1
|
Yang X, Dai J, Wu C, Liu Z. Alzheimer's Disease and Cancer: Common Targets. Mini Rev Med Chem 2024; 24:983-1000. [PMID: 38037912 DOI: 10.2174/0113895575263108231031132404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/13/2023] [Accepted: 10/09/2023] [Indexed: 12/02/2023]
Abstract
There is growing epidemiologic evidence of an inverse association between cancer and AD. In addition, both cell survival and death are regulated by the same signaling pathways, and their abnormal regulation may be implicated in the occurrence and development of cancer and AD. Research shows that there may be a common molecular mechanism between cancer and AD. This review will discuss the role of GSK3, DAPK1, PP2A, P53 and CB2R in the pathogenesis of cancer and AD and describe the current research status of drug development based on these targets.
Collapse
Affiliation(s)
- Xueqing Yang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Jinlian Dai
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Chenglong Wu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Zongliang Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| |
Collapse
|
2
|
Peris I, Romero-Murillo S, Vicente C, Narla G, Odero MD. Regulation and role of the PP2A-B56 holoenzyme family in cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188953. [PMID: 37437699 DOI: 10.1016/j.bbcan.2023.188953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 07/14/2023]
Abstract
Protein phosphatase 2A (PP2A) inactivation is common in cancer, leading to sustained activation of pro-survival and growth-promoting pathways. PP2A consists of a scaffolding A-subunit, a catalytic C-subunit, and a regulatory B-subunit. The functional complexity of PP2A holoenzymes arises mainly through the vast repertoire of regulatory B-subunits, which determine both their substrate specificity and their subcellular localization. Therefore, a major challenge for developing more effective therapeutic strategies for cancer is to identify the specific PP2A complexes to be targeted. Of note, the development of small molecules specifically directed at PP2A-B56α has opened new therapeutic avenues in both solid and hematological tumors. Here, we focus on the B56/PR61 family of PP2A regulatory subunits, which have a central role in directing PP2A tumor suppressor activity. We provide an overview of the mechanisms controlling the formation and regulation of these complexes, the pathways they control, and the mechanisms underlying their deregulation in cancer.
Collapse
Affiliation(s)
- Irene Peris
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain; Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| | - Silvia Romero-Murillo
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain; Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain
| | - Carmen Vicente
- Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor, MI, USA
| | - Maria D Odero
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain; Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
3
|
Yu H, Zaveri S, Sattar Z, Schaible M, Perez Gandara B, Uddin A, McGarvey LR, Ohlmeyer M, Geraghty P. Protein Phosphatase 2A as a Therapeutic Target in Pulmonary Diseases. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1552. [PMID: 37763671 PMCID: PMC10535831 DOI: 10.3390/medicina59091552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023]
Abstract
New disease targets and medicinal chemistry approaches are urgently needed to develop novel therapeutic strategies for treating pulmonary diseases. Emerging evidence suggests that reduced activity of protein phosphatase 2A (PP2A), a complex heterotrimeric enzyme that regulates dephosphorylation of serine and threonine residues from many proteins, is observed in multiple pulmonary diseases, including lung cancer, smoke-induced chronic obstructive pulmonary disease, alpha-1 antitrypsin deficiency, asthma, and idiopathic pulmonary fibrosis. Loss of PP2A responses is linked to many mechanisms associated with disease progressions, such as senescence, proliferation, inflammation, corticosteroid resistance, enhanced protease responses, and mRNA stability. Therefore, chemical restoration of PP2A may represent a novel treatment for these diseases. This review outlines the potential impact of reduced PP2A activity in pulmonary diseases, endogenous and exogenous inhibitors of PP2A, details the possible PP2A-dependent mechanisms observed in these conditions, and outlines potential therapeutic strategies for treatment. Substantial medicinal chemistry efforts are underway to develop therapeutics targeting PP2A activity. The development of specific activators of PP2A that selectively target PP2A holoenzymes could improve our understanding of the function of PP2A in pulmonary diseases. This may lead to the development of therapeutics for restoring normal PP2A responses within the lung.
Collapse
Affiliation(s)
- Howard Yu
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | - Sahil Zaveri
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | - Zeeshan Sattar
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | - Michael Schaible
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | - Brais Perez Gandara
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | - Anwar Uddin
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | - Lucas R. McGarvey
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | | | - Patrick Geraghty
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| |
Collapse
|
4
|
Bergkamp ND, van Senten JR, Brink HJ, Bebelman MP, van den Bor J, Çobanoğlu TS, Dinkla K, Köster J, Klau G, Siderius M, Smit MJ. A virally encoded GPCR drives glioblastoma through feed-forward activation of the SK1-S1P 1 signaling axis. Sci Signal 2023; 16:eade6737. [PMID: 37582160 DOI: 10.1126/scisignal.ade6737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 07/27/2023] [Indexed: 08/17/2023]
Abstract
The G protein-coupled receptor (GPCR) US28 encoded by the human cytomegalovirus (HCMV) is associated with accelerated progression of glioblastomas, aggressive brain tumors with a generally poor prognosis. Here, we showed that US28 increased the malignancy of U251 glioblastoma cells by enhancing signaling mediated by sphingosine-1-phosphate (S1P), a bioactive lipid that stimulates oncogenic pathways in glioblastoma. US28 expression increased the abundance of the key components of the S1P signaling axis, including an enzyme that generates S1P [sphingosine kinase 1 (SK1)], an S1P receptor [S1P receptor 1 (S1P1)], and S1P itself. Enhanced S1P signaling promoted glioblastoma cell proliferation and survival by activating the kinases AKT and CHK1 and the transcriptional regulators cMYC and STAT3 and by increasing the abundance of cancerous inhibitor of PP2A (CIP2A), driving several feed-forward signaling loops. Inhibition of S1P signaling abrogated the proliferative and anti-apoptotic effects of US28. US28 also activated the S1P signaling axis in HCMV-infected cells. This study uncovers central roles for S1P and CIP2A in feed-forward signaling that contributes to the US28-mediated exacerbation of glioblastoma.
Collapse
Affiliation(s)
- Nick D Bergkamp
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Jeffrey R van Senten
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Hendrik J Brink
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Maarten P Bebelman
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Jelle van den Bor
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Tuğçe S Çobanoğlu
- Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | - Johannes Köster
- Algorithms for Reproducible Bioinformatics, Institute of Human Genetics, Faculty of Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Medical Oncology, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Gunnar Klau
- Algorithmic Bioinformatics, Department of Computer Science, Heinrich Heine University, Düsseldorf, Germany
| | - Marco Siderius
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Martine J Smit
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
5
|
Fujiki H, Sueoka E, Watanabe T, Komori A, Suganuma M. Cancer progression by the okadaic acid class of tumor promoters and endogenous protein inhibitors of PP2A, SET and CIP2A. J Cancer Res Clin Oncol 2023:10.1007/s00432-023-04800-4. [PMID: 37097392 PMCID: PMC10374699 DOI: 10.1007/s00432-023-04800-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 04/15/2023] [Indexed: 04/26/2023]
Abstract
PURPOSE Okadaic acid class of tumor promoters are transformed into endogenous protein inhibitors of PP2A, SET, and CIP2A in human cancers. This indicates that inhibition of PP2A activity is a common mechanism of cancer progression in humans. It is important to study the roles of SET and CIP2A vis-à-vis their clinical significance on the basis of new information gathered from a search of PubMed. RESULTS AND DISCUSSION The first part of this review introduces the carcinogenic roles of TNF-α and IL-1, which are induced by the okadaic acid class of compounds. The second part describes unique features of SET and CIP2A in cancer progression for several types of human cancer: (1) SET-expressing circulating tumor cells (SET-CTCs) in breast cancer, (2) knockdown of CIP2A and increased PP2A activity in chronic myeloid leukemia, (3) CIP2A and epidermal growth factor receptor (EGFR) activity in erlotinib sensitive- and resistant-non-small cell lung cancer, (4) SET antagonist EMQA plus radiation therapy against hepatocellular carcinoma, (5) PP2A inactivation as a common event in colorectal cancer, (6) prostate cancer susceptibility variants, homeobox transcription factor (HOXB13 T) and CIP2A T, and (7) SET inhibitor OP449 for pre-clinical investigation of pancreatic cancer. In the Discussion, the binding complex of SET is briefly introduced, and overexpression of SET and CIP2A proteins is discussed in relation to age-associated chronic inflammation (inflammaging). CONCLUSION This review establishes the concept that inhibition of PP2A activity is a common mechanism of human cancer progression and activation of PP2A activity leads to effective anticancer therapy.
Collapse
Affiliation(s)
- Hirota Fujiki
- Department of Clinical Laboratory Medicine, Faculty of Medicine, Saga University, Nabeshima, Saga, 849-8501, Japan.
| | - Eisaburo Sueoka
- Department of Clinical Laboratory Medicine, Faculty of Medicine, Saga University, Nabeshima, Saga, 849-8501, Japan
| | - Tatsuro Watanabe
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Nabeshima, Saga, 849-8501, Japan
| | - Atsumasa Komori
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center and Department of Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Omura, Nagasaki, 856-8562, Japan
| | - Masami Suganuma
- Department of Strategic Research, Graduate School of Science and Engineering, Saitama University, Saitama, 338-8570, Japan
| |
Collapse
|
6
|
Chen B, Hu H, Chen X. From Basic Science to Clinical Practice: The Role of Cancerous Inhibitor of Protein Phosphatase 2A (CIP2A)/p90 in Cancer. Front Genet 2023; 14:1110656. [PMID: 36911405 PMCID: PMC9998691 DOI: 10.3389/fgene.2023.1110656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/03/2023] [Indexed: 03/14/2023] Open
Abstract
Cancerous inhibitor of protein phosphatase 2A (CIP2A), initially reported as a tumor-associated antigen (known as p90), is highly expressed in most solid and hematological tumors. The interaction of CIP2A/p90, protein phosphatase 2A (PP2A), and c-Myc can hinder the function of PP2A toward c-Myc S62 induction, thus stabilizing c-Myc protein, which represents a potential role of CIP2A/p90 in tumorigeneses such as cell proliferation, invasion, and migration, as well as cancer drug resistance. The signaling pathways and regulation networks of CIP2A/p90 are complex and not yet fully understood. Many previous studies have also demonstrated that CIP2A/p90 can be used as a potential therapeutic cancer target. In addition, the autoantibody against CIP2A/p90 in sera may be used as a promising biomarker in the diagnosis of certain types of cancer. In this Review, we focus on recent advances relating to CIP2A/p90 and their implications for future research.
Collapse
Affiliation(s)
- Beibei Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China.,Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| | - Huihui Hu
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China.,Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| | - Xiaobing Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China.,Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| |
Collapse
|
7
|
Hu W, Liuyang Z, Tian Y, Liang J, Zhang X, Zhang H, Wang G, Huo Y, Shentu Y, Wang J, Wang X, Lu Y, Westermarck J, Man H, Liu R. CIP2A deficiency promotes depression-like behaviors in mice through inhibition of dendritic arborization. EMBO Rep 2022; 23:e54911. [PMID: 36305233 PMCID: PMC9724669 DOI: 10.15252/embr.202254911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 08/25/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022] Open
Abstract
Major depressive disorder (MDD) is a severe mental illness. Decreased brain plasticity and dendritic fields have been consistently found in MDD patients and animal models; however, the underlying molecular mechanisms remain to be clarified. Here, we demonstrate that the deletion of cancerous inhibitor of PP2A (CIP2A), an endogenous inhibitor of protein phosphatase 2A (PP2A), leads to depression-like behaviors in mice. Hippocampal RNA sequencing analysis of CIP2A knockout mice shows alterations in the PI3K-AKT pathway and central nervous system development. In primary neurons, CIP2A stimulates AKT activity and promotes dendritic development. Further analysis reveals that the effect of CIP2A in promoting dendritic development is dependent on PP2A-AKT signaling. In vivo, CIP2A deficiency-induced depression-like behaviors and impaired dendritic arborization are rescued by AKT activation. Decreased CIP2A expression and impaired dendrite branching are observed in a mouse model of chronic unpredictable mild stress (CUMS). Indicative of clinical relevance to humans, CIP2A expression is found decreased in transcriptomes from MDD patients. In conclusion, we discover a novel mechanism that CIP2A deficiency promotes depression through the regulation of PP2A-AKT signaling and dendritic arborization.
Collapse
Affiliation(s)
- Wen‐Ting Hu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of PathologyPeking University Shenzhen HospitalShenzhenChina
| | - Zhen‐Yu Liuyang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of General Surgery, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Yuan Tian
- Department of BiologyBoston UniversityBostonUSA
| | - Jia‐Wei Liang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiao‐Lin Zhang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hui‐Liang Zhang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Guan Wang
- Department of BiologyBoston UniversityBostonUSA
| | - Yuda Huo
- Department of BiologyBoston UniversityBostonUSA
| | - Yang‐Ping Shentu
- Department of NephrologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Jian‐Zhi Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiao‐Chuan Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - You‐ming Lu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- The Institute of Brain Research, Collaborative Innovation Center for Brain ScienceHuazhong University of Science and TechnologyWuhanChina
| | - Jukka Westermarck
- Turku Bioscience CentreUniversity of TurkuTurkuFinland
- Åbo Akademi UniversityTurkuFinland
- Institute of BiomedicineUniversity of TurkuTurkuFinland
| | - Heng‐Ye Man
- Department of BiologyBoston UniversityBostonUSA
| | - Rong Liu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- The Institute of Brain Research, Collaborative Innovation Center for Brain ScienceHuazhong University of Science and TechnologyWuhanChina
- Department of Pediatrics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
8
|
Prodigiosin inhibits the proliferation of glioblastoma by regulating the KIAA1524/PP2A signaling pathway. Sci Rep 2022; 12:18527. [PMID: 36323805 PMCID: PMC9630538 DOI: 10.1038/s41598-022-23186-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 10/26/2022] [Indexed: 11/20/2022] Open
Abstract
Prodigiosin (PG), a member of a family of natural red pigments produced by a variety of bacteria, was first discovered in Serratia marcescens. PG has been reported to have an apoptosis-inducing effect in many cancers, such as lymphoma, colon cancer and nasopharyngeal carcinoma. For this study, we used three glioblastoma (GBM) cell lines (LN229, U251 and A172) to explore the effect of prodigiosin on GBM cells. A CCK8 assay was used to evaluate cell viability. We determinedthe cell cycle distribution by flow cytometry and measured proliferation by an EdU incorporation assay. The expression of different molecules was investigated by western blotting and RT-PCR. We further confirmed our results by plasmid transfection and lentiviral transduction. The LN229 xenograft model was used to study the effect of prodigiosin in vivo. We confirmed that prodigiosin played an anticancer role in several GBM cell lines through the KIAA1524/PP2A/Akt signalling pathway. Prodigiosin inhibited the protein expression of KIAA1524 by suppressing its transcription, which led to activation of PP2A. Afterward, PP2A inhibited the phosphorylation of Akt, thereby inducing increased expression of p53/p21. Furthermore, it was verified that prodigiosin inhibited the KIAA1524/PP2A/Akt axis in vivo in the LN229 xenograft model. These data improve the understanding of the anticancer effects of prodigiosin and further highlight the potential of prodigiosin for the development of anti-glioma drugs.
Collapse
|
9
|
Omeroglu Ulu Z, Bolat ZB, Sahin F. Integrated transcriptome and in vitro analysis revealed anti-proliferative effect of sodium perborate on hepatocellular carcinoma cells. J Trace Elem Med Biol 2022; 73:127011. [PMID: 35716648 DOI: 10.1016/j.jtemb.2022.127011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/20/2022] [Accepted: 05/27/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Hepatocelular carcinoma is one of the leading cancer types with no effective cure as poor prognosis is still a challenging aspect. Thus, alternative therapeutics are necessary to control hepatocelular carcinoma. Boron derivatives such as boric acid (BA), sodium perborate tetrahydrate (SPT) and sodium pentaborate pentahydrate (NaB) have been discovered to have anti-cancer effect. This study investigated the anti-proliferative effects of SPT against hepatocelular carcinoma (HCC) using in vitro and transcriptome approaches. METHODS Cytotoxic level of SPT on cell survival were detected using MTS assay. The apoptotic cell death and cell cycle arrest was determined using Annexin V/PI and cell cycle assay, respectively. Transcriptome analysis was performed using RNA-seq, followed by functional and KEGG pathway enrichment analysis. qPCR was used to validate the different genes. RESULTS SPT treated HepG2 and Hep3B cells induced cytotoxicity having IC50 values of 1.13 mM and 0.91 mM, respectively. SPT caused mitotic arrest in G0/G1 phase at 48 h and subsequent apoptotic cell death. RNA-seq revealed a total number of 822 and 1075 differentially expressed genes (DEGs) which after SPT treatment in HepG2 and Hep3B cells, respectively. Functional and KEGG pathway enrichment results suggested that there are several genes involved to induce apoptosis related pathways. The DEGs in p53 signaling pathway may have closely relationships to the cells apoptosis caused by SPT treatment. qPCR results validated dynamic changes in p53 signaling pathway, DNA replication and cell cycle related genes, such as CDKN1A, SERPINE1, PMAIP1, MCM3, MCM5 and MCM6. CONCLUSION In vitro experiments and RNA-seq analysis show anti-proliferative and apoptotic effect of SPT in HCC cells. Further studies might help in understanding the molecular mechanisms of SPT.
Collapse
Affiliation(s)
- Zehra Omeroglu Ulu
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Kayisdagi Cad., Atasehir, Istanbul 34755, Turkey
| | - Zeynep Busra Bolat
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Kayisdagi Cad., Atasehir, Istanbul 34755, Turkey; Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Istanbul Sabahattin Zaim University, Halkali Cad. 281, Kucukcekmece, Istanbul 34303, Turkey
| | - Fikrettin Sahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Kayisdagi Cad., Atasehir, Istanbul 34755, Turkey.
| |
Collapse
|
10
|
Pan J, Zhou L, Zhang C, Xu Q, Sun Y. Targeting protein phosphatases for the treatment of inflammation-related diseases: From signaling to therapy. Signal Transduct Target Ther 2022; 7:177. [PMID: 35665742 PMCID: PMC9166240 DOI: 10.1038/s41392-022-01038-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/28/2022] [Accepted: 05/25/2022] [Indexed: 11/09/2022] Open
Abstract
Inflammation is the common pathological basis of autoimmune diseases, metabolic diseases, malignant tumors, and other major chronic diseases. Inflammation plays an important role in tissue homeostasis. On one hand, inflammation can sense changes in the tissue environment, induce imbalance of tissue homeostasis, and cause tissue damage. On the other hand, inflammation can also initiate tissue damage repair and maintain normal tissue function by resolving injury and restoring homeostasis. These opposing functions emphasize the significance of accurate regulation of inflammatory homeostasis to ameliorate inflammation-related diseases. Potential mechanisms involve protein phosphorylation modifications by kinases and phosphatases, which have a crucial role in inflammatory homeostasis. The mechanisms by which many kinases resolve inflammation have been well reviewed, whereas a systematic summary of the functions of protein phosphatases in regulating inflammatory homeostasis is lacking. The molecular knowledge of protein phosphatases, and especially the unique biochemical traits of each family member, will be of critical importance for developing drugs that target phosphatases. Here, we provide a comprehensive summary of the structure, the "double-edged sword" function, and the extensive signaling pathways of all protein phosphatases in inflammation-related diseases, as well as their potential inhibitors or activators that can be used in therapeutic interventions in preclinical or clinical trials. We provide an integrated perspective on the current understanding of all the protein phosphatases associated with inflammation-related diseases, with the aim of facilitating the development of drugs that target protein phosphatases for the treatment of inflammation-related diseases.
Collapse
Affiliation(s)
- Jie Pan
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Lisha Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Chenyang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China.
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
11
|
Luque M, Cristóbal I, Sanz-Álvarez M, Santos A, Zazo S, Eroles P, Arpí O, Rovira A, Albanell J, Madoz-Gúrpide J, García-Foncillas J, Rojo F. CIP2A as a Key Regulator for AKT Phosphorylation Has Partial Impact Determining Clinical Outcome in Breast Cancer. J Clin Med 2022; 11:jcm11061610. [PMID: 35329936 PMCID: PMC8955826 DOI: 10.3390/jcm11061610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/03/2022] [Accepted: 03/12/2022] [Indexed: 12/07/2022] Open
Abstract
Together with its reported ability to modulate AKT phosphorylation (p-AKT) status in several tumor types, the oncoprotein CIP2A has been described to induce breast cancer progression and drug resistance. However, the clinical and therapeutic relevance of the CIP2A/AKT interplay in breast cancer remains to be fully clarified. Here, we found high p-AKT levels in 80 out of 220 cases (36.4%), which were associated with negative estrogen receptor expression (p = 0.049) and CIP2A overexpression (p < 0.001). Interestingly, p-AKT determined substantially shorter overall (p = 0.002) and progression-free survival (p = 0.003), and multivariate analyses showed its CIP2A-independent prognostic value. Moreover, its clinical relevance was further confirmed in the triple negative and HER2-positive subgroups after stratifying our series by molecular subtype. Functionally, we confirmed in vitro the role of CIP2A as a regulator of p-AKT levels in breast cancer cell lines, and the importance of the CIP2A/AKT axis was also validated in vivo. Finally, p-AKT also showed a higher predictive value of response to doxorubicin than CIP2A in ex vivo analyses. In conclusion, our findings suggest that CIP2A overexpression is a key contributing event to AKT phosphorylation and highlights the CIP2A/AKT axis as a promising therapeutic target in breast cancer. However, our observations highlight the existence of alternative mechanisms that regulate AKT signaling in a subgroup of breast tumors without altered CIP2A expression that determines its independent value as a marker of poor outcome in this disease.
Collapse
Affiliation(s)
- Melani Luque
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Ion Cristóbal
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, ISS-FJD-UAM, 28040 Madrid, Spain;
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain;
- Correspondence: (I.C.); (F.R.); Tel.: +34-915-504-800 (I.C. & F.R.)
| | - Marta Sanz-Álvarez
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Andrea Santos
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, ISS-FJD-UAM, 28040 Madrid, Spain;
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain;
| | - Sandra Zazo
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Pilar Eroles
- Institute of Health Research INCLIVA, 46010 Valencia, Spain;
| | - Oriol Arpí
- Medical Oncology Department, Hospital del Mar, 08003 Barcelona, Spain; (O.A.); (A.R.); (J.A.)
| | - Ana Rovira
- Medical Oncology Department, Hospital del Mar, 08003 Barcelona, Spain; (O.A.); (A.R.); (J.A.)
| | - Joan Albanell
- Medical Oncology Department, Hospital del Mar, 08003 Barcelona, Spain; (O.A.); (A.R.); (J.A.)
| | - Juan Madoz-Gúrpide
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Jesús García-Foncillas
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain;
| | - Federico Rojo
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
- Correspondence: (I.C.); (F.R.); Tel.: +34-915-504-800 (I.C. & F.R.)
| |
Collapse
|
12
|
Tarek MM, Yahia A, El-Nakib MM, Elhefnawi M. Integrative assessment of CIP2A overexpression and mutational effects in human malignancies identifies possible deleterious variants. Comput Biol Med 2021; 139:104986. [PMID: 34739970 DOI: 10.1016/j.compbiomed.2021.104986] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/23/2021] [Accepted: 10/24/2021] [Indexed: 10/19/2022]
Abstract
KIAA1524 is the gene encoding the human cancerous inhibitor of PP2A (CIP2A) protein which is regarded as a novel target for cancer therapy. It is overexpressed in 65%-90% of tissues in almost all studied human cancers. CIP2A expression correlates with cancer progression, disease aggressivity in lung cancer besides poor survival and resistance to chemotherapy in breast cancer. Herein, a pan-cancer analysis of public gene expression datasets was conducted showing significant upregulation of CIP2A in cancerous and metastatic tissues. CIP2A overexpression also correlated with poor survival of cancer patients. To determine the non-coding variants associated with CIP2A overexpression, 5'UTR and 3'UTR variants were annotated and scored using RegulomeDB and Enformer deep learning model. The 5'UTR variants rs1239349555, rs1576326380, and rs1231839144 were predicted to be potential regulators of CIP2A overexpression scoring best on RegulomeDB annotations with a high "2a" rank of supporting experimental data. These variants also scored the highest on Enformer predictions. Analysis of the 3'UTR variants of CIP2A predicted rs56255137 and rs58758610 to alter binding sites of hsa-miR-500a-5 and (hsa-miR-3671, hsa-miR-5692a) respectively. Both variants were also found in linkage disequilibrium with rs11709183 and rs147863209 respectively at r2 ≥ 0.8. The aforementioned variants were found to be eQTL hits significantly associated with CIP2A overexpression. Further, analysis of rs11709183 and rs147863209 revealed a high "2b" rank on RegulomeDB annotations indicating a probable effect on DNAse transcription factors binding. The MuTarget analysis indicated that somatic mutations in TP53 are significantly associated with upregulated CIP2A in human cancers. Analysis of missense SNPs on CIP2A solved structure predicted seven deleterious effects. Four of these variants were also predicted as structurally and functionally destabilizing to CIP2A including; rs375108755, rs147942716, rs368722879, and rs367941403. Variant rs1193091427 was predicted as a potential intronic splicing mutation that might be responsible for the novel CIP2A variant (NOCIVA) in multiple myeloma. Finally, Enrichment of the Wnt/β-catenin pathway within the CIP2A regulatory gene network suggested potential of therapeutic combinations between FTY720 with Wnt/β-catenin, Plk1 and/or HDAC inhibitors to downregulate CIP2A which has been shown to be essential for the survival of different cancer cell lines.
Collapse
Affiliation(s)
- Mohammad M Tarek
- Bioinformatics Department, Armed Forces College of Medicine (AFCM) Cairo, Egypt.
| | - Ahmed Yahia
- Otolaryngology Department, Armed Forces College of Medicine (AFCM) Cairo, Egypt
| | | | - Mahmoud Elhefnawi
- Biomedical Informatics and Chemo-Informatics Group, Centre of Excellence for Medical Research, Informatics and Systems Department, National Research Centre, Cairo, Egypt
| |
Collapse
|
13
|
Penke LRK, Speth J, Wettlaufer S, Draijer C, Peters-Golden M. Bortezomib Inhibits Lung Fibrosis and Fibroblast Activation Without Proteasome Inhibition. Am J Respir Cell Mol Biol 2021; 66:23-37. [PMID: 34236953 DOI: 10.1165/rcmb.2021-0112oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The FDA-approved proteasomal inhibitor bortezomib (BTZ) has attracted interest for its potential anti-fibrotic actions. However, neither its in vivo efficacy in lung fibrosis nor its dependence on proteasome inhibition has been conclusively defined. In this study, we assessed the therapeutic efficacy of BTZ in a mouse model of pulmonary fibrosis, developed an in vitro protocol to define its actions on diverse fibroblast activation parameters, determined its reliance on proteasome inhibition for these actions in vivo and in vitro and explored alternative mechanisms of action. The therapeutic administration of BTZ diminished the severity of pulmonary fibrosis without reducing proteasome activity in the lung. In experiments designed to mimic this lack of proteasome inhibition in vitro, BTZ reduced fibroblast proliferation, differentiation into myofibroblasts, and collagen synthesis. It promoted de-differentiation of myofibroblasts and overcame their characteristic resistance to apoptosis. Mechanistically, BTZ inhibited kinases important for fibroblast activation while inducing expression of dual-specificity phosphatase 1 or DUSP1, and knockdown of DUSP1 abolished its anti-fibrotic actions in fibroblasts. Collectively, these findings suggest that BTZ exhibits a multidimensional profile of robust inhibitory actions on lung fibroblasts as well as anti-fibrotic actions in vivo. Unexpectedly, these actions appear to be independent of proteasome inhibition, and instead attributable to induction of DUSP1.
Collapse
Affiliation(s)
| | - Jennifer Speth
- University of Michigan, 1259, Ann Arbor, Michigan, United States
| | - Scott Wettlaufer
- University of Michigan, 1259, Division of Pulmonary and Critical Care Medicine, Ann Arbor, Michigan, United States
| | | | - Marc Peters-Golden
- University of Michigan Health System, 21707, Ann Arbor, Michigan, United States;
| |
Collapse
|
14
|
Zhu Z, Wei Z. CIP2A silencing alleviates doxorubicin resistance in MCF7/ADR cells through activating PP2A and autophagy. Clin Transl Oncol 2021; 23:1542-1548. [PMID: 33948919 PMCID: PMC8238779 DOI: 10.1007/s12094-021-02616-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/31/2020] [Indexed: 12/15/2022]
Abstract
Background Cancerous inhibitor of protein phosphatase 2A (CIP2A) plays a critical role in the pathogenesis of various types of cancer. Here, we investigated whether manipulating CIP2A abundance could enhance the treatment effects of doxorubicin in MCF-7/ADR cells. Methods CIP2A silencing was achieved by specific siRNAs. Proliferation of breast cancer cell line MCF-7/ADR under effective doxorubicin concentrations after CIP2A silencing was examined by MTT assay. Wound healing assay was performed to quantify cell migration and caspase-3/-7 activities were measured for assessing the extent of apoptosis. Results First, our data confirmed that MCF-7/ADR cell proliferation was suppressed by doxorubicin in a dose-dependent manner. Additionally, knocking down of CIP2A could further decrease MCF-7 cell proliferation and migration, even in the presence of doxorubicin. Mechanistically, we have found that CIP2A silencing promoted cell apoptosis relative to doxorubicin alone or vehicle control groups. Lastly, phosphatase2A (PP2A) activity was potentiated and the autophagy markers, LC3B and Beclin1, were upregulated after knocking down CIP2A. Conclusion Our findings support the potential benefits of using CIP2A inhibitor as a therapeutic agent to treat doxorubicin-resistant breast cancer. Supplementary Information The online version contains supplementary material available at 10.1007/s12094-021-02616-7.
Collapse
Affiliation(s)
- Z Zhu
- Department of Radiotherapy, Cangzhou Central Hospital, No.16 Xinhua West Rd, Cangzhou city, Hebei Province, 061000, China.
| | - Z Wei
- Thyroid and Breast Department, Cangzhou Central Hospital, No.16 Xinhua West Rd, Cangzhou city, Hebei Province, 061000, China
| |
Collapse
|
15
|
Saafan H, Alahdab A, Michelet R, Gohlke L, Ziemann J, Holdenrieder S, McLaughlin KM, Wass MN, Cinatl J, Michaelis M, Kloft C, Ritter CA. Constitutive Cell Proliferation Regulating Inhibitor of Protein Phosphatase 2A (CIP2A) Mediates Drug Resistance to Erlotinib in an EGFR Activating Mutated NSCLC Cell Line. Cells 2021; 10:716. [PMID: 33804833 PMCID: PMC8103245 DOI: 10.3390/cells10040716] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 11/21/2022] Open
Abstract
Exploring mechanisms of drug resistance to targeted small molecule drugs is critical for an extended clinical benefit in the treatment of non-small cell lung cancer (NSCLC) patients carrying activating epidermal growth factor receptor (EGFR) mutations. Here, we identified constitutive cell proliferation regulating inhibitor of protein phosphatase 2A (CIP2A) in the HCC4006rErlo0.5 NSCLC cell line adapted to erlotinib as a model of acquired drug resistance. Constitutive CIP2A resulted in a constitutive activation of Akt signaling. The proteasome inhibitor bortezomib was able to reduce CIP2A levels, which resulted in an activation of protein phosphatase 2A and deactivation of Akt. Combination experiments with erlotinib and bortezomib revealed a lack of interaction between the two drugs. However, the effect size of bortezomib was higher in HCC4006rErlo0.5, compared to the erlotinib-sensitive HCC4006 cells, as indicated by an increase in Emax (0.911 (95%CI 0.867-0.954) vs. 0.585 (95%CI 0.568-0.622), respectively) and decrease in EC50 (52.4 µM (95%CI 46.1-58.8 µM) vs. 73.0 µM (95%CI 60.4-111 µM), respectively) in the concentration-effect model, an earlier onset of cell death induction, and a reduced colony surviving fraction (0.38 ± 0.18 vs. 0.95 ± 0.25, respectively, n = 3, p < 0.05). Therefore, modulation of CIP2A with bortezomib could be an interesting approach to overcome drug resistance to erlotinib treatment in NSCLC.
Collapse
Affiliation(s)
- Hisham Saafan
- Institute of Pharmacy, Clinical Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, 17489 Greifswald, Germany; (H.S.); (A.A.); (L.G.)
| | - Ahmad Alahdab
- Institute of Pharmacy, Clinical Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, 17489 Greifswald, Germany; (H.S.); (A.A.); (L.G.)
| | - Robin Michelet
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 14195 Berlin, Germany; (R.M.); (C.K.)
| | - Linus Gohlke
- Institute of Pharmacy, Clinical Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, 17489 Greifswald, Germany; (H.S.); (A.A.); (L.G.)
| | - Janine Ziemann
- Central Unit for Infection Prevention and Control, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Stefan Holdenrieder
- Institute of Laboratory Medicine, German Heart Center, Munich Technical University, 80636 Munich, Germany;
| | - Katie-May McLaughlin
- Industrial Biotechnology Centre, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK; (K.-M.M.); (M.N.W.); (M.M.)
| | - Mark N. Wass
- Industrial Biotechnology Centre, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK; (K.-M.M.); (M.N.W.); (M.M.)
| | - Jindrich Cinatl
- Institute of Medical Virology, Goethe-University, 60596 Frankfurt am Main, Germany;
| | - Martin Michaelis
- Industrial Biotechnology Centre, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK; (K.-M.M.); (M.N.W.); (M.M.)
| | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 14195 Berlin, Germany; (R.M.); (C.K.)
| | - Christoph A Ritter
- Institute of Pharmacy, Clinical Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, 17489 Greifswald, Germany; (H.S.); (A.A.); (L.G.)
| |
Collapse
|
16
|
The Molecular 'Myc-anisms' Behind Myc-Driven Tumorigenesis and the Relevant Myc-Directed Therapeutics. Int J Mol Sci 2020; 21:ijms21249486. [PMID: 33322239 PMCID: PMC7764474 DOI: 10.3390/ijms21249486] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 02/07/2023] Open
Abstract
MYC, a well-studied proto-oncogene that is overexpressed in >20% of tumors across all cancers, is classically known as “undruggable” due to its crucial roles in cell processes and its lack of a drug binding pocket. Four decades of research and creativity led to the discovery of a myriad of indirect (and now some direct!) therapeutic strategies targeting Myc. This review explores the various mechanisms in which Myc promotes cancer and highlights five key therapeutic approaches to disrupt Myc, including transcription, Myc-Max dimerization, protein stability, cell cycle regulation, and metabolism, in order to develop more specific Myc-directed therapies.
Collapse
|
17
|
Goguet-Rubio P, Amin P, Awal S, Vigneron S, Charrasse S, Mechali F, Labbé JC, Lorca T, Castro A. PP2A-B55 Holoenzyme Regulation and Cancer. Biomolecules 2020; 10:biom10111586. [PMID: 33266510 PMCID: PMC7700614 DOI: 10.3390/biom10111586] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 01/05/2023] Open
Abstract
Protein phosphorylation is a post-translational modification essential for the control of the activity of most enzymes in the cell. This protein modification results from a fine-tuned balance between kinases and phosphatases. PP2A is one of the major serine/threonine phosphatases that is involved in the control of a myriad of different signaling cascades. This enzyme, often misregulated in cancer, is considered a tumor suppressor. In this review, we will focus on PP2A-B55, a particular holoenzyme of the family of the PP2A phosphatases whose specific role in cancer development and progression has only recently been highlighted. The discovery of the Greatwall (Gwl)/Arpp19-ENSA cascade, a new pathway specifically controlling PP2A-B55 activity, has been shown to be frequently altered in cancer. Herein, we will review the current knowledge about the mechanisms controlling the formation and the regulation of the activity of this phosphatase and its misregulation in cancer.
Collapse
|
18
|
Shih CT, Shiau CW, Chen YL, Chen LJ, Chao TI, Wang CY, Huang CY, Hung MH, Chen KF. TD-92, a novel erlotinib derivative, depletes tumor-associated macrophages in non-small cell lung cancer via down-regulation of CSF-1R and enhances the anti-tumor effects of anti-PD-1. Cancer Lett 2020; 498:142-151. [PMID: 33232786 DOI: 10.1016/j.canlet.2020.10.043] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 10/15/2020] [Accepted: 10/26/2020] [Indexed: 01/19/2023]
Abstract
Recent advances in immune checkpoint inhibition, which augment T-cell immune responses, have highlighted the potential of exploiting one's immune system to combat cancer. However, only a relatively small number of non-small cell lung cancer (NSCLC) patients benefit from immune checkpoint blockade due to the immunosuppressive tumor microenvironment. Therefore, combination immunotherapies are now being developed to achieve maximal therapeutic benefits. In this study, we assessed whether a novel erlotinib derivative, TD-92, which possesses anti-tumor effects across several cancer cell lines, could enhance anti-PD-1 treatment. Our results demonstrated that the combined treatment of anti-PD-1 and TD-92 resulted in a potent anti-tumor response in a Lewis lung carcinoma cancer model, as evidenced by the reduced tumor growth and increased survival. Analysis of immune cell population counts revealed that TD-92 reduced the number of pro-tumorigenic CD11b+ F4/80+ tumor-associated macrophages, without significantly affecting the total numbers of other major immunocytes. Further experiments showed that TD-92 induced a marked decline in colony stimulating factor 1 receptor (CSF-1R) expression in macrophage cell lines. The results also suggested that c-Cbl-mediated proteasome degradation was involved in TD-92-mediated CSF-1R downregulation. Our data paves the way for the development of additional combination immunotherapies for NSCLC patients.
Collapse
Affiliation(s)
- Chi-Ting Shih
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Chung-Wai Shiau
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Yen-Lin Chen
- Department of Pathology, Cardinal Tien Hospital, School of Medicine, Fu Jen Catholic University, New Taipei city, Taiwan
| | - Li-Ju Chen
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | | | - Cheng-Yi Wang
- Department of Internal Medicine, Cardinal Tien Hospital and School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.
| | - Chao-Yuan Huang
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Man-Hsin Hung
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan; Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | | |
Collapse
|
19
|
Pecoraro A, Pagano M, Russo G, Russo A. Role of Autophagy in Cancer Cell Response to Nucleolar and Endoplasmic Reticulum Stress. Int J Mol Sci 2020; 21:ijms21197334. [PMID: 33020404 PMCID: PMC7582989 DOI: 10.3390/ijms21197334] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 12/15/2022] Open
Abstract
Eukaryotic cells are exposed to many internal and external stimuli that affect their fate. In particular, the exposure to some of these stimuli induces stress triggering a variety of stress responses aimed to re-establish cellular homeostasis. It is now established that the deregulation of stress response pathways plays a central role in cancer initiation and progression, allowing the adaptation of cells to an altered state in the new environment. Autophagy is a tightly regulated pathway which exerts “housekeeping” role in physiological processes. Recently, a growing amount of evidence highlighted the crucial role of autophagy in the regulation of integrated stress responses, including nucleolar and endoplasmic reticulum. In this review, we attempt to afford an overview of the complex role of nucleolar and endoplasmic reticulum stress-response mechanisms in the regulation of autophagy in cancer and cancer treatment.
Collapse
Affiliation(s)
| | | | - Giulia Russo
- Correspondence: (G.R.); (A.R.); Tel.: +39-081-678415 (G.R.); +39-081-678414 (A.R.)
| | - Annapina Russo
- Correspondence: (G.R.); (A.R.); Tel.: +39-081-678415 (G.R.); +39-081-678414 (A.R.)
| |
Collapse
|
20
|
Saralamma VVG, Vetrivel P, Lee HJ, Kim SM, Ha SE, Murugesan R, Kim EH, Heo JD, Kim GS. Comparative proteomic analysis uncovers potential biomarkers involved in the anticancer effect of Scutellarein in human gastric cancer cells. Oncol Rep 2020; 44:939-958. [PMID: 32705238 PMCID: PMC7388386 DOI: 10.3892/or.2020.7677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 05/28/2020] [Indexed: 02/07/2023] Open
Abstract
Scutellarein (SCU), a flavone that belongs to the flavonoid family and abundantly present in Scutellaria baicalensis a flowering plant in the family Lamiaceae, has been reported to exhibit anticancer effects in several cancer cell lines including gastric cancer (GC). Although our previous study documented the mechanisms of Scutellarein‑induced cytotoxic effects, the literature shows that the proteomic changes that are associated with the cellular response to SCU have been poorly understood. To avoid adverse side‑effects and significant toxicity of chemotherapy in patients who react poorly, biomarkers anticipating therapeutic responses are imperative. In the present study, we utilized a comparative proteomic analysis to identify proteins associated with Scutellarein (SCU)‑induced cell death in GC cells (AGS and SNU484), by integrating two‑dimensional gel electrophoresis (2‑DE), mass spectrometry (MS), and bioinformatics to analyze the proteins. Proteomic analysis between SCU‑treated and DMSO (control) samples successfully identified 41 (AGS) and 31 (SNU484) proteins by MALDI‑TOF/MS analysis and protein database search. Comparative proteomics analysis between AGS and SNU484 cells treated with SCU revealed a total of 7 protein identities commonly expressed and western blot analysis validated a subset of identified critical proteins, which were consistent with those of the 2‑DE outcome. Molecular docking studies also confirmed the binding affinity of SCU towards these critical proteins. Phosphatidylinositol 4,5‑bisphosphate 3‑kinase catalytic subunit β isoform (PIK3CB) protein expression was accompanied by a distinct group of cellular functions, including cell growth, and proliferation. Cancerous inhibitor of protein phosphatase 2A (CIP2A), is one of the oncogenic molecules that have been shown to promote tumor growth and resistance to apoptosis and senescence‑inducing therapies. In the present study, both PIK3CB and CIP2A proteins were downregulated in SCU‑treated cells, which boosts our previous results of SCU to induce apoptosis and inhibits GC cell growth by regulating these critical proteins. The comparative proteomic analysis has yielded candidate biomarkers of response to SCU treatment in GC cell models and further validation of these biomarkers will help the future clinical development of SCU as a novel therapeutic drug.
Collapse
Affiliation(s)
- Venu Venkatarame Gowda Saralamma
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| | - Preethi Vetrivel
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| | - Ho Jeong Lee
- Gyeongnam Department of Environment Toxicology and Chemistry, Biological Resources Research Group, Korea Institute of Toxicology, Jinju, Gyeongnam 52834, Republic of Korea
| | - Seong Min Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| | - Sang Eun Ha
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| | - Rajeswari Murugesan
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu 641043, India
| | - Eun Hee Kim
- Department of Nursing Science, International University of Korea, Jinju, Gyeongnam 52833, Republic of Korea
| | - Jeong Doo Heo
- Gyeongnam Department of Environment Toxicology and Chemistry, Biological Resources Research Group, Korea Institute of Toxicology, Jinju, Gyeongnam 52834, Republic of Korea
| | - Gon Sup Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| |
Collapse
|
21
|
Qi J, Zhou J, Tang XQ, Wang Y. Gene Biomarkers Derived from Clinical Data of Hepatocellular Carcinoma. Interdiscip Sci 2020; 12:226-236. [PMID: 32297074 DOI: 10.1007/s12539-020-00366-8] [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] [Received: 09/09/2019] [Revised: 03/05/2020] [Accepted: 04/02/2020] [Indexed: 12/25/2022]
Abstract
Hepatocellular carcinoma (HCC) is a common cancer of high mortality, mainly due to the difficulty in diagnosis during its clinical stage. Here we aim to find the gene biomarkers, which are of important significance for diagnosis and treatment. In this work, 3682 differentially expressed genes on HCC were firstly differentiated based on the Cancer Genome Atlas database (TCGA). Co-expression modules of these differentially expressed genes were then constructed based on the weighted correlation network algorithm. The correlation coefficient between the co-expression module and clinical data from the Broad GDAC Firehose was thereafter derived. Finally, the interactive network of genes was then constructed. Then, the hub genes were used to implement enrichment analysis and pathway analysis in the Database for Annotation, Visualization and Integrated Discovery (DAVID) database. Results revealed that the abnormally expressed genes in the module played an important role in the biological process including cell division, sister chromatid cohesion, DNA repair, and G1/S transition of mitotic cell cycle. Meanwhile, these genes also enriched in a few crucial pathways related to Cell cycle, Oocyte meiosis, and p53 signaling. Via investigating the closeness centrality of the interactive network, eight gene biomarkers including the CKAP2, TPX2, CDCA8, KIFC1, MELK, SGO1, RACGAP1, and KIAA1524 gene were discovered, whose functions had been indeed revealed to be correlated with HCC. This study, therefore, suggests that the abnormal expression of those eight genes may be taken as gene biomarkers of HCC.
Collapse
Affiliation(s)
- Jiaming Qi
- School of Science, Jiangnan University, Wuxi, 214122, China
| | - Jiaxing Zhou
- School of Science, Jiangnan University, Wuxi, 214122, China
| | - Xu-Qing Tang
- School of Science, Jiangnan University, Wuxi, 214122, China. .,Wuxi Engineering Research Center for Biocomputing, Jiangnan University, Wuxi, 214122, China.
| | - Yaolai Wang
- Wuxi Engineering Research Center for Biocomputing, Jiangnan University, Wuxi, 214122, China
| |
Collapse
|
22
|
FL118 inhibits viability and induces apoptosis of colorectal cancer cells via inactivating the CIP2A/PP2A axis. Life Sci 2019; 239:117074. [PMID: 31751585 DOI: 10.1016/j.lfs.2019.117074] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/07/2019] [Accepted: 11/14/2019] [Indexed: 12/24/2022]
Abstract
AIMS FL118, a novel camptothecin analogue, has been extensively studied for its superior antitumor potency. The aim of this research study is to explore its potential mechanism of action in anti- colorectal cancer (CRC). MAIN METHODS The effect of FL118 on CRC cell proliferation was assessed using CCK-8 assay, while apoptosis was detected using Hoechst staining and Flow cytometry assays. The expression levels of CIP2A were analyzed using qRT-PCR. The expression of CIP2A, PP2A-C, Bax, cleaved caspase-3 and PARP were analyzed using western blotting analysis. The expressions of related proteins in CRC tissues were detected using immunohistochemical staining. TUNEL assay was used to detect apoptosis of tissue. Toxicity of FL118 in primary organs were examined using H&E staining. KEY FINDINGS The results show that FL118 can inhibit the proliferation and clonogenic potential of CRC cells and increase the expression of pro-apoptosis proteins, Bax, cleaved caspase-3 and PARP. Microarray analyses found that FL118 treatment significantly decreases cancerous inhibition of protein phosphatase 2A (CIP2A). Further validation found that CIP2A is aberrantly upregulated in CRC tissues, and is positively correlated with the progression of CRC. In vitro findings confirm that FL118 mediates the downregulation of CIP2A, at both protein and mRNA levels. Co-treatment with Okadaic acid (OA) (a PP2A inhibitor) partially abolishes the anti-proliferative and pro-apoptotic effect of FL118. Consistently, in vivo experiment demonstrates that FL118 can effectively suppress tumorigenesis without any obvious toxic effects. SIGNIFICANCE Collectively, these findings exhibit the anti-neoplastic effects of FL118 against CRC through the down regulation of CIP2A, which subsequently enhances the activity of PP2A.
Collapse
|
23
|
Shen F, Chen Y, Chen L, Qin J, Li Z, Xu J. Amentoflavone Promotes Apoptosis in Non-Small-Cell Lung Cancer by Modulating Cancerous Inhibitor of PP2A. Anat Rec (Hoboken) 2019; 302:2201-2210. [PMID: 31433570 DOI: 10.1002/ar.24229] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/16/2019] [Accepted: 05/07/2019] [Indexed: 12/28/2022]
Abstract
Non-small-cell lung cancer (NSCLC) is one of the most common human malignancies. Amentoflavone (AF) is one of bioflavonoid compounds isolated from Selaginella tamariscina Spring. This study was designed to examine the effect of AF on NSCLC. Our results indicated that AF decreased cell viability of both H1299 and H358 cells. Colony formation assay also showed that AF was able to suppress the anchorage-independent growth of NSCLC cells. AF also triggered cell cycle arrest by downregulating cyclin D1, CDK4, and CDK6. The pro-apoptotic activity of AF was confirmed by Hoechst staining and flow cytometry. The effect of AF on activation of caspase-3, upregulation of Bax, and downregulation of Bcl-2 was examined by western blot. The anti-growth and pro-apoptotic activities of AF were further validated in xenograft murine model. iTRAQ assay showed that cancerous inhibitor of PP2A (CIP2A) expression was markedly downregulated by AF treatment in H1299 cells. In addition, qRT-PCR and western blot also showed that AF was able to dose-dependently inhibit CIP2A expression. Meanwhile, the activity of protein phosphatase 2A (PP2A) was enhanced by AF treatment. The mRNA and protein expression of CIP2A as well as PP2A activity in xenograft tumor tissue were examined, which indicated that the in vivo anticancer activity of AF was associated with downregulation of CIP2A and reactivation of PP2A. Moreover, our results showed that the anti-growth and pro-apoptotic activities of AF were augmented by CIP2A knockdown and attenuated by ectopic CIP2A expression. Our results indicated that AF exhibited anticancer activity in NSCLC by targeting CIP2A. Anat Rec, 302:2201-2210, 2019. © 2019 American Association for Anatomy.
Collapse
Affiliation(s)
- Fei Shen
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yijiang Chen
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liang Chen
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jianwei Qin
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhi Li
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Xu
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
24
|
Gao H, Li Y, Lin T, Cheng Y, Ma Y. Downregulation of CIP2A inhibits cancer cell proliferation and vascularization in renal clear cell carcinoma. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2019; 164:196-202. [PMID: 31431792 DOI: 10.5507/bp.2019.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 06/26/2019] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND CIP2A has been proved to play a role as an oncogene in various types of malignancies while its functionality in renal clear cell carcinoma has not been investigated. Our study aimed to investigate the role of CIP2A in renal clear cell carcinoma and to explore the possible mechanisms. METHODS A total of 80 patients with renal clear cell carcinoma and 32 healthy people were included in the study. Expression of CIP2A was detected by qRT-PCR. CIP2A silencing renal clear cell carcinoma cell line was established. Its effects on cell proliferation and migration were verified by CCK-8 assay and Transwell cell assay, respectively. The effects of CIP2A overexpression on AKT and VEGF were investigated. RESULTS CIP2A expression level was increased in tumor tissues compared to adjacent healthy tissues. Serum levels of CIP2A protein were higher in cancer patients than in healthy controls, and serum levels of CIP2A protein were increased with increased stage of primary tumor. Serum CIP2A protein can be used to accurately predict renal clear cell carcinoma and its prognosis. CIP2A siRNA silencing inhibited tumor cell proliferation, and treatment with Akt activator reduced this inhibitory effect. CIP2A siRNA silencing decreased the expression level of VEGF and phosphorylation levels of AKT in renal clear cell carcinoma cells, while AKT activator treatment showed no significant effects on CIP2A expression. CONCLUSION Downregulation of CIP2A can inhibit cancer cell proliferation and vascularization in renal clear cell carcinoma through inactivation of the Akt pathway and its downstream VEGF.
Collapse
Affiliation(s)
- Hui Gao
- Department of Urology, Liaocheng People's Hospital, Liaocheng City, Shandong Province, P. R. China, 252000
| | - Yuqiao Li
- Department of Urology, Liaocheng People's Hospital, Liaocheng City, Shandong Province, P. R. China, 252000
| | - Tao Lin
- Department of Urology, Liaocheng People's Hospital, Liaocheng City, Shandong Province, P. R. China, 252000
| | - Yan Cheng
- Department of Urology, Liaocheng People's Hospital, Liaocheng City, Shandong Province, P. R. China, 252000
| | - Yunbo Ma
- Department of Urology, Liaocheng People's Hospital, Liaocheng City, Shandong Province, P. R. China, 252000
| |
Collapse
|
25
|
Fang L, Zhang Y, Zang Y, Chai R, Zhong G, Li Z, Duan Z, Ren J, Xu Z. HP-1 inhibits the progression of ccRCC and enhances sunitinib therapeutic effects by suppressing EMT. Carbohydr Polym 2019; 223:115109. [PMID: 31427001 DOI: 10.1016/j.carbpol.2019.115109] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/09/2019] [Accepted: 07/18/2019] [Indexed: 12/29/2022]
Abstract
Trametes robiniophila Murr (Huaier) has been used for many years as an adjuvant treatment for tumors. Sunitinib is the first-line therapy for end-stage renal cancer, but its side effects and drug resistance limit its clinical application. Cell counting kit- 8 (CCK-8), colony formation, scratch, and Transwell assays showed that Huaier polysaccharide (HP-1) reduced tumor progression. Its combination with sunitinib elicited stronger antitumor effects, including induction of apoptosis and cycle arrest. HP-1-induced effects depended on CIP2A downregulation and suppression of the EMT process. Moreover, qPCR and western blotting experiments showed that CIP2A downregulation was particularly pronounced after treatment with the combination therapy and was associated with EMT suppression. In addition, the HP-1/sunitinib combination inhibited the PI3K/Akt/VEGFR pathway, reducing the expression of pathway-related proteins. The HP-1-induced enhancement of sunitinib effects on tumor growth were also observed in vivo in a xenograft mouse model. Overall, these results indicated that HP-1 exerted antitumor effects against clear cell renal cell carcinoma (ccRCC) and enhanced the therapeutic efficacy of sunitinib.
Collapse
Affiliation(s)
- Liang Fang
- Department of Urology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Yongzhen Zhang
- Department of Urology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Yuanwei Zang
- Department of Urology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Rong Chai
- Department of Emergency, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Guangxin Zhong
- Department of Urology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Zeyan Li
- Department of Urology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Zhichen Duan
- Department of Urology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Juchao Ren
- Department of Urology, Qilu Hospital, Shandong University, Jinan 250012, China.
| | - Zhonghua Xu
- Department of Urology, Qilu Hospital, Shandong University, Jinan 250012, China.
| |
Collapse
|
26
|
Remmerie M, Janssens V. PP2A: A Promising Biomarker and Therapeutic Target in Endometrial Cancer. Front Oncol 2019; 9:462. [PMID: 31214504 PMCID: PMC6558005 DOI: 10.3389/fonc.2019.00462] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
Over the last decade, the use of targeted therapies has immensely increased in the treatment of cancer. However, treatment for endometrial carcinomas (ECs) has lagged behind, although potential molecular markers have been identified. This is particularly problematic for the type II ECs, since these aggressive tumors are usually not responsive toward the current standard therapies. Therefore, type II ECs are responsible for most EC-related deaths, indicating the need for new treatment options. Interestingly, molecular analyses of type II ECs have uncovered frequent genetic alterations (up to 40%) in PPP2R1A, encoding the Aα subunit of the tumor suppressive heterotrimeric protein phosphatase type 2A (PP2A). PPP2R1A mutations were also reported in type I ECs and other common gynecologic cancers, albeit at much lower frequencies (0-7%). Nevertheless, PP2A inactivation in the latter cancer types is common via other mechanisms, in particular by increased expression of Cancerous Inhibitor of PP2A (CIP2A) and PP2A Methylesterase-1 (PME-1) proteins. In this review, we discuss the therapeutic potential of direct and indirect PP2A targeting compounds, possibly in combination with other anti-cancer drugs, in EC. Furthermore, we investigate the potential of the PP2A status as a predictive and/or prognostic marker for type I and II ECs.
Collapse
Affiliation(s)
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
27
|
Javadpour P, Dargahi L, Ahmadiani A, Ghasemi R. To be or not to be: PP2A as a dual player in CNS functions, its role in neurodegeneration, and its interaction with brain insulin signaling. Cell Mol Life Sci 2019; 76:2277-2297. [PMID: 30874837 PMCID: PMC11105459 DOI: 10.1007/s00018-019-03063-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 02/16/2019] [Accepted: 03/07/2019] [Indexed: 12/26/2022]
Abstract
Accumulating evidence has reached the consensus that the balance of phosphorylation state of signaling molecules is a pivotal point in the regulation of cell signaling. Therefore, characterizing elements (kinases-phosphatases) in the phosphorylation balance are at great importance. However, the role of phosphatase enzymes is less investigated than kinase enzymes. PP2A is a member of serine/threonine protein phosphatase that its imbalance has been reported in neurodegenerative diseases. Therefore, we reviewed the superfamily of phosphatases and more specifically PP2A, its regulation, and physiological functions participate in CNS. Thereafter, we discussed the latest findings about PP2A dysregulation in Alzheimer and Parkinson diseases and possible interplay between this phosphatase and insulin signaling pathways. Finally, activating/inhibitory modulators for PP2A activity as well as experimental methods for PP2A study have been reviewed.
Collapse
Affiliation(s)
- Pegah Javadpour
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Ghasemi
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Elgenaidi IS, Spiers JP. Regulation of the phosphoprotein phosphatase 2A system and its modulation during oxidative stress: A potential therapeutic target? Pharmacol Ther 2019; 198:68-89. [PMID: 30797822 DOI: 10.1016/j.pharmthera.2019.02.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 02/15/2019] [Indexed: 02/06/2023]
Abstract
Phosphoprotein phosphatases are of growing interest in the pathophysiology of many diseases and are often the neglected partner of protein kinases. One family member, PP2A, accounts for dephosphorylation of ~55-70% of all serine/threonine phosphosites. Interestingly, dysregulation of kinase signalling is a hallmark of many diseases in which an increase in oxidative stress is also noted. With this in mind, we assess the evidence to support oxidative stress-mediated regulation of the PP2A system In this article, we first present an overview of the PP2A system before providing an analysis of the regulation of PP2A by endogenous inhibitors, post translational modification, and miRNA. Next, a detailed critique of data implicating reactive oxygen species, ischaemia, ischaemia-reperfusion, and hypoxia in regulating the PP2A holoenzyme and associated regulators is presented. Finally, the pharmacological targeting of PP2A, its endogenous inhibitors, and enzymes responsible for its post-translational modification are covered. There is extensive evidence that oxidative stress modulates multiple components of the PP2A system, however, most of the data pertains to the catalytic subunit of PP2A. Irrespective of the underlying aetiology, free radical-mediated attenuation of PP2A activity is an emerging theme. However, in many instances, a dichotomy exists, which requires clarification and mechanistic insight. Nevertheless, this raises the possibility that pharmacological activation of PP2A, either through small molecule activators of PP2A or CIP2A/SET antagonists may be beneficial in modulating the cellular response to oxidative stress. A better understanding of which, will have wide ranging implications for cancer, heart disease and inflammatory conditions.
Collapse
Affiliation(s)
- I S Elgenaidi
- Department of Pharmacology and Therapeutics, Trinity College Dublin, Ireland
| | - J P Spiers
- Department of Pharmacology and Therapeutics, Trinity College Dublin, Ireland.
| |
Collapse
|
29
|
Targeting SET to restore PP2A activity disrupts an oncogenic CIP2A-feedforward loop and impairs triple negative breast cancer progression. EBioMedicine 2019; 40:263-275. [PMID: 30651219 PMCID: PMC6412013 DOI: 10.1016/j.ebiom.2018.12.032] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 12/13/2018] [Accepted: 12/17/2018] [Indexed: 12/29/2022] Open
Abstract
Background Triple-negative breast cancer (TNBC) remains difficult to be targeted. SET and cancerous inhibitor of protein phosphatase 2A (CIP2A) are intrinsic protein-interacting inhibitors of protein phosphatase 2A (PP2A) and frequently overexpressed in cancers, whereas reactivating PP2A activity has been postulated as an anti-cancer strategy. Here we explored this strategy in TNBC. Methods Data from The Cancer Genome Atlas (TCGA) database was analyzed. TNBC cell lines were used for in vitro studies. Cell viability was examined by MTT assay. The apoptotic cells were examined by flow cytometry and Western blot. A SET-PP2A protein-protein interaction antagonist TD19 was used to disrupt signal transduction. In vivo efficacy of TD19 was tested in MDA-MB-468-xenografted animal model. Findings TCGA data revealed upregulation of SET and CIP2A and positive correlation of these two gene expressions in TNBC tumors. Ectopic SET or CIP2A increased cell viability, migration, and invasion of TNBC cells. Notably ERK inhibition increased PP2A activity. ERK activation is known crucial for Elk-1 activity, a transcriptional factor regulating CIP2A expression, we hypothesized an oncogenic feedforward loop consisting of pERK/pElk-1/CIP2A/PP2A. This loop was validated by knockdown of PP2A and ectopic expression of Elk-1, showing reciprocal changes in loop members. In addition, ectopic expression of SET increased pAkt, pERK, pElk-1 and CIP2A expressions, suggesting a positive linkage between SET and CIP2A signaling. Moreover, TD19 disrupted this CIP2A-feedforward loop by restoring PP2A activity, demonstrating in vitro and in vivo anti-cancer activity. Mechanistically, TD19 downregulated CIP2A mRNA via inhibiting pERK-mediated Elk-1 nuclear translocation thereby decreased Elk-1 binding to the CIP2A promoter. Interpretation These findings suggested that a novel oncogenic CIP2A-feedforward loop contributes to TNBC progression and targeting SET to disrupt this oncogenic CIP2A loop showed therapeutic potential in TNBC.
Collapse
|
30
|
Huang IT, Dhungel B, Shrestha R, Bridle KR, Crawford DHG, Jayachandran A, Steel JC. Spotlight on Bortezomib: potential in the treatment of hepatocellular carcinoma. Expert Opin Investig Drugs 2018; 28:7-18. [PMID: 30474444 DOI: 10.1080/13543784.2019.1551359] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION This study reviews the evidence for the use of Bortezomib (BZB), a first-in-class proteasome inhibitor in advanced Hepatocellular carcinoma (HCC). This review aims to delineate the role of BZB within the management of non-surgical and metastatic HCC, either as an alternative or as an adjunct to the current treatment paradigm. AREAS COVERED In addition to BZB pharmacology and mechanism of action, safety and tolerance profiles of the drug obtained from clinical trials are explored. The utility of BZB as a therapeutic agent either alone or in combination with other therapies against HCC, including its application in both preclinical and clinical settings has been reviewed. In particular, we highlight the importance of preclinical evaluation of BZB as a combinatorial agent in synergism with other therapies for the use in the management of HCC. EXPERT OPINION There has been much interest surrounding the use of BZB, a first-in-class proteasome inhibitor for HCC therapy. The discernment of outcomes of BZB clinical trials for HCC need to take into consideration the disease-specific factors that can affect survival outcomes including patient selection and aetiological differences. Further preclinical testing of BZB in combination with other therapeutic modalities can be important for eliciting enhanced anti-HCC effects.
Collapse
Affiliation(s)
- I-Tao Huang
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Bijay Dhungel
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Ritu Shrestha
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Kim R Bridle
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Darrell H G Crawford
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Aparna Jayachandran
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Jason C Steel
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,c School of Health, Medical and Applied Sciences , CQ University , Rockhampton , Australia
| |
Collapse
|
31
|
Li W, Zhang H, Yang L, Wang Y. Cancerous inhibitor of protein phosphatase 2A regulates cisplatin resistance in ovarian cancer. Oncol Lett 2018; 17:1211-1216. [PMID: 30655886 DOI: 10.3892/ol.2018.9653] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 02/06/2018] [Indexed: 11/05/2022] Open
Abstract
Ovarian cancer is the most aggressive type of gynecological cancer. The cause of the poor survival rate is the development of chemotherapy resistance to platinum-based therapies, including cisplatin. The present study aimed to investigate the mechanism of cancerous inhibitor of protein phosphatase 2A (CIP2A)-induced chemoresistance in ovarian cancer. The present study initially investigated the expression of CIP2A in the ovarian tumor tissue, cisplatin-sensitive SKOV-3 cell line, and cisplatin-resistant ovarian carcinoma SKOV-3CDDP/R cell line. In addition, CIP2A was knocked down using small interference RNA in ovarian cancer cells and the chemosensitivity of these cells was analyzed. The results demonstrated that CIP2A expression was significantly higher in patients with ovarian cancer and in the cisplatin-resistant ovarian carcinoma SKOV-3CDDP/R cell line at the mRNA and protein levels. The proliferation and chemosensitivity were decreased and enhanced, respectively, when CIP2A was knocked down. CIP2A silencing significantly promoted the apoptosis induced by cisplatin in SKOV-3CDDP/R cells, suggesting that CIP2A participated in the cisplatin resistance of ovarian cancer cells and that CIP2A silencing enhanced the apoptosis induced by cisplatin. CIP2A may be considered as a potential candidate for modulating cisplatin therapy in ovarian cancer.
Collapse
Affiliation(s)
- Wanbin Li
- Department of Gynecology, Jining Medical University, Jining, Shandong 272100, P.R. China
| | - Hongyan Zhang
- Department of Gynecology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272100, P.R. China
| | - Linqing Yang
- Department of Gynecology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272100, P.R. China
| | - Yunfei Wang
- Department of Gynecology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272100, P.R. China
| |
Collapse
|
32
|
Duan J, Zhan JC, Wang GZ, Zhao XC, Huang WD, Zhou GB. The red wine component ellagic acid induces autophagy and exhibits anti-lung cancer activity in vitro and in vivo. J Cell Mol Med 2018; 23:143-154. [PMID: 30353639 PMCID: PMC6307804 DOI: 10.1111/jcmm.13899] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 07/17/2018] [Accepted: 08/14/2018] [Indexed: 12/26/2022] Open
Abstract
Red wine consists of a large amount of compounds such as resveratrol, which exhibits chemopreventive and therapeutic effects against several types of cancers by targeting cancer driver molecules. In this study, we tested the anti‐lung cancer activity of 11 red wine components and reported that a natural polyphenol compound ellagic acid (EA) inhibited lung cancer cell proliferation at an efficacy approximately equal to that of resveratrol. EA markedly increased the expression of the autophagosomal marker LC3‐II as well as inactivation of the mechanistic target of rapamycin signalling pathway. EA elevated autophagy‐associated cell death by down‐regulating the expression of cancerous inhibitor of protein phosphatase 2A (CIP2A), and CIP2A overexpression attenuated EA‐induced autophagy of lung cancer cells. Treating tumour‐bearing mice with EA resulted in significant inhibition of tumour growth with suppression of CIP2A levels and increased autophagy. In addition, EA potentiated the inhibitory effects of the natural compound celastrol on lung cancer cells in vitro and in vivo by enhancing autophagy and down‐regulating CIP2A. These findings indicate that EA may be a promising chemotherapeutic agent for lung cancer, and that the combination of EA and celastrol may have applicability for the treatment of this disease.
Collapse
Affiliation(s)
- Jing Duan
- Beijing Advanced Innovation Center for Food Nutrition and Human Heath, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Ji-Cheng Zhan
- Beijing Advanced Innovation Center for Food Nutrition and Human Heath, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Gui-Zhen Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xin-Chun Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wei-Dong Huang
- Beijing Advanced Innovation Center for Food Nutrition and Human Heath, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Guang-Biao Zhou
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
33
|
Zhang Y, Fang L, Zang Y, Ren J, Xu Z. CIP2A Promotes Proliferation, Invasion and Chemoresistance to Cisplatin in Renal Cell Carcinoma. J Cancer 2018; 9:4029-4038. [PMID: 30410608 PMCID: PMC6218763 DOI: 10.7150/jca.25005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 08/23/2018] [Indexed: 12/15/2022] Open
Abstract
CIP2A is a well-known oncoprotein whose expression is elevated in multiple human solid tumor types. However, its role in renal cell carcinoma (RCC) development is poorly understood. Thus, in our present study, we used the renal cancer cell lines 786-O, A498 and CAKI-1 and the renal epithelial cell line HK-2 to clarify the function of CIP2A in RCC. We found that CIP2A expression is much higher in the RCC cells than in the normal renal epithelial cell. Lentivirus covered coding region CIP2A cDNA sequence and CIP2A siRNA were used to up and down regulate CIP2A expression in vitro. We found that overexpression of CIP2A promoted G1/S transition and cell proliferation. In addition, up-regulation of CIP2A significantly enhanced the invasion and migration capabilities of the cells. Furthermore, CIP2A promoted epithelial-mesenchymal transformation (EMT) and chemoresistance to cisplatin in RCC cells. Taken together, our findings demonstrate that CIP2A plays an important role in proliferation, invasion and chemoresistance to cisplatin in RCC cells. CIP2A may serve as an ideal molecular target for RCC therapeutics.
Collapse
Affiliation(s)
- Yongzhen Zhang
- Department of Urology, Qilu Hospital, Shandong University, 107# Wenhua Xi Road, Jinan 250012, PRC
- Department of Cancer Biology, University of Cincinnati, Cincinnati 45219, USA
| | - Liang Fang
- Department of Urology, Qilu Hospital, Shandong University, 107# Wenhua Xi Road, Jinan 250012, PRC
| | - Yuanwei Zang
- Department of Urology, Qilu Hospital, Shandong University, 107# Wenhua Xi Road, Jinan 250012, PRC
| | - Juchao Ren
- Department of Urology, Qilu Hospital, Shandong University, 107# Wenhua Xi Road, Jinan 250012, PRC
| | - Zhonghua Xu
- Department of Urology, Qilu Hospital, Shandong University, 107# Wenhua Xi Road, Jinan 250012, PRC
| |
Collapse
|
34
|
Wang N, Zhou F, Guo J, Zhu H, Luo S, Cao J. Euxanthone suppresses tumor growth and metastasis in colorectal cancer via targeting CIP2A/PP2A pathway. Life Sci 2018; 209:498-506. [PMID: 30144452 DOI: 10.1016/j.lfs.2018.08.052] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/17/2018] [Accepted: 08/21/2018] [Indexed: 02/06/2023]
Abstract
AIM Colorectal cancer (CRC) accounts for over 600,000 deaths annually worldwide. Euxanthone is a flavonoid compound extracted from Polygala caudata, with documented anti-neoplastic actions. The current study aimed to determine the therapeutic potential of euxanthone in CRC. METHODS AND MATERIALS Cell Counting Kit-8 (CCK-8) assay was used to analyze the effect of euxanthone on the cell viability, and apoptosis was detected by the TUNEL assay. The in vitro migratory capacity was determined by wound healing and the invasiveness was assessed by Transwell assay. Western blotting was used to determine the level of relevant proteins. Furthermore, a CRC xenograft murine model was used to analyze the therapeutic efficacy of euxanthone in vivo. Isobaric tags for relative and absolute quantification (iTRAQ) was then performed to identify the potential targets of euxanthone. To validate the role of cancerous inhibitor of PP2A (CIP2A) in the anti-cancer effects of euxanthone, plasmid overexpressing CIP2A and shRNA targeting CIP2A were used in in vitro assays. KEY FINDINGS Euxanthone decreased cell viability and increased apoptosis in CRC cells, in addition to restraining migration, invasion and EMT. Similarly, euxanthone also effectively suppressed tumor growth and pulmonary metastasis in vivo. iTRAQ analysis identified CIP2A as the primary target responsible for the anticancer effects of euxanthone. The mediatory role of CIP2A was validated when the anticancer activity of euxanthone was significantly blocked by CIP2A overexpression, while CIP2A knockdown sensitized the CRC cells to euxanthone. SIGNIFICANCE Euxanthone exerts anti-cancer effects in vitro and in vivo in CRC by targeting CIP2A/PP2A signaling.
Collapse
Affiliation(s)
- Ning Wang
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China.
| | - Fang Zhou
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Jinhui Guo
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Huaiyuan Zhu
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Shanshui Luo
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | | |
Collapse
|
35
|
Ji J, Zhen W, Si Y, Ma W, Zheng L, Li C, Zhang Y, Qin S, Zhang T, Liu P, Zheng X, Liu Y. Increase in CIP2A expression is associated with cisplatin chemoresistance in gastric cancer. Cancer Biomark 2018; 21:307-316. [PMID: 29103022 DOI: 10.3233/cbm-170416] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The cancerous inhibitor of protein phosphatase 2A (CIP2A) is an oncoprotein which involves in the progression of several human malignancies. Development of cisplatin (DDP) resistance is the obstacle to an effective control of gastric cancer (GC) clinically. OBJECTIVE We thus assessed whether CIP2A expression is associated with sensitivity of GC to DDP. METHODS Real-time quantitative PCR, immunohistochemical analysis, or western blotting was performed to detect CIP2A expression in GC patients' tissues. SGC7901/DDP cells were transfected with CIP2A siRNA. MTT assay was used to determine the DDP-sensitivity of cells. Flow cytometry was used to measure cell apoptosis. RESULTS CIP2A has higher expression in DDP-resistant GC patients. DDP-resistant GC patients with high CIP2A expression presented with poorer overall survival rates than those with low CIP2A expression. CIP2A knockdown in DDP-resistant GC cells resulted in attenuated proliferative abilities and increased apoptosis level. CIP2A depletion sensitizes DDP-resistant cells to DDP and CIP2A overexpression antagonizes DDP-sensitive cells to DDP. CIP2A influences the expression of multidrug resistance-related proteins in GC cells. CONCLUSIONS Our results suggested that CIP2A oncoprotein plays an important role in DDP resistance of GC and could serve as a novel therapeutic target for the treatment of GC patients with DDP resistance.
Collapse
Affiliation(s)
- Juanli Ji
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China.,School of Biomedical Engineering, Hubei University of Medicine, Shiyan, Hubei 442000, China.,Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Weiguo Zhen
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China.,Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Yuan Si
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Wenjing Ma
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China.,School of Biomedical Engineering, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Lanlan Zheng
- Laboratory of Medicinal Plant, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Chen Li
- Laboratory of Medicinal Plant, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Yonghong Zhang
- Laboratory of Medicinal Plant, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Shanshan Qin
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Te Zhang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Pengfei Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China.,School of Biomedical Engineering, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Xin Zheng
- Department of Gastrointestinal Surgery, Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Ying Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China
| |
Collapse
|
36
|
Liu CY, Hsu CC, Huang TT, Lee CH, Chen JL, Yang SH, Jiang JK, Chen WS, Lee KD, Teng HW. ER stress-related ATF6 upregulates CIP2A and contributes to poor prognosis of colon cancer. Mol Oncol 2018; 12:1706-1717. [PMID: 30063110 PMCID: PMC6166000 DOI: 10.1002/1878-0261.12365] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/27/2018] [Accepted: 07/22/2018] [Indexed: 01/20/2023] Open
Abstract
Endoplasmic reticulum (ER) stress is an adaptive response to various stress conditions and plays emerging roles in cancer. Activating transcription factor 6 (ATF6), one of the three major ER stress transducers, has been shown to contribute to chemoresistance by altering cancer cell survival. Cancerous inhibitor of protein phosphatase 2A (CIP2A) is an oncogene, and its expression has been correlated with the prognosis of patients with cancer. In this study, we aimed to explore the relationship between ER stress‐related ATF signaling and CIP2A. We found that CIP2A expression was positively correlated with ATF6 expression by analyzing publicly available RNA sequence data of patients with colorectal cancer (The Cancer Genome Atlas, TCGA). In addition, we demonstrated that tunicamycin‐induced ER stress in vitro upregulated ATF6 and CIP2A. Mechanistically, we found that ATF6 directly bound to the CIP2A promoter and induced CIP2A gene expression, which contributed to colon cancer cell survival. Furthermore, knockdown of CIP2A reduced the viability of cells under ER stress. Most importantly, immunohistochemical analysis of a tissue microarray from a colon cancer patient cohort showed that higher expression levels of ATF6 and CIP2A were associated with a trend toward poor prognosis. Taken together, our results show that ER stress‐related ATF6 upregulates CIP2A and contributes to the prognosis of colon cancer. Targeting CIP2A may disrupt ER stress‐mediated colon cancer cell survival and thus improve the prognosis of patients with colon cancer.
Collapse
Affiliation(s)
- Chun-Yu Liu
- Division of Medical Oncology, Department of Oncology, Center for Immuno-Oncology, Taipei Veterans General Hospital, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taiwan.,Division of Transfusion Medicine, Department of Medicine, Taipei Veterans General Hospital, Taiwan
| | - Chia-Chi Hsu
- Division of Medical Oncology, Department of Oncology, Center for Immuno-Oncology, Taipei Veterans General Hospital, Taiwan.,Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taiwan
| | - Tzu-Ting Huang
- Division of Medical Oncology, Department of Oncology, Center for Immuno-Oncology, Taipei Veterans General Hospital, Taiwan.,Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taiwan
| | - Chia-Han Lee
- Division of Medical Oncology, Department of Oncology, Center for Immuno-Oncology, Taipei Veterans General Hospital, Taiwan
| | - Ji-Lin Chen
- Division of Medical Oncology, Department of Oncology, Center for Immuno-Oncology, Taipei Veterans General Hospital, Taiwan.,Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taiwan
| | - Shung-Haur Yang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Colon and Rectum Surgery, Department of Surgery, Taipei Veterans General Hospital, Taiwan.,Department of Surgery, National Yang-Ming University Hospital, Yilan, Taiwan
| | - Jeng-Kai Jiang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Colon and Rectum Surgery, Department of Surgery, Taipei Veterans General Hospital, Taiwan
| | - Wei-Shone Chen
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Colon and Rectum Surgery, Department of Surgery, Taipei Veterans General Hospital, Taiwan
| | - Kuan-Der Lee
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University Hospital, Taiwan.,School of Medicine, Taipei Medical University, Taiwan
| | - Hao-Wei Teng
- Division of Medical Oncology, Department of Oncology, Center for Immuno-Oncology, Taipei Veterans General Hospital, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
37
|
Gao F, Wang X, Chen S, Xu T, Wang X, Shen Y, Dong F, Zhong S, Shen Z. CIP2A depletion potentiates the chemosensitivity of cisplatin by inducing increased apoptosis in bladder cancer cells. Oncol Rep 2018; 40:2445-2454. [PMID: 30106121 PMCID: PMC6151887 DOI: 10.3892/or.2018.6641] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/17/2018] [Indexed: 01/10/2023] Open
Abstract
Poor response and chemotherapy resistance to cisplatin (DDP)-based therapy frequently lead to treatment failure in advanced bladder cancer; however the underlying mechanism is extremely complex and unclear. Furthermore, cancerous inhibitor of protein phosphatase 2A (CIP2A), a recently identified human oncoprotein, has been shown to play important regulatory roles in cancer cell survival. The present study aimed to investigate the correlation of CIP2A with sensitivity to DDP in bladder cancer cells. In the present study, knockdown of CIP2A was performed using short hairpin-RNA. IC50 determination was used to estimate the chemosensitivity of cells to DDP. Apoptosis and DNA damage indicators were tested in vitro and in vivo to clarify the role of CIP2A in enhancing DDP sensitivity. We observed that CIP2A knockdown enhanced DDP sensitivity. CIP2A depletion accelerated the process of DNA damage caused by DDP treatment. Furthermore, DDP triggered inhibition of CIP2A by preventing AKT Ser473 phosphorylation. In vivo, CIP2A suppression increased the cytotoxicity of DDP, which resulted in a decrease in the subcutaneous tumor growth in a xenograft mouse model. Our findings revealed that the mechanism underlying the involvement of CIP2A in DDP sensitivity enhancement is that CIP2A mediates DDP-induced cell apoptosis and DNA damage. CIP2A is a potential target to improve the response to DDP-based therapy in bladder cancer patients.
Collapse
Affiliation(s)
- Fengbin Gao
- Department of Urology, Ruijin Hospital Affiliated to The School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Xiaojing Wang
- Department of Urology, Ruijin Hospital Affiliated to The School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Shanwen Chen
- Department of Urology, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, P.R. China
| | - Tianyuan Xu
- Department of Urology, Ruijin Hospital Affiliated to The School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Xianjin Wang
- Department of Urology, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, P.R. China
| | - Yifan Shen
- Department of Urology, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, P.R. China
| | - Fan Dong
- Department of Urology, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, P.R. China
| | - Shan Zhong
- Department of Urology, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, P.R. China
| | - Zhoujun Shen
- Department of Urology, Ruijin Hospital Affiliated to The School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| |
Collapse
|
38
|
Enhanced expression of MycN/CIP2A drives neural crest toward a neural stem cell-like fate: Implications for priming of neuroblastoma. Proc Natl Acad Sci U S A 2018; 115:E7351-E7360. [PMID: 30021854 DOI: 10.1073/pnas.1800039115] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Neuroblastoma is a neural crest-derived childhood tumor of the peripheral nervous system in which MycN amplification is a hallmark of poor prognosis. Here we show that MycN is expressed together with phosphorylation-stabilizing factor CIP2A in regions of the neural plate destined to form the CNS, but MycN is excluded from the neighboring neural crest stem cell domain. Interestingly, ectopic expression of MycN or CIP2A in the neural crest domain biases cells toward CNS-like neural stem cells that express Sox2. Consistent with this, some forms of neuroblastoma have been shown to share transcriptional resemblance with CNS neural stem cells. As high MycN/CIP2A levels correlate with poor prognosis, we posit that a MycN/CIP2A-mediated cell-fate bias may reflect a possible mechanism underlying early priming of some aggressive forms of neuroblastoma. In contrast to MycN, its paralogue cMyc is normally expressed in the neural crest stem cell domain and typically is associated with better overall survival in clinical neuroblastoma, perhaps reflecting a more "normal" neural crest-like state. These data suggest that priming for some forms of aggressive neuroblastoma may occur before neural crest emigration from the CNS and well before sympathoadrenal specification.
Collapse
|
39
|
Huang CY, Hung MH, Shih CT, Hsieh FS, Kuo CW, Tsai MH, Chang SS, Hsiao YJ, Chen LJ, Chao TI, Chen KF. Antagonizing SET Augments the Effects of Radiation Therapy in Hepatocellular Carcinoma through Reactivation of PP2A-Mediated Akt Downregulation. J Pharmacol Exp Ther 2018; 366:410-421. [PMID: 29914877 DOI: 10.1124/jpet.118.249102] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/13/2018] [Indexed: 12/23/2022] Open
Abstract
Increasing evidence suggests that SET functions as an oncoprotein and promotes cancer survival and therapeutic resistance. However, whether SET affects radiation therapy (RT)-mediated anticancer effects has not yet been explored. We investigated the impact of SET on RT sensitivity in hepatocellular carcinoma (HCC). Using colony and hepatosphere formation assays, we found that RT-induced proliferative inhibition was critically associated with SET expression. We next tested a novel SET antagonist, N4-(3-ethynylphenyl)-6,7-dimethoxy-N2-(4-phenoxyphenyl) quinazoline-2,4-diamine (EMQA), in combination with RT. We showed that additive use of EMQA significantly enhanced the effects of RT against HCC in vitro and in vivo. Notably, compared with mice receiving either RT or EMQA alone, the growth of PLC5 xenografted tumor in mice receiving RT plus EMQA was significantly reduced without compromising treatment tolerability. Furthermore, we proved that antagonizing SET to restore protein phosphatase 2A-mediated phospho-Akt (p-AKT) downregulation was responsible for the synergism between EMQA and RT. Our data demonstrate a new oncogenic property of SET and provide preclinical evidence that combining a SET antagonist and RT may be effective for treatment of HCC. Further investigation is warranted to validate the clinical relevance of this approach.
Collapse
Affiliation(s)
- Chao-Yuan Huang
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (C.-Y.H.); Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan (M.-H.H.); Institute of Biopharmaceutical Sciences (C.-T.S.) and School of Medicine (M.-H.H.), National Yang-Ming University, Taipei, Taiwan; Department of Medical Research (F.-S.H., M.-H.T., S.-S.C., Y.-J.H, L.-J.C., K.-F.C.) and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Imaging and Radiological Technology, Yuanpei University, Hsinchu, Taiwan (C.-Y.H., C.-W.K.); and SupremeCure Pharma Inc., Taipei, Taiwan (T.-I.C.)
| | - Man-Hsin Hung
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (C.-Y.H.); Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan (M.-H.H.); Institute of Biopharmaceutical Sciences (C.-T.S.) and School of Medicine (M.-H.H.), National Yang-Ming University, Taipei, Taiwan; Department of Medical Research (F.-S.H., M.-H.T., S.-S.C., Y.-J.H, L.-J.C., K.-F.C.) and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Imaging and Radiological Technology, Yuanpei University, Hsinchu, Taiwan (C.-Y.H., C.-W.K.); and SupremeCure Pharma Inc., Taipei, Taiwan (T.-I.C.)
| | - Chi-Ting Shih
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (C.-Y.H.); Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan (M.-H.H.); Institute of Biopharmaceutical Sciences (C.-T.S.) and School of Medicine (M.-H.H.), National Yang-Ming University, Taipei, Taiwan; Department of Medical Research (F.-S.H., M.-H.T., S.-S.C., Y.-J.H, L.-J.C., K.-F.C.) and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Imaging and Radiological Technology, Yuanpei University, Hsinchu, Taiwan (C.-Y.H., C.-W.K.); and SupremeCure Pharma Inc., Taipei, Taiwan (T.-I.C.)
| | - Feng-Shu Hsieh
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (C.-Y.H.); Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan (M.-H.H.); Institute of Biopharmaceutical Sciences (C.-T.S.) and School of Medicine (M.-H.H.), National Yang-Ming University, Taipei, Taiwan; Department of Medical Research (F.-S.H., M.-H.T., S.-S.C., Y.-J.H, L.-J.C., K.-F.C.) and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Imaging and Radiological Technology, Yuanpei University, Hsinchu, Taiwan (C.-Y.H., C.-W.K.); and SupremeCure Pharma Inc., Taipei, Taiwan (T.-I.C.)
| | - Chiung-Wen Kuo
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (C.-Y.H.); Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan (M.-H.H.); Institute of Biopharmaceutical Sciences (C.-T.S.) and School of Medicine (M.-H.H.), National Yang-Ming University, Taipei, Taiwan; Department of Medical Research (F.-S.H., M.-H.T., S.-S.C., Y.-J.H, L.-J.C., K.-F.C.) and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Imaging and Radiological Technology, Yuanpei University, Hsinchu, Taiwan (C.-Y.H., C.-W.K.); and SupremeCure Pharma Inc., Taipei, Taiwan (T.-I.C.)
| | - Ming-Hsien Tsai
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (C.-Y.H.); Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan (M.-H.H.); Institute of Biopharmaceutical Sciences (C.-T.S.) and School of Medicine (M.-H.H.), National Yang-Ming University, Taipei, Taiwan; Department of Medical Research (F.-S.H., M.-H.T., S.-S.C., Y.-J.H, L.-J.C., K.-F.C.) and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Imaging and Radiological Technology, Yuanpei University, Hsinchu, Taiwan (C.-Y.H., C.-W.K.); and SupremeCure Pharma Inc., Taipei, Taiwan (T.-I.C.)
| | - Shih-Shin Chang
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (C.-Y.H.); Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan (M.-H.H.); Institute of Biopharmaceutical Sciences (C.-T.S.) and School of Medicine (M.-H.H.), National Yang-Ming University, Taipei, Taiwan; Department of Medical Research (F.-S.H., M.-H.T., S.-S.C., Y.-J.H, L.-J.C., K.-F.C.) and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Imaging and Radiological Technology, Yuanpei University, Hsinchu, Taiwan (C.-Y.H., C.-W.K.); and SupremeCure Pharma Inc., Taipei, Taiwan (T.-I.C.)
| | - Yung-Jen Hsiao
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (C.-Y.H.); Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan (M.-H.H.); Institute of Biopharmaceutical Sciences (C.-T.S.) and School of Medicine (M.-H.H.), National Yang-Ming University, Taipei, Taiwan; Department of Medical Research (F.-S.H., M.-H.T., S.-S.C., Y.-J.H, L.-J.C., K.-F.C.) and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Imaging and Radiological Technology, Yuanpei University, Hsinchu, Taiwan (C.-Y.H., C.-W.K.); and SupremeCure Pharma Inc., Taipei, Taiwan (T.-I.C.)
| | - Li-Ju Chen
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (C.-Y.H.); Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan (M.-H.H.); Institute of Biopharmaceutical Sciences (C.-T.S.) and School of Medicine (M.-H.H.), National Yang-Ming University, Taipei, Taiwan; Department of Medical Research (F.-S.H., M.-H.T., S.-S.C., Y.-J.H, L.-J.C., K.-F.C.) and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Imaging and Radiological Technology, Yuanpei University, Hsinchu, Taiwan (C.-Y.H., C.-W.K.); and SupremeCure Pharma Inc., Taipei, Taiwan (T.-I.C.)
| | - Tzu-I Chao
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (C.-Y.H.); Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan (M.-H.H.); Institute of Biopharmaceutical Sciences (C.-T.S.) and School of Medicine (M.-H.H.), National Yang-Ming University, Taipei, Taiwan; Department of Medical Research (F.-S.H., M.-H.T., S.-S.C., Y.-J.H, L.-J.C., K.-F.C.) and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Imaging and Radiological Technology, Yuanpei University, Hsinchu, Taiwan (C.-Y.H., C.-W.K.); and SupremeCure Pharma Inc., Taipei, Taiwan (T.-I.C.)
| | - Kuen-Feng Chen
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (C.-Y.H.); Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan (M.-H.H.); Institute of Biopharmaceutical Sciences (C.-T.S.) and School of Medicine (M.-H.H.), National Yang-Ming University, Taipei, Taiwan; Department of Medical Research (F.-S.H., M.-H.T., S.-S.C., Y.-J.H, L.-J.C., K.-F.C.) and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Imaging and Radiological Technology, Yuanpei University, Hsinchu, Taiwan (C.-Y.H., C.-W.K.); and SupremeCure Pharma Inc., Taipei, Taiwan (T.-I.C.)
| |
Collapse
|
40
|
CIP2A is overexpressed in human endometrioid adenocarcinoma and regulates cell proliferation, invasion and apoptosis. Pathol Res Pract 2018; 214:233-239. [DOI: 10.1016/j.prp.2017.11.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/01/2017] [Accepted: 11/13/2017] [Indexed: 12/22/2022]
|
41
|
CIP2A down regulation enhances the sensitivity of pancreatic cancer cells to gemcitabine. Oncotarget 2018; 7:14831-40. [PMID: 26894380 PMCID: PMC4924755 DOI: 10.18632/oncotarget.7447] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/29/2016] [Indexed: 01/05/2023] Open
Abstract
Cancerous inhibitor of protein phosphatase 2A (CIP2A) is an oncoprotein which participates in inhibiting tumor apoptosis in pancreatic cancer cells. Using immunohistochemical staining, we investigated the expression of CIP2A protein in 72 cases of human pancreatic ductal adenocarcinoma (PDAC) tissue and 27 cases of adjacent normal pancreatic tissue. The positive rate of CIP2A protein expression in pancreatic cancer tissue was70.83 %, which was significantly higher than that in adjacent non- cancerous pancreatic tissue (11.11%). The expression of CIP2A was found to be correlated with TNM stage, but not correlated with age, gender, tumor location, smoking status, alcohol consumption, diabetes, high blood pressure, BMI, tumor size, lymph node metastasis or distant metastases. Kaplan- Meier survival analysis showed that patients with positive CIP2A protein expression had a lower overall survival rate than patients without CIP2A expression. COX regression analysis indicated that expression of CIP2A was an independent prognostic factor for pancreatic ductal adenocarcinoma. In addition, down-regulation of CIP2A inhibited cell proliferation and increased sensitivity to gemcitabine in pancreatic cancer cells by decreasing AKT signaling pathway. Our results indicated that down-regulation of CIP2A could be a novel therapeutic strategy for pancreatic cancer.
Collapse
|
42
|
Cha G, Xu J, Xu X, Li B, Lu S, Nanding A, Hu S, Liu S. High expression of CIP2A protein is associated with tumor aggressiveness in stage I-III NSCLC and correlates with poor prognosis. Onco Targets Ther 2017; 10:5907-5914. [PMID: 29276394 PMCID: PMC5731340 DOI: 10.2147/ott.s148250] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The aim of this work was to examine the expression of cancerous inhibitor of protein phosphatase 2A (CIP2A) in non-small cell lung cancer (NSCLC) and analyze its correlation with clinical outcomes. CIP2A protein levels were detected by immunohistochemistry (IHC). One hundred and eighty-four of 209 (88.3%) primary stage I-III NSCLC specimens and 4 of 38 (10.5%) adjacent normal lung tissue specimens expressed CIP2A protein. High expression of CIP2A was detected in 38.8% (81/209) of the NSCLC specimens. Patients diagnosed histologically with late-stage NSCLC (p<0.001) and malignant nodes (p=0.001) exhibited high CIP2A expression. Univariate analysis using the log-rank test identified CIP2A expression as a prognostic predictor for overall survival (p=0.005). In multivariate analyses using the Cox regression test, CIP2A expression, T stage, N stage, histological type, and chemotherapy were identified as independent prognostic factors (p=0.007, 0.001, 0.003, <0.001, and <0.001, respectively). Furthermore, Kaplan-Meier survival curves demonstrated that high CIP2A expression indicated poor prognosis in the subgroup of patients with squamous cell carcinoma (p=0.008). Similar results were noted in the subgroup of patients with adenocarcinoma, but the results did not reach statistical significance (p=0.084). We also used univariate analysis and multivariate analysis to assess the prognostic factors for overall survival in the subgroup of patients who received postoperative chemotherapy. CIP2A expression was also an independent prognostic factor in NSCLC patients who received postoperative chemotherapy (p=0.009), along with histological type (p=0.001) and N stage (p=0.034). In conclusion, adding to the accumulating evidence, our research suggested that the CIP2A expression is associated with aggressiveness and correlates with poor prognosis in NSCLC. Our findings also indicated that CIP2A might be a potential therapeutic target against NSCLC.
Collapse
Affiliation(s)
- Geqi Cha
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang
| | - Jianyu Xu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang
| | - Xiangying Xu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang
| | - Bin Li
- Department of Plastic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong
| | - Shan Lu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang
| | - Abiyasi Nanding
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Songliu Hu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang
| | - Shilong Liu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang
| |
Collapse
|
43
|
Therapeutic targeting of PP2A. Int J Biochem Cell Biol 2017; 96:182-193. [PMID: 29107183 DOI: 10.1016/j.biocel.2017.10.008] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/13/2017] [Accepted: 10/16/2017] [Indexed: 12/19/2022]
Abstract
Protein phosphatase 2A (PP2A) is a major serine/threonine phosphatase that regulates many cellular processes. Given the central role of PP2A in regulating diverse biological functions and its dysregulation in many diseases, including cancer, PP2A directed therapeutics have become of great interest. The main approaches leveraged thus far can be categorized as follows: 1) inhibiting endogenous inhibitors of PP2A, 2) targeted disruption of post translational modifications on PP2A subunits, or 3) direct targeting of PP2A. Additional insight into the structural, molecular, and biological framework driving the efficacy of these therapeutic strategies will provide a foundation for the refinement and development of novel and clinically tractable PP2A targeted therapies.
Collapse
|
44
|
Kim MO, Choe MH, Yoon YN, Ahn J, Yoo M, Jung KY, An S, Hwang SG, Oh JS, Kim JS. Antihelminthic drug niclosamide inhibits CIP2A and reactivates tumor suppressor protein phosphatase 2A in non-small cell lung cancer cells. Biochem Pharmacol 2017; 144:78-89. [PMID: 28813646 DOI: 10.1016/j.bcp.2017.08.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 08/10/2017] [Indexed: 02/07/2023]
Abstract
Protein phosphatase 2A (PP2A) is a critical tumor suppressor complex responsible for the inactivation of various oncogenes. Recently, PP2A reactivation has emerged asan anticancer strategy. Cancerous inhibitor of protein phosphatase 2A (CIP2A), an endogenous inhibitor of PP2A, is upregulated in many cancer cells, including non-small cell lung cancer (NSCLC) cells. We demonstrated that the antihelminthic drug niclosamide inhibited the expression of CIP2A and reactivated the tumor suppressor PP2A in NSCLC cells. We performed a drug-repurposing screen and identified niclosamide asa CIP2A suppressor in NSCLC cells. Niclosamide inhibited cell proliferation, colony formation, and tumor sphere formation, and induced mitochondrial dysfunction through increased mitochondrial ROS production in NSCLC cells; however, these effects were rescued by CIP2A overexpression, which indicated that the antitumor activity of niclosamide was dependent on CIP2A. We found that niclosamide increased PP2A activity through CIP2A inhibition, which reduced the phosphorylation of several oncogenic proteins. Moreover, we found that a niclosamide analog inhibited CIP2A expression and increased PP2A activity in several types of NSCLC cells. Finally, we showed that other well-known PP2A activators, including forskolin and FTY720, did not inhibit CIP2A and that their activities were not dependent on CIP2A. Collectively, our data suggested that niclosamide effectively suppressed CIP2A expression and subsequently activated PP2A in NSCLC cells. This provided strong evidence for the potential use of niclosamide asa PP2A-activating drug in the clinical treatment of NSCLC.
Collapse
Affiliation(s)
- Myeong-Ok Kim
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea; Molecular-Targeted Drug Research Center and Korea Institute for Skin and Clinical Sciences, Konkuk University, Seoul, South Korea
| | - Min Ho Choe
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea; Department of Life Sciences and Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, South Korea
| | - Yi Na Yoon
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea; Radiological and Medico-Oncological Sciences, University of Science and Technology, South Korea
| | - Jiyeon Ahn
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Minjin Yoo
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology, South Korea
| | - Kwan-Young Jung
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology, South Korea; Center for Medicinal Chemistry, Korea Research Institute of Chemical Technology, Daejeon, South Korea
| | - Sungkwan An
- Molecular-Targeted Drug Research Center and Korea Institute for Skin and Clinical Sciences, Konkuk University, Seoul, South Korea
| | - Sang-Gu Hwang
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Jeong Su Oh
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, South Korea.
| | - Jae-Sung Kim
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea; Radiological and Medico-Oncological Sciences, University of Science and Technology, South Korea.
| |
Collapse
|
45
|
Wu CG, Chen H, Guo F, Yadav VK, Mcilwain SJ, Rowse M, Choudhary A, Lin Z, Li Y, Gu T, Zheng A, Xu Q, Lee W, Resch E, Johnson B, Day J, Ge Y, Ong IM, Burkard ME, Ivarsson Y, Xing Y. PP2A-B' holoenzyme substrate recognition, regulation and role in cytokinesis. Cell Discov 2017; 3:17027. [PMID: 28884018 PMCID: PMC5586252 DOI: 10.1038/celldisc.2017.27] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 07/12/2017] [Indexed: 12/11/2022] Open
Abstract
Protein phosphatase 2A (PP2A) is a major Ser/Thr phosphatase; it forms diverse heterotrimeric holoenzymes that counteract kinase actions. Using a peptidome that tiles the disordered regions of the human proteome, we identified proteins containing [LMFI]xx[ILV]xEx motifs that serve as interaction sites for B′-family PP2A regulatory subunits and holoenzymes. The B′-binding motifs have important roles in substrate recognition and in competitive inhibition of substrate binding. With more than 100 novel ligands identified, we confirmed that the recently identified LxxIxEx B′α-binding motifs serve as common binding sites for B′ subunits with minor variations, and that S/T phosphorylation or D/E residues at positions 2, 7, 8 and 9 of the motifs reinforce interactions. Hundreds of proteins in the human proteome harbor intrinsic or phosphorylation-responsive B′-interaction motifs, and localize at distinct cellular organelles, such as midbody, predicting kinase-facilitated recruitment of PP2A-B′ holoenzymes for tight spatiotemporal control of phosphorylation at mitosis and cytokinesis. Moroever, Polo-like kinase 1-mediated phosphorylation of Cyk4/RACGAP1, a centralspindlin component at the midbody, facilitates binding of both RhoA guanine nucleotide exchange factor (epithelial cell transforming sequence 2 (Ect2)) and PP2A-B′ that in turn dephosphorylates Cyk4 and disrupts Ect2 binding. This feedback signaling loop precisely controls RhoA activation and specifies a restricted region for cleavage furrow ingression. Our results provide a framework for further investigation of diverse signaling circuits formed by PP2A-B′ holoenzymes in various cellular processes.
Collapse
Affiliation(s)
- Cheng-Guo Wu
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA.,Biophysics Program, University of Wisconsin at Madison, Madison, WI, USA
| | - Hui Chen
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Feng Guo
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Vikash K Yadav
- Department of Chemistry-BMC, Uppsala University, Uppsala, Sweden
| | - Sean J Mcilwain
- Biostatistics and Medical Informatics, Wisconsin Institutes of Medical Research, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Michael Rowse
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Alka Choudhary
- Department of Medicine, Hematology/Oncology, UW Carbone Cancer Center, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Ziqing Lin
- Department of Cell and Regenerative Biology, Human Proteomic Program, School of Medicine and Public Health, Madison, WI, USA
| | - Yitong Li
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Tingjia Gu
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Aiping Zheng
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Qingge Xu
- Department of Cell and Regenerative Biology, Human Proteomic Program, School of Medicine and Public Health, Madison, WI, USA
| | - Woojong Lee
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Eduard Resch
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology TMP, Frankfurt am Main, Germany
| | - Benjamin Johnson
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Jenny Day
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Ying Ge
- Department of Cell and Regenerative Biology, Human Proteomic Program, School of Medicine and Public Health, Madison, WI, USA
| | - Irene M Ong
- Biostatistics and Medical Informatics, Wisconsin Institutes of Medical Research, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Mark E Burkard
- Department of Medicine, Hematology/Oncology, UW Carbone Cancer Center, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Ylva Ivarsson
- Department of Chemistry-BMC, Uppsala University, Uppsala, Sweden
| | - Yongna Xing
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA.,Biophysics Program, University of Wisconsin at Madison, Madison, WI, USA
| |
Collapse
|
46
|
Zhao M, Howard EW, Parris AB, Guo Z, Zhao Q, Ma Z, Xing Y, Liu B, Edgerton SM, Thor AD, Yang X. Activation of cancerous inhibitor of PP2A (CIP2A) contributes to lapatinib resistance through induction of CIP2A-Akt feedback loop in ErbB2-positive breast cancer cells. Oncotarget 2017; 8:58847-58864. [PMID: 28938602 PMCID: PMC5601698 DOI: 10.18632/oncotarget.19375] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 07/11/2017] [Indexed: 11/25/2022] Open
Abstract
Lapatinib, a small molecule ErbB2/EGFR inhibitor, is FDA-approved for the treatment of metastatic ErbB2-overexpressing breast cancer; however, lapatinib resistance is an emerging clinical challenge. Understanding the molecular mechanisms of lapatinib-mediated anti-cancer activities and identifying relevant resistance factors are of pivotal significance. Cancerous inhibitor of protein phosphatase 2A (CIP2A) is a recently identified oncoprotein that is overexpressed in breast cancer. Our study investigated the role of CIP2A in the anti-cancer efficacy of lapatinib in ErbB2-overexpressing breast cancer cells. We found that lapatinib concurrently downregulated CIP2A and receptor tyrosine kinase signaling in ErbB2-overexpressing SKBR3 and 78617 cells; however, these effects were attenuated in lapatinib-resistant (LR) cells. CIP2A overexpression rendered SKBR3 and 78617 cells resistant to lapatinib-induced apoptosis and growth inhibition. Conversely, CIP2A knockdown via lentiviral shRNA enhanced cell sensitivity to lapatinib-induced growth inhibition and apoptosis. Results also suggested that lapatinib downregulated CIP2A through regulation of protein stability. We further demonstrated that lapatinib-induced CIP2A downregulation can be recapitulated by LY294002, suggesting that Akt mediates CIP2A upregulation. Importantly, lapatinib induced differential CIP2A downregulation between parental BT474 and BT474/LR cell lines. Moreover, CIP2A shRNA knockdown significantly sensitized the BT474/LR cells to lapatinib. Collectively, our results demonstrate that CIP2A is a molecular target and resistance factor of lapatinib with a critical role in lapatinib-induced cellular responses, including the inhibition of the CIP2A-Akt feedback loop. Further investigation of lapatinib-mediated CIP2A regulation will advance our understanding of lapatinib-associated anti-tumor activities and drug resistance.
Collapse
Affiliation(s)
- Ming Zhao
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biological and Biomedical Sciences, North Carolina Central University, Kannapolis, North Carolina, USA
| | - Erin W Howard
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biological and Biomedical Sciences, North Carolina Central University, Kannapolis, North Carolina, USA
| | - Amanda B Parris
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biological and Biomedical Sciences, North Carolina Central University, Kannapolis, North Carolina, USA
| | - Zhiying Guo
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biological and Biomedical Sciences, North Carolina Central University, Kannapolis, North Carolina, USA
| | - Qingxia Zhao
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biological and Biomedical Sciences, North Carolina Central University, Kannapolis, North Carolina, USA.,Basic Medical College of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Zhikun Ma
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biological and Biomedical Sciences, North Carolina Central University, Kannapolis, North Carolina, USA
| | - Ying Xing
- Basic Medical College of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Bolin Liu
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Susan M Edgerton
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ann D Thor
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Xiaohe Yang
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biological and Biomedical Sciences, North Carolina Central University, Kannapolis, North Carolina, USA.,College of Medicine, Henan University of Sciences and Technology, Luoyang, Henan, P.R. China
| |
Collapse
|
47
|
Dysregulation of the causative genes for hereditary parkinsonism in the midbrain in Parkinson's disease. Mov Disord 2017; 32:1211-1220. [DOI: 10.1002/mds.27019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 02/26/2017] [Accepted: 03/17/2017] [Indexed: 11/07/2022] Open
|
48
|
Liu X, Cao W, Qin S, Zhang T, Zheng J, Dong Y, Ming P, Cheng Q, Lu Z, Guo Y, Zhang B, Liu Y. Overexpression of CIP2A is associated with poor prognosis in multiple myeloma. Signal Transduct Target Ther 2017; 2:17013. [PMID: 29263916 PMCID: PMC5661621 DOI: 10.1038/sigtrans.2017.13] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/04/2017] [Accepted: 03/02/2017] [Indexed: 11/09/2022] Open
Abstract
Cancerous inhibitor of protein phosphatase 2A (CIP2A), an endogenous protein phosphatase 2A (PP2A) inhibitor, has been identified as an oncoprotein in promoting cancer initiation and progression of several types of cancer. However, the expression and the role played by CIP2A in the pathogenesis of multiple myeloma (MM) remain unclear. In this study, we showed that CIP2A was overexpressed in human MM cell lines and MM patients' bone marrow tissues. Clinicopathologic analysis showed that CIP2A expression was significantly correlated with clinical stage and percent of plasma cells in bone marrow. Kaplan-Meier analysis revealed that patients with high CIP2A expression presented with poorer overall survival rates than those with low CIP2A expression. Moreover, CIP2A knockdown in MM cells resulted in attenuated proliferative abilities. In addition, CIP2A depletion sensitizes dexamethasone (Dex)-resistant cells to Dex. The effect of CIP2A on proliferation and Dex therapy was mediated by the inhibition of PP2A, which in turn activated Akt. In vivo studies confirmed that CIP2A regulated MM tumorigenesis and the phosphorylation of Akt. Taken together, our results suggest that CIP2A oncoprotein plays an important role in MM progression and could serve as a prognosis marker and a novel therapeutic target for the treatment of patients with MM.
Collapse
Affiliation(s)
- Xuewen Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences; Hubei University of Medicine, Shiyan, China
| | - Wei Cao
- MOE Key Laboratory of Industrial Fermentation Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Shanshan Qin
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences; Hubei University of Medicine, Shiyan, China
| | - Te Zhang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences; Hubei University of Medicine, Shiyan, China
| | - Junnian Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Ying Dong
- Department of Oncology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Pinghong Ming
- Department of Pathology, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Qian Cheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Zheng Lu
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Yang Guo
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences; Hubei University of Medicine, Shiyan, China
| | - Baofu Zhang
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Ying Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences; Hubei University of Medicine, Shiyan, China
| |
Collapse
|
49
|
EGFR-independent Elk1/CIP2A signalling mediates apoptotic effect of an erlotinib derivative TD52 in triple-negative breast cancer cells. Eur J Cancer 2017; 72:112-123. [DOI: 10.1016/j.ejca.2016.11.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 10/27/2016] [Accepted: 11/15/2016] [Indexed: 12/23/2022]
|
50
|
Li C, Hu J, Li W, Song G, Shen J. Combined bortezomib-based chemotherapy and p53 gene therapy using hollow mesoporous silica nanospheres for p53 mutant non-small cell lung cancer treatment. Biomater Sci 2017; 5:77-88. [DOI: 10.1039/c6bm00449k] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Hollow mesoporous silica nanospheres (HMSN)-based co-delivery of bortezomib (BTZ) and the tumor suppressor gene p53 was developed for p53 signal impaired NSCLC therapy.
Collapse
Affiliation(s)
- Chun Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Materials Science and Engineering
- Donghua University
- Shanghai 201620
- China
| | - Junqing Hu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Materials Science and Engineering
- Donghua University
- Shanghai 201620
- China
| | - Wenyao Li
- School of Material Engineering
- Research & Development Center for Key Technologies of Intelligent Ultra-Intense Laser Processing Equipments
- Shanghai University of Engineering Science
- Shanghai 201620
- China
| | - Guosheng Song
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Materials Science and Engineering
- Donghua University
- Shanghai 201620
- China
| | - Jia Shen
- State Key Laboratory of Molecular Biology; Institute of Biochemistry and Cell Biology
- Shanghai Institutes for Biological Sciences
- Chinese Academy of Sciences
- Shanghai 200031
- China
| |
Collapse
|