101
|
Pokharel YR, Saarela J, Szwajda A, Rupp C, Rokka A, Lal Kumar Karna S, Teittinen K, Corthals G, Kallioniemi O, Wennerberg K, Aittokallio T, Westermarck J. Relevance Rank Platform (RRP) for Functional Filtering of High Content Protein-Protein Interaction Data. Mol Cell Proteomics 2015; 14:3274-83. [PMID: 26499835 DOI: 10.1074/mcp.m115.050773] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Indexed: 11/06/2022] Open
Abstract
High content protein interaction screens have revolutionized our understanding of protein complex assembly. However, one of the major challenges in translation of high content protein interaction data is identification of those interactions that are functionally relevant for a particular biological question. To address this challenge, we developed a relevance ranking platform (RRP), which consist of modular functional and bioinformatic filters to provide relevance rank among the interactome proteins. We demonstrate the versatility of RRP to enable a systematic prioritization of the most relevant interaction partners from high content data, highlighted by the analysis of cancer relevant protein interactions for oncoproteins Pin1 and PME-1. We validated the importance of selected interactions by demonstration of PTOV1 and CSKN2B as novel regulators of Pin1 target c-Jun phosphorylation and reveal previously unknown interacting proteins that may mediate PME-1 effects via PP2A-inhibition. The RRP framework is modular and can be modified to answer versatile research problems depending on the nature of the biological question under study. Based on comparison of RRP to other existing filtering tools, the presented data indicate that RRP offers added value especially for the analysis of interacting proteins for which there is no sufficient prior knowledge available. Finally, we encourage the use of RRP in combination with either SAINT or CRAPome computational tools for selecting the candidate interactors that fulfill the both important requirements, functional relevance, and high confidence interaction detection.
Collapse
Affiliation(s)
- Yuba Raj Pokharel
- From the ‡Institute for Molecular Medicine Finland FIMM, University of Helsinki, PO Box 20, FIN-00014 Helsinki, Finland; §Centre for Biotechnology, ‖Faculty of Life Science and Biotechnology, South Asian University, New Delhi 110021, India
| | - Jani Saarela
- From the ‡Institute for Molecular Medicine Finland FIMM, University of Helsinki, PO Box 20, FIN-00014 Helsinki, Finland
| | - Agnieszka Szwajda
- From the ‡Institute for Molecular Medicine Finland FIMM, University of Helsinki, PO Box 20, FIN-00014 Helsinki, Finland
| | | | | | | | - Kaisa Teittinen
- **Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Tampere University Hospital, FIN-33014, Tampere, Finland
| | | | - Olli Kallioniemi
- From the ‡Institute for Molecular Medicine Finland FIMM, University of Helsinki, PO Box 20, FIN-00014 Helsinki, Finland
| | - Krister Wennerberg
- From the ‡Institute for Molecular Medicine Finland FIMM, University of Helsinki, PO Box 20, FIN-00014 Helsinki, Finland
| | - Tero Aittokallio
- From the ‡Institute for Molecular Medicine Finland FIMM, University of Helsinki, PO Box 20, FIN-00014 Helsinki, Finland
| | - Jukka Westermarck
- From the ‡Institute for Molecular Medicine Finland FIMM, University of Helsinki, PO Box 20, FIN-00014 Helsinki, Finland; §Centre for Biotechnology, ¶Department of Pathology,University of Turku and Åbo Akademi, Turku, Finland, PO Box 123, FIN-20521 Turku, Finland.;
| |
Collapse
|
102
|
Cheng YS, Seibert O, Klöting N, Dietrich A, Straßburger K, Fernández-Veledo S, Vendrell JJ, Zorzano A, Blüher M, Herzig S, Berriel Diaz M, Teleman AA. PPP2R5C Couples Hepatic Glucose and Lipid Homeostasis. PLoS Genet 2015; 11:e1005561. [PMID: 26440364 PMCID: PMC4595073 DOI: 10.1371/journal.pgen.1005561] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 09/10/2015] [Indexed: 01/12/2023] Open
Abstract
In mammals, the liver plays a central role in maintaining carbohydrate and lipid homeostasis by acting both as a major source and a major sink of glucose and lipids. In particular, when dietary carbohydrates are in excess, the liver converts them to lipids via de novo lipogenesis. The molecular checkpoints regulating the balance between carbohydrate and lipid homeostasis, however, are not fully understood. Here we identify PPP2R5C, a regulatory subunit of PP2A, as a novel modulator of liver metabolism in postprandial physiology. Inactivation of PPP2R5C in isolated hepatocytes leads to increased glucose uptake and increased de novo lipogenesis. These phenotypes are reiterated in vivo, where hepatocyte specific PPP2R5C knockdown yields mice with improved systemic glucose tolerance and insulin sensitivity, but elevated circulating triglyceride levels. We show that modulation of PPP2R5C levels leads to alterations in AMPK and SREBP-1 activity. We find that hepatic levels of PPP2R5C are elevated in human diabetic patients, and correlate with obesity and insulin resistance in these subjects. In sum, our data suggest that hepatic PPP2R5C represents an important factor in the functional wiring of energy metabolism and the maintenance of a metabolically healthy state. After a meal, dietary glucose travels through the hepatic portal vein to the liver. A substantial part of this glucose is taken up by liver, which converts it to glycogen which is stored, and lipids which are in part stored and in part secreted as VLDL particles. The rest of the organs receive whatever glucose the liver leaves in circulation, plus the secreted lipids. Hence the liver plays a crucial role in determining the balance of sugar versus lipids in the body after a meal. This balance is very important, because too much glucose in circulation leads to diabetic complications whereas too much VLDL increases risk of atherosclerosis. Little is known about how the liver strikes this balance. We identify here a phosphatase—the PP2A holoenzyme containing the PPP2R5C regulatory subunit—as a regulator of this process. We find that knockdown of PPP2R5C in mouse liver specifically causes it to uptake elevated levels of glucose, and secrete elevated levels of VLDL into circulation. This leads to a phenotype of improved glucose tolerance and insulin sensitivity. The prediction from these functional studies in mice is that elevated levels of PPP2R5C expression should lead to insulin resistance. Indeed, we find that PPP2R5C expression levels are elevated in diabetic patients, or healthy controls with visceral obesity, raising the possibility that dysregulation of PPP2R5C expression in humans may contribute towards metabolic dysfunction.
Collapse
Affiliation(s)
| | - Oksana Seibert
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nora Klöting
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Arne Dietrich
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | | | - Sonia Fernández-Veledo
- Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili. Universitat, Rovira i Virgili, CIBERDEM, Tarragona, Spain
| | - Joan J. Vendrell
- Hospital Universitari de Tarragona Joan XXIII. Institut d´Investigació Sanitària Pere Virgili Universitat Rovira i Virgili, CIBERDEM, Tarragona, Spain
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), Departament de Bioquímica i Biologia Molecular, Universitat de Barcelona, and CIBERDEM, Barcelona, , Spain
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Stephan Herzig
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany, and Joint Heidelberg-IDC Translational Diabetes Program, University Hospital Heidelberg, Heidelberg, Germany
- Chair Molecular Metabolic Control, Medical Faculty, Technical University Munich, Germany
| | - Mauricio Berriel Diaz
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany, and Joint Heidelberg-IDC Translational Diabetes Program, University Hospital Heidelberg, Heidelberg, Germany
- * E-mail: (MBD); (AAT)
| | - Aurelio A. Teleman
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- * E-mail: (MBD); (AAT)
| |
Collapse
|
103
|
CIP2A is associated with multidrug resistance in cervical adenocarcinoma by a P-glycoprotein pathway. Tumour Biol 2015; 37:2673-82. [PMID: 26404133 DOI: 10.1007/s13277-015-4032-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 09/01/2015] [Indexed: 12/13/2022] Open
Abstract
Cancerous inhibitor of protein phosphatase 2A (CIP2A) is a recently identified oncoprotein. Here, we investigated its role in the formation of multidrug resistance (MDR) of cervical adenocarcinoma in vitro and in vivo. MTT assay showed that knockdown of CIP2A expression increased the drug sensitivity of HeLa and Dox-resistant HeLa cells (HeLa-Dox) to doxorubicin, cisplatin, and paclitaxel significantly, while overexpression of CIP2A decreased the sensitivity of HeLa cells to chemo-drugs dramatically. When treated with different chemotherapeutics, CIP2A and P-glycoprotein (P-gp) protein levels were increased in HeLa cells simultaneously. In accordance with it, knockdown or overexpression of CIP2A expression inhibited or increased the P-gp expression in the transcription level separately. The effects of CIP2A on P-gp expression was achieved partly through its regulation on the transcription factor E2F1. Moreover, the interference of CIP2A could decrease the P-gp protein activity elucidated by Rhodamine 123 (Rh123) efflux assay in HeLa and HeLa/Dox cells. In the in vivo level, confocal microscopy data demonstrated the strong co-localization of CIP2A and P-gp protein in HeLa cells, and CIP2A protein expression was significantly associated with that of P-gp in cervical adenocarcinoma tissues. Thus, CIP2A is involved in regulating multidrug resistance of cervical adenocarcinoma upon chemotherapy by enhancing P-gp expression through E2F1. CIP2A may be an attractive target in anticancer strategies to improve the effect of chemotherapy in cervical adenocarcinoma.
Collapse
|
104
|
Routila J, Mäkelä JA, Luukkaa H, Leivo I, Irjala H, Westermarck J, Mäkitie A, Ventelä S. Potential role for inhibition of protein phosphatase 2A tumor suppressor in salivary gland malignancies. Genes Chromosomes Cancer 2015; 55:69-81. [DOI: 10.1002/gcc.22312] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 09/06/2015] [Accepted: 09/07/2015] [Indexed: 12/14/2022] Open
Affiliation(s)
- Johannes Routila
- The Centre for Biotechnology; University of Turku and Åbo Akademi University; Tykistökatu BioCity Turku FI-20521 Finland
| | - Juho-Antti Mäkelä
- Department of Physiology; University of Turku; Kiinamyllynkatu 10 Turku FI-20520 Finland
| | - Heikki Luukkaa
- Department of Otorhinolaryngology-Head and Neck Surgery; Turku University Hospital; Kiinamyllynkatu 4-8 Turku FI-20521 Finland
| | - Ilmo Leivo
- Department of Pathology; University of Turku; Kiinamyllynkatu 10 Turku FI-20520 Finland
| | - Heikki Irjala
- Department of Otorhinolaryngology-Head and Neck Surgery; Turku University Hospital; Kiinamyllynkatu 4-8 Turku FI-20521 Finland
| | - Jukka Westermarck
- The Centre for Biotechnology; University of Turku and Åbo Akademi University; Tykistökatu BioCity Turku FI-20521 Finland
- Department of Pathology; University of Turku; Kiinamyllynkatu 10 Turku FI-20520 Finland
| | - Antti Mäkitie
- Department of Otorhinolaryngology-Head and Neck Surgery; Helsinki University Central Hospital and University of Helsinki; HUCH Helsinki FI-00029 Finland
| | - Sami Ventelä
- The Centre for Biotechnology; University of Turku and Åbo Akademi University; Tykistökatu BioCity Turku FI-20521 Finland
- Department of Otorhinolaryngology-Head and Neck Surgery; Turku University Hospital; Kiinamyllynkatu 4-8 Turku FI-20521 Finland
| |
Collapse
|
105
|
Kaur A, Elzagheid A, Birkman EM, Avoranta T, Kytölä V, Korkeila E, Syrjänen K, Westermarck J, Sundström J. Protein phosphatase methylesterase-1 (PME-1) expression predicts a favorable clinical outcome in colorectal cancer. Cancer Med 2015; 4:1798-808. [PMID: 26377365 PMCID: PMC5123709 DOI: 10.1002/cam4.541] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 08/19/2015] [Accepted: 08/20/2015] [Indexed: 12/25/2022] Open
Abstract
Colorectal cancer (CRC) accounts for high mortality. So far, there is lack of markers capable of predicting which patients are at risk of aggressive course of the disease. Protein phosphatase-2A (PP2A) inhibitor proteins have recently gained interest as markers of more aggressive disease in certain cancers. Here, we report the role of PP2A inhibitor PME-1 in CRC. PME-1 expression was assessed from a rectal cancer patient cohort by immunohistochemistry, and correlations were performed for various clinicopathological variables and patient survival. Rectal cancer patients with higher cytoplasmic PME-1 protein expression (above median) had less recurrences (P = 0.003, n = 195) and better disease-free survival (DFS) than the patients with low cytoplasmic PME-1 protein expression (below median). Analysis of PPME-1 mRNA expression from TCGA dataset of colon and rectal adenocarcinoma (COADREAD) patient cohort confirmed high PPME1 expression as an independent protective factor predicting favorable overall survival (OS) (P = 0.005, n = 396) compared to patients with low PPME1 expression. CRC cell lines were used to study the effect of PME-1 knockdown by siRNA on cell survival. Contrary to other cancer types, PME-1 inhibition in CRC cell lines did not reduce the viability of cells or the expression of active phosphorylated AKT and ERK proteins. In conclusion, PME-1 expression predicts for a favorable outcome of CRC patients. The unexpected role of PME-1 in CRC in contrast with the oncogenic role of PP2A inhibitor proteins in other malignancies warrants further studies of cancer-specific function for each of these proteins.
Collapse
Affiliation(s)
- Amanpreet Kaur
- Department of Pathology, University of Turku, Turku, 20520, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, 20520, Finland.,TuBS and TuDMM Doctoral Programmes, Turku, 20520, Finland
| | - Adam Elzagheid
- Department of Pathology, University of Turku, Turku, 20520, Finland.,Department of Pathology, Faculty of Medicine, Benghazi University, PO Box 1308, Benghazi, Libya.,Biotechnology Research Center, Tripoli, Libya
| | | | - Tuulia Avoranta
- Department of Pathology, University of Turku, Turku, 20520, Finland.,Department of Oncology and Radiotherapy, University of Turku and Turku University Hospital, Turku, 20521, Finland.,Department of Social Services and Healthcare, City of Helsinki, Helsinki, 00099, Finland
| | - Ville Kytölä
- BioMediTech, University of Tampere, Tampere, 33520, Finland
| | - Eija Korkeila
- Department of Oncology and Radiotherapy, University of Turku and Turku University Hospital, Turku, 20521, Finland
| | - Kari Syrjänen
- Department of Clinical Research, Biohit Oyj, Helsinki, 00880, Finland.,Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, 14784-400, Brazil
| | - Jukka Westermarck
- Department of Pathology, University of Turku, Turku, 20520, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, 20520, Finland
| | - Jari Sundström
- Department of Pathology, University of Turku, Turku, 20520, Finland
| |
Collapse
|
106
|
Ventelä S, Sittig E, Mannermaa L, Mäkelä JA, Kulmala J, Löyttyniemi E, Strauss L, Cárpen O, Toppari J, Grénman R, Westermarck J. CIP2A is an Oct4 target gene involved in head and neck squamous cell cancer oncogenicity and radioresistance. Oncotarget 2015; 6:144-58. [PMID: 25474139 PMCID: PMC4381584 DOI: 10.18632/oncotarget.2670] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 11/02/2014] [Indexed: 12/31/2022] Open
Abstract
Radiotherapy is a mainstay for treatment of many human cancer types, including head and neck squamous cell carcinoma (HNSCC). Thereby, it is clinically very relevant to understand the mechanisms determining radioresistance. Here, we identify CIP2A as an Oct4 target gene and provide evidence that they co-operate in radioresistance. Oct4 positively regulates CIP2A expression both in testicular cancer cell lines as well as in embryonic stem cells. To expand the relevance of these findings we show that Oct4 and CIP2A are co-expressed in CD24 positive side-population of patient-derived HNSCC cell lines. Most importantly, all Oct4 positive HNSCC patient samples were CIP2A positive and this double positivity was linked to poor differentiation level, and predicted for decreased patient survival among radiotherapy treated HNSCC patients. Oct4 and CIP2A expression was also linked with increased aggressiveness and radioresistancy in HNSCC cell lines. Together we demonstrate that CIP2A is a novel Oct4 target gene in stem cells and in human cancer cell lines. Clinically these results suggest that diagnostic evaluation of HNSCC tumors for Oct4 or Oct4/CIP2A positivity might help to predict HNSCC tumor radioresistancy. These results also identify both Oct4 and CIP2A as potential targets for radiosensitation.
Collapse
Affiliation(s)
- Sami Ventelä
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi, Turku, Finland. Department of Physiology, University of Turku, Finland. Department of Otorhinolaryngology - Head and Neck Surgery, Turku University Hospital, Turku, Finland
| | - Eleonora Sittig
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi, Turku, Finland
| | - Leni Mannermaa
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi, Turku, Finland
| | | | - Jarmo Kulmala
- Department of Oncology and Radiotherapy, Turku University Hospital, Turku, Finland
| | | | - Leena Strauss
- Institute of Biomedicine and Turku Center for Disease Modeling, University of Turku, Finland
| | - Olli Cárpen
- Department of Pathology, University of Turku, Finland
| | - Jorma Toppari
- Department of Physiology, University of Turku, Finland. Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Reidar Grénman
- Department of Otorhinolaryngology - Head and Neck Surgery, Turku University Hospital, Turku, Finland
| | - Jukka Westermarck
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi, Turku, Finland. Department of Pathology, University of Turku, Finland
| |
Collapse
|
107
|
Myant K, Qiao X, Halonen T, Come C, Laine A, Janghorban M, Partanen JI, Cassidy J, Ogg EL, Cammareri P, Laiterä T, Okkeri J, Klefström J, Sears RC, Sansom OJ, Westermarck J. Serine 62-Phosphorylated MYC Associates with Nuclear Lamins and Its Regulation by CIP2A Is Essential for Regenerative Proliferation. Cell Rep 2015; 12:1019-31. [PMID: 26235622 PMCID: PMC4535171 DOI: 10.1016/j.celrep.2015.07.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 04/24/2015] [Accepted: 07/01/2015] [Indexed: 02/07/2023] Open
Abstract
An understanding of the mechanisms determining MYC's transcriptional and proliferation-promoting activities in vivo could facilitate approaches for MYC targeting. However, post-translational mechanisms that control MYC function in vivo are poorly understood. Here, we demonstrate that MYC phosphorylation at serine 62 enhances MYC accumulation on Lamin A/C-associated nuclear structures and that the protein phosphatase 2A (PP2A) inhibitor protein CIP2A is required for this process. CIP2A is also critical for serum-induced MYC phosphorylation and for MYC-elicited proliferation induction in vitro. Complementary transgenic approaches and an intestinal regeneration model further demonstrated the in vivo importance of CIP2A and serine 62 phosphorylation for MYC activity upon DNA damage. However, targeting of CIP2A did not influence the normal function of intestinal crypt cells. These data underline the importance of nuclear organization in the regulation of MYC phosphorylation, leading to an in vivo demonstration of a strategy for inhibiting MYC activity without detrimental physiological effects.
Collapse
Affiliation(s)
- Kevin Myant
- The Beatson Institute for Cancer Research, Glasgow G61 1BD, UK
| | - Xi Qiao
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Department of Pathology, University of Turku, 20520 Turku, Finland
| | - Tuuli Halonen
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Christophe Come
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Anni Laine
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Mahnaz Janghorban
- Department of Molecular and Medical Genetics and Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Johanna I Partanen
- Research Programs Unit, Translational Cancer Biology and Institute of Biomedicine, University of Helsinki, 00014 Helsinki, Finland
| | - John Cassidy
- The Beatson Institute for Cancer Research, Glasgow G61 1BD, UK
| | - Erinn-Lee Ogg
- The Beatson Institute for Cancer Research, Glasgow G61 1BD, UK
| | | | - Tiina Laiterä
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Juha Okkeri
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Juha Klefström
- Research Programs Unit, Translational Cancer Biology and Institute of Biomedicine, University of Helsinki, 00014 Helsinki, Finland
| | - Rosalie C Sears
- Department of Molecular and Medical Genetics and Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Owen J Sansom
- The Beatson Institute for Cancer Research, Glasgow G61 1BD, UK.
| | - Jukka Westermarck
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Department of Pathology, University of Turku, 20520 Turku, Finland.
| |
Collapse
|
108
|
Ohama T. [Targeting PP2A inhibitors as a novel anti-cancer strategy
]. Nihon Yakurigaku Zasshi 2015; 145:293-8. [PMID: 26063151 DOI: 10.1254/fpj.145.293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
109
|
Golebiewska A, Fritah S, Girotti MR. OECI-EACR precision medicine for cancer: Conference report 1-4 March 2015, Luxembourg. Ecancermedicalscience 2015; 9:519. [PMID: 25932043 PMCID: PMC4404038 DOI: 10.3332/ecancer.2015.519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Indexed: 11/25/2022] Open
Abstract
The ‘Precision Medicine for Cancer’ was the first meeting of a new series of conferences organised biannually by the European Association for Cancer Research (EACR) and the Organisation for European Cancer Institutes (OECI). The main objective of the meeting was to focus on novel topics in precision medicine by allowing strong interactions between participants and to access the speakers easily. As the first implementations of personalised medicine are appreciated in the clinic, the aim of the meeting was to further educate both researchers and clinicians and learn more from the novel approaches in the field. Similarly, the interaction between two organisations—the research-oriented EACR and the clinic-oriented OECI—was of a great value for the meeting. This OECI-EACR 2015 report will highlight the major findings of this outstanding meeting.
Collapse
Affiliation(s)
- Anna Golebiewska
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (L.I.H), Luxembourg, L-1526 Luxembourg
| | - Sabrina Fritah
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (L.I.H), Luxembourg, L-1526 Luxembourg
| | - Maria Romina Girotti
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, M20 4BX, UK
| |
Collapse
|
110
|
Khanna A. DNA Damage in Cancer Therapeutics: A Boon or a Curse? Cancer Res 2015; 75:2133-8. [DOI: 10.1158/0008-5472.can-14-3247] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 02/17/2015] [Indexed: 11/16/2022]
|
111
|
Strategies to make protein serine/threonine (PP1, calcineurin) and tyrosine phosphatases (PTP1B) druggable: achieving specificity by targeting substrate and regulatory protein interaction sites. Bioorg Med Chem 2015; 23:2781-5. [PMID: 25771485 DOI: 10.1016/j.bmc.2015.02.040] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/09/2015] [Accepted: 02/19/2015] [Indexed: 12/18/2022]
Abstract
The established dogma is that protein serine/threonine (PSPs) and tyrosine (PTPs) phosphatases are unattainable drug targets. This is because natural product inhibitors of PSP active sites are lethal, while the active sites of PTPs are exceptionally conserved and charged, making it nearly impossible to develop PTP inhibitors that are selective. However, due to a series of recent structural and functional studies, this view of phosphatases is about to undergo a radical change. Rather than target active sites, these studies have demonstrated that targeting PSP/PTP protein (substrate/regulatory) interaction sites, which are distal from the active sites, are highly viable and suitable drugs targets. This is especially true for calcineurin (CN), in which the blockbuster immunosuppressant drugs FK506 and cyclosporin A were recently demonstrated to bind and block one of the key CN substrate interaction sites, the LxVP site. Additional studies show that this approach-targeting substrate and/or regulatory protein interaction sites-also holds incredible promise for protein phosphatase 1 (PP1)-related diseases. Finally, domains outside PTP catalytic domains have also recently been demonstrated to directly alter PTP activity. Collectively, these novel insights offer new, transformative perspectives for the therapeutic targeting of PSPs by interfering with the binding of PIPs or substrates and PTPs by targeting allosteric sites outside their catalytic domains.
Collapse
|
112
|
Flørenes VA, Emilsen E, Dong HP, Førsund M, Holm R, Slipicevic A. Cellular localization of CIP2A determines its prognostic impact in superficial spreading and nodular melanoma. Cancer Med 2015; 4:903-13. [PMID: 25663244 PMCID: PMC4472213 DOI: 10.1002/cam4.425] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 12/15/2014] [Accepted: 12/23/2014] [Indexed: 01/31/2023] Open
Abstract
Cancerous inhibitor of protein phosphatase 2A (CIP2A) is an important oncogene contributing to cancer progression partially by regulating cMYC and AKT. We examined CIP2A expression in cutaneous melanomas, its association with clinicopathological parameters and mapped molecular mechanisms regulated by CIP2A in vitro. CIP2A expression was analyzed by immunohistochemistry in 17 nevi, 132 primary melanomas and 49 metastases. Effects of siRNA-mediated down-regulation on proliferation, apoptosis and signaling pathways were assessed in melanoma cell lines. In superficial spreading melanomas (SSM), high nuclear CIP2A expression was associated with poor overall survival (OS) (P = 0.0018). Surprisingly, high cytoplasmic expression was related to improved relapse-free (P = 0.031) and OS (P = 0.014) in nodular melanomas (NM). In vitro experiments revealed that CIP2A can regulate proliferation and/or apoptosis partially through the PI3K/AKT pathway but also independently. In summary, CIP2A could represent a potential therapeutic target in SSM. However, in NM cytoplasmic CIP2A is associated with improved prognosis indicating that CIP2A has distinct, complex functions dependent on the molecular context and histological subtype. As seen in other cancer types, CIP2A can influence cMYC and AKT, but our data also suggest that in melanoma it has additional targets which need to be identified.
Collapse
Affiliation(s)
- Vivi Ann Flørenes
- Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, N-0310, Oslo, Norway
| | - Elisabeth Emilsen
- Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, N-0310, Oslo, Norway
| | - Hiep Phuc Dong
- Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, N-0310, Oslo, Norway
| | - Mette Førsund
- Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, N-0310, Oslo, Norway
| | - Ruth Holm
- Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, N-0310, Oslo, Norway
| | - Ana Slipicevic
- Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, N-0310, Oslo, Norway
| |
Collapse
|
113
|
Barragán E, Chillón MC, Castelló-Cros R, Marcotegui N, Prieto MI, Hoyos M, Pippa R, Llop M, Etxabe A, Cervera J, Rodríguez G, Buño I, Rifón J, Sierra J, González M, Calasanz MJ, Sanz MA, Odero MD. CIP2A high expression is a poor prognostic factor in normal karyotype acute myeloid leukemia. Haematologica 2015; 100:e183-5. [PMID: 25637054 DOI: 10.3324/haematol.2014.118117] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Eva Barragán
- Clinical Laboratory and Department of Hematology, University Hospital La Fe, Valencia, Spain
| | - María Carmen Chillón
- Department of Hematology, University Hospital of Salamanca, Salamanca Institute for Biomedical Research (IBSAL), Spain
| | | | - Nerea Marcotegui
- Program of Hematology-Oncology, CIMA, University of Navarra, Pamplona, Spain
| | - María Isabel Prieto
- Department of Hematology, University Hospital of Salamanca, Salamanca Institute for Biomedical Research (IBSAL), Spain
| | - Montserrat Hoyos
- Department of Hematology, Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | - Raffaella Pippa
- Program of Hematology-Oncology, CIMA, University of Navarra, Pamplona, Spain
| | - Marta Llop
- Clinical Laboratory and Department of Hematology, University Hospital La Fe, Valencia, Spain
| | - Amaia Etxabe
- Program of Hematology-Oncology, CIMA, University of Navarra, Pamplona, Spain
| | - José Cervera
- Clinical Laboratory and Department of Hematology, University Hospital La Fe, Valencia, Spain
| | - Gabriela Rodríguez
- Department of Hematology, Gregorio Marañón Institute for Health Research, Madrid, Spain
| | - Ismael Buño
- Department of Hematology, Gregorio Marañón Institute for Health Research, Madrid, Spain
| | - José Rifón
- Department of Hematology, Clinical University of Navarra, Pamplona, Spain
| | - Jorge Sierra
- Department of Hematology, Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | - Marcos González
- Department of Hematology, University Hospital of Salamanca, Salamanca Institute for Biomedical Research (IBSAL), Spain
| | - María J Calasanz
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain
| | - Miguel A Sanz
- Clinical Laboratory and Department of Hematology, University Hospital La Fe, Valencia, Spain
| | - María D Odero
- Program of Hematology-Oncology, CIMA, University of Navarra, Pamplona, Spain Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain
| |
Collapse
|
114
|
Khanna A, Pimanda JE. Clinical significance of cancerous inhibitor of protein phosphatase 2A in human cancers. Int J Cancer 2015; 138:525-32. [DOI: 10.1002/ijc.29431] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Revised: 12/28/2014] [Accepted: 12/29/2014] [Indexed: 01/03/2023]
Affiliation(s)
- Anchit Khanna
- Lowy Cancer Research Centre and Prince of Wales Clinical School, University of New South Wales (UNSW) Medicine Department; Sydney New South Wales 2052 Australia
| | - John E. Pimanda
- Lowy Cancer Research Centre and Prince of Wales Clinical School, University of New South Wales (UNSW) Medicine Department; Sydney New South Wales 2052 Australia
- Department of Haematology; the Prince of Wales Hospital; Randwick New South Wales Australia
| |
Collapse
|
115
|
Lindskog C, Edlund K, Mattsson JSM, Micke P. Immunohistochemistry-based prognostic biomarkers in NSCLC: novel findings on the road to clinical use? Expert Rev Mol Diagn 2015; 15:471-90. [DOI: 10.1586/14737159.2015.1002772] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
116
|
Haesen D, Sents W, Lemaire K, Hoorne Y, Janssens V. The Basic Biology of PP2A in Hematologic Cells and Malignancies. Front Oncol 2014; 4:347. [PMID: 25566494 PMCID: PMC4263090 DOI: 10.3389/fonc.2014.00347] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 11/20/2014] [Indexed: 12/30/2022] Open
Abstract
Reversible protein phosphorylation plays a crucial role in regulating cell signaling. In normal cells, phosphoregulation is tightly controlled by a network of protein kinases counterbalanced by several protein phosphatases. Deregulation of this delicate balance is widely recognized as a central mechanism by which cells escape external and internal self-limiting signals, eventually resulting in malignant transformation. A large fraction of hematologic malignancies is characterized by constitutive or unrestrained activation of oncogenic kinases. This is in part achieved by activating mutations, chromosomal rearrangements, or constitutive activation of upstream kinase regulators, in part by inactivation of their anti-oncogenic phosphatase counterparts. Protein phosphatase 2A (PP2A) represents a large family of cellular serine/threonine phosphatases with suspected tumor suppressive functions. In this review, we highlight our current knowledge about the complex structure and biology of these phosphatases in hematologic cells, thereby providing the rationale behind their diverse signaling functions. Eventually, this basic knowledge is a key to truly understand the tumor suppressive role of PP2A in leukemogenesis and to allow further rational development of therapeutic strategies targeting PP2A.
Collapse
Affiliation(s)
- Dorien Haesen
- Laboratory of Protein Phosphorylation and Proteomics, Department Cellular and Molecular Medicine, University of Leuven , Leuven , Belgium
| | - Ward Sents
- Laboratory of Protein Phosphorylation and Proteomics, Department Cellular and Molecular Medicine, University of Leuven , Leuven , Belgium
| | - Katleen Lemaire
- Gene Expression Unit, Department Cellular and Molecular Medicine, University of Leuven , Leuven , Belgium
| | - Yana Hoorne
- Laboratory of Protein Phosphorylation and Proteomics, Department Cellular and Molecular Medicine, University of Leuven , Leuven , Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation and Proteomics, Department Cellular and Molecular Medicine, University of Leuven , Leuven , Belgium
| |
Collapse
|
117
|
Li T, Wang G. Computer-aided targeting of the PI3K/Akt/mTOR pathway: toxicity reduction and therapeutic opportunities. Int J Mol Sci 2014; 15:18856-91. [PMID: 25334061 PMCID: PMC4227251 DOI: 10.3390/ijms151018856] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 09/21/2014] [Accepted: 10/08/2014] [Indexed: 12/14/2022] Open
Abstract
The PI3K/Akt/mTOR pathway plays an essential role in a wide range of biological functions, including metabolism, macromolecular synthesis, cell growth, proliferation and survival. Its versatility, however, makes it a conspicuous target of many pathogens; and the consequential deregulations of this pathway often lead to complications, such as tumorigenesis, type 2 diabetes and cardiovascular diseases. Molecular targeted therapy, aimed at modulating the deregulated pathway, holds great promise for controlling these diseases, though side effects may be inevitable, given the ubiquity of the pathway in cell functions. Here, we review a variety of factors found to modulate the PI3K/Akt/mTOR pathway, including gene mutations, certain metabolites, inflammatory factors, chemical toxicants, drugs found to rectify the pathway, as well as viruses that hijack the pathway for their own synthetic purposes. Furthermore, this evidence of PI3K/Akt/mTOR pathway alteration and related pathogenesis has inspired the exploration of computer-aided targeting of this pathway to optimize therapeutic strategies. Herein, we discuss several possible options, using computer-aided targeting, to reduce the toxicity of molecularly-targeted therapy, including mathematical modeling, to reveal system-level control mechanisms and to confer a low-dosage combination therapy, the potential of PP2A as a therapeutic target, the formulation of parameters to identify patients who would most benefit from specific targeted therapies and molecular dynamics simulations and docking studies to discover drugs that are isoform specific or mutation selective so as to avoid undesired broad inhibitions. We hope this review will stimulate novel ideas for pharmaceutical discovery and deepen our understanding of curability and toxicity by targeting the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Tan Li
- Department of Biology, South University of Science and Technology of China, 1088 Xueyuan Rd., Shenzhen 518055, China.
| | - Guanyu Wang
- Department of Biology, South University of Science and Technology of China, 1088 Xueyuan Rd., Shenzhen 518055, China.
| |
Collapse
|
118
|
Lei N, Peng B, Zhang JY. CIP2A regulates cell proliferation via the AKT signaling pathway in human lung cancer. Oncol Rep 2014; 32:1689-94. [PMID: 25109354 PMCID: PMC4148383 DOI: 10.3892/or.2014.3375] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 05/28/2014] [Indexed: 12/17/2022] Open
Abstract
Cancerous inhibitor of PP2A (CIP2A) is an intracellular endogenous protein phosphatase 2A (PP2A) inhibitor with oncogenic activities. Initially identified as a tumor-associated antigen (TAA) in gastric and liver cancer patients, CIP2A was overexpressed in a variety of cancer types. The overexpression of CIP2A in cancer cells is associated with increased cell proliferation. However, the mechanism of CIP2A in cancer cell proliferation remains poorly understood. In the present study, we reported that CIP2A can regulate AKT phosphorylation at S473 under growth factor stimulation and our results also showed that CIP2A may promote cell proliferation through the AKT signaling pathway. Notably, depletion of CIP2A did not induce a global change of AKT phosphatase activity, which indicated that CIP2A may recognize specific AKT targets and play certain roles in the signaling pathway. In addition, we detected that CIP2A expression was associated with mTOR phosphorylation. Our further analysis corroborated the relationship between CIP2A and AKT-mTOR signaling pathway. Therefore, our study addressed a novel role of CIP2A in mediating cancer progression through interacting with the AKT-mTOR signaling pathway.
Collapse
Affiliation(s)
- Ningjing Lei
- Border Biomedical Research Center and Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Bo Peng
- Border Biomedical Research Center and Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Jian-Ying Zhang
- Border Biomedical Research Center and Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
| |
Collapse
|
119
|
Erlotinib derivative inhibits hepatocellular carcinoma by targeting CIP2A to reactivate protein phosphatase 2A. Cell Death Dis 2014; 5:e1359. [PMID: 25077545 PMCID: PMC4123111 DOI: 10.1038/cddis.2014.325] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 06/27/2014] [Accepted: 07/01/2014] [Indexed: 12/23/2022]
Abstract
Protein phosphatase 2A (PP2A) is a tumor suppressor, which is functionally defective in various cancers. Previously, we found that PP2A activity determined the anticancer effect of bortezomib and erlotinib in hepatocellular carcinoma (HCC) cells. Here, we tested a novel erlotinib derivative, TD52, in four HCC cell lines, PLC5, Huh-7, Hep3B and Sk-Hep1. Using MTT and flow cytometry, we showed that TD52 had more potent apoptotic effects than erlotinib in HCC cells. TD52-induced apoptosis was associated with dose- and time- dependent reactivation of PP2A and downregulation of cancerous inhibitor of protein phosphatase 2A (CIP2A) and p-Akt. Inhibition of PP2A or ectopic expression of CIP2A or Akt in PLC5 cells abolished the effects of TD52. Furthermore, we demonstrated that TD52 affected the binding of Elk-1 to the proximal promoter of the CIP2A gene, thus downregulating transcription of CIP2A. Importantly, TD52-induced tumor inhibition was associated with reactivation of PP2A and downregulation of CIP2A and p-Akt in vivo. In conclusion, we found that enhancement of PP2A activity by inhibition of CIP2A determines the apoptotic effect induced by TD52. Our findings disclose the therapeutic mechanism of this novel targeted agent, and suggest the therapeutic potential and feasibility of developing PP2A enhancers as a novel anticancer strategy.
Collapse
|
120
|
De P, Carlson J, Leyland-Jones B, Dey N. Oncogenic nexus of cancerous inhibitor of protein phosphatase 2A (CIP2A): an oncoprotein with many hands. Oncotarget 2014; 5:4581-602. [PMID: 25015035 PMCID: PMC4148086 DOI: 10.18632/oncotarget.2127] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 06/20/2014] [Indexed: 12/23/2022] Open
Abstract
Oncoprotein CIP2A a Cancerous Inhibitor of PP2A forms an "oncogenic nexus" by virtue of its control on PP2A and MYC stabilization in cancer cells. The expression and prognostic function of CIP2A in different solid tumors including colorectal carcinoma, head and neck cancers, gastric cancers, lung carcinoma, cholangiocarcinoma, esophageal cancers, pancreatic carcinoma, brain cancers, breast carcinoma, bladder cancers, ovarian carcinoma, renal cell carcinomas, tongue cancers, cervical carcinoma, prostate cancers, and oral carcinoma as well as a number of hematological malignancies are just beginning to emerge. Herein, we reviewed the recent progress in our understanding of (1) how an "oncogenic nexus" of CIP2A participates in the tumorigenic transformation of cells and (2) how we can prospect/view the clinical relevance of CIP2A in the context of cancer therapy. The review will try to understand the role of CIP2A (a) as a biomarker in cancers and evaluate the prognostic value of CIP2A in different cancers (b) as a therapeutic target in cancers and (c) in drug response and developing chemo-resistance in cancers.
Collapse
Affiliation(s)
- Pradip De
- Department of Molecular & Experimental Medicine, Avera Research Institute, Sioux Falls, SD
- Department of Internal Medicine, SSOM, University of South Dakota, Sioux Falls, SD
| | - Jennifer Carlson
- Department of Molecular & Experimental Medicine, Avera Research Institute, Sioux Falls, SD
| | - Brian Leyland-Jones
- Department of Molecular & Experimental Medicine, Avera Research Institute, Sioux Falls, SD
- Department of Internal Medicine, SSOM, University of South Dakota, Sioux Falls, SD
| | - Nandini Dey
- Department of Molecular & Experimental Medicine, Avera Research Institute, Sioux Falls, SD
- Department of Internal Medicine, SSOM, University of South Dakota, Sioux Falls, SD
| |
Collapse
|
121
|
Liu N, He QM, Chen JW, Li YQ, Xu YF, Ren XY, Sun Y, Mai HQ, Shao JY, Jia WH, Kang TB, Zeng MS, Ma J. Overexpression of CIP2A is an independent prognostic indicator in nasopharyngeal carcinoma and its depletion suppresses cell proliferation and tumor growth. Mol Cancer 2014; 13:111. [PMID: 24884612 PMCID: PMC4046003 DOI: 10.1186/1476-4598-13-111] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 05/15/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cancerous inhibitor of protein phosphatase 2A (CIP2A) is an oncoprotein that acts as a prognostic marker for several human malignancies. In this study, we investigated the clinical significance of CIP2A and its function in nasopharyngeal carcinoma (NPC). METHODS Quantitative RT-PCR, western blot, and immunohistochemistry analyses were used to quantify CIP2A expression in NPC cell lines and clinical samples. Kaplan-Meier curves were used to estimate the association between CIP2A expression and patient survival. The functional role of CIP2A in NPC cell lines was evaluated by small interfering RNA-mediated depletion of the protein followed by analyses of cell proliferation and xenograft growth. RESULTS CIP2A levels were upregulated in NPC cell lines and clinical samples at both the mRNA and protein levels (P < 0.01). Patients with high CIP2A expression had poorer overall survival (HR, 1.98; 95% CI, 1.16-3.34; P = 0.01) and poorer disease-free survival (HR, 1.68; 95% CI, 1.07-2.62; P = 0.02) rates than patients with low CIP2A expression. In addition, CIP2A expression status was an independent prognostic indicator for NPC patients. The depletion of CIP2A expression inhibited c-Myc protein expression in NPC cell lines, suppressed cell viability, colony formation, and anchorage-independent growth in vitro, and inhibited xenograft tumor growth in vivo. CONCLUSIONS Our data demonstrate that high CIP2A expression in patients was associated with poor survival in NPC, and depletion of CIP2A expression inhibited NPC cell proliferation and tumor growth. Thus, these results warrant further investigation of CIP2A as a novel therapeutic target for the treatment of NPC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Jun Ma
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, 651 Dongfeng Road East, Guangzhou, People's Republic of China.
| |
Collapse
|
122
|
Tan DSP, Bedard PL, Kuruvilla J, Siu LL, Razak ARA. Promising SINEs for embargoing nuclear-cytoplasmic export as an anticancer strategy. Cancer Discov 2014; 4:527-37. [PMID: 24743138 DOI: 10.1158/2159-8290.cd-13-1005] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In cancer cells, the nuclear-cytoplasmic transport machinery is frequently disrupted, resulting in mislocalization and loss of function for many key regulatory proteins. In this review, the mechanisms by which tumor cells co-opt the nuclear transport machinery to facilitate carcinogenesis, cell survival, drug resistance, and tumor progression will be elucidated, with a particular focus on the role of the nuclear-cytoplasmic export protein. The recent development of a new generation of selective inhibitors of nuclear export (XPO1 antagonists) and how these novel anticancer drugs may bring us closer to the implementation of this therapeutic strategy in the clinic will be discussed.
Collapse
Affiliation(s)
- David S P Tan
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | | | | | | | | |
Collapse
|
123
|
Laine A, Westermarck J. Molecular pathways: harnessing E2F1 regulation for prosenescence therapy in p53-defective cancer cells. Clin Cancer Res 2014; 20:3644-50. [PMID: 24788101 DOI: 10.1158/1078-0432.ccr-13-1942] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Induction of terminal proliferation arrest, senescence, is important for in vivo tumor-suppressive function of p53. Moreover, p53-mutant cells are highly resistant to senescence induction by either oncogenic signaling during cellular transformation or in response to different therapies. Senescence resistance in p53-mutant cells has been attributed mostly to inhibition of the checkpoint function of p53 in response to senescence-inducing stress signals. Here, we review very recent evidence that offers an alternative explanation for senescence resistance in p53-defective cancer cells: p21-mediated E2F1 expression. We discuss the potential relevance of these findings for senescence-inducing therapies and highlight cyclin-dependent kinases (CDK) and mechanisms downstream of retinoblastoma protein (RB) as prospective prosenescence therapeutic targets. In particular, we discuss recent findings indicating an important role for the E2F1-CIP2A feedback loop in causing senescence resistance in p53-compromised cancer cells. We further propose that targeting of the E2F1-CIP2A feedback loop could provide a prosenescence therapeutic approach that is effective in both p53-deficient and RB-deficient cancer cells, which together constitute the great majority of all cancer cells. Diagnostic evaluation of the described senescence resistance mechanisms in human tumors might also be informative for patient stratification for already existing therapies.
Collapse
Affiliation(s)
- Anni Laine
- Authors' Affiliations: Turku Centre for Biotechnology, University of Turku and Åbo Akademi University; and
| | - Jukka Westermarck
- Authors' Affiliations: Turku Centre for Biotechnology, University of Turku and Åbo Akademi University; and Department of Pathology, University of Turku, Turku, Finland
| |
Collapse
|
124
|
Mangos JA, Boyd RL, Loughlin GM, Cockrell A, Fucci R. Transductal fluxes of water and monovalent ions in ferret salivary glands. J Dent Res 1981; 130:231-246. [PMID: 6934197 DOI: 10.1172/jci126390] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 09/25/2019] [Indexed: 12/19/2022] Open
Abstract
The net transductal fluxes of water and monovalent ions were measured in the parotid and submandibular salivary glands of the ferret, Mustela putorius furo, during stimulation of secretion with pilocarpine. The duct systems of these glands were found to be impermeable to water using the split-oil droplet method for stationary microperfusion of lobular and main ducts. The net transductal fluxes of Na+, K+, Cl-, and HCO3- were characterized by analysis of ductal fluid samples obtained simultaneously from the intercalated, lobular, and main ducts of these glands.
Collapse
|