1
|
Liu C, Zhang X, Zhang Q, Cao Y, Dong L, Suo F, Dong J, Zhang L, Ma S. CaMKK as a Potential Target for the Natural Product Insecticide Cytisine against Megoura japonica Matsumura. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40331368 DOI: 10.1021/acs.jafc.4c12549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Cytisine, a botanical compound, has strong contact activity against a variety of aphids. However, the target and mechanism of its aphidicidal action remain unclear. In this study, the biochemical characteristics of cytisine against Megoura japonica were tested, and the potential target proteins of cytisine were identified, and further verified by fluorescence quenching and molecular docking. Cytisine can affect the activity of Ca2+, Mg2+-ATPase, and Na+, K+-ATPase, with inhibitory rates of 50.00% and 65.22%, respectively. Sixty-eight and fifty-one major binding proteins were identified by drug affinity responsive target stability (DARTS) and cellular thermal shift assay and mass spectrometry (MS-CETSA), respectively. 125 up-regulated and 68 down-regulated genes were obtained by transcriptome sequencing. By combining the candidate target genes of transcriptomics with the potential target proteins of DARTS and MS-CETSA, CaMKK and PPP2R3B were speculated as potential target proteins. The molecular docking results showed that the binding energies of cytisine to CaMKK and PPP2R3B were -6.61 kcal/mol and -6.53 kcal/mol. The fluorescence intensity of CaMKK protein decreased by 28.50, 34.86, 39.68, 51.00, 55.16, 73.99, and 83.29% after different concentrations of cytisine treatment. This research proved that CaMKK is the potential target of cytisine, providing a new target resource for the creation of new pesticides.
Collapse
Affiliation(s)
- Chunli Liu
- College of Plant Protection/Key Laboratory of Hebei Province for Plant Physiology and Molecular Pathology, Hebei Agricultural University, Baoding 071001, China
| | - Xinxin Zhang
- College of Plant Protection/Key Laboratory of Hebei Province for Plant Physiology and Molecular Pathology, Hebei Agricultural University, Baoding 071001, China
| | - Quanguo Zhang
- Institute of Cereal and Oil Crops, Hebei Academy of Agricalture and Foristry Sciences, Shijiazhuang 050000, China
| | - Yuxin Cao
- College of Plant Protection/Key Laboratory of Hebei Province for Plant Physiology and Molecular Pathology, Hebei Agricultural University, Baoding 071001, China
| | - Lili Dong
- College of Plant Protection/Key Laboratory of Hebei Province for Plant Physiology and Molecular Pathology, Hebei Agricultural University, Baoding 071001, China
| | - Fengyue Suo
- College of Plant Protection/Key Laboratory of Hebei Province for Plant Physiology and Molecular Pathology, Hebei Agricultural University, Baoding 071001, China
| | - Jingao Dong
- College of Plant Protection/Key Laboratory of Hebei Province for Plant Physiology and Molecular Pathology, Hebei Agricultural University, Baoding 071001, China
| | - Lihui Zhang
- College of Plant Protection/Key Laboratory of Hebei Province for Plant Physiology and Molecular Pathology, Hebei Agricultural University, Baoding 071001, China
| | - Shujie Ma
- College of Plant Protection/Key Laboratory of Hebei Province for Plant Physiology and Molecular Pathology, Hebei Agricultural University, Baoding 071001, China
| |
Collapse
|
2
|
Zhang F, Luo W, Liu S, Zhao L, Su Y. Protein phosphatase 2A regulates blood cell proliferation and differentiation in Drosophila larval lymph glands. FEBS J 2024; 291:4558-4580. [PMID: 39185698 DOI: 10.1111/febs.17247] [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: 09/28/2023] [Revised: 05/31/2024] [Accepted: 08/02/2024] [Indexed: 08/27/2024]
Abstract
Protein phosphatase 2A (PP2A), one of the most abundant protein phosphatases, has divergent functions in multiple types of cells. Its inactivation has been closely associated with leukemia diseases. However, the physiological function of PP2A for hematopoiesis has been poorly understood in organisms. Drosophila hematopoiesis parallels the vertebrate counterpart in developmental and functional features but involves a much simpler hematopoietic system. Here, utilizing the Drosophila major larval hematopoietic organ lymph gland, we studied the function of PP2A for hematopoiesis in vivo. By knocking down the expression of Pp2A-29B that encodes the scaffold subunit of the PP2A holoenzyme complex, we found that PP2A silencing in the differentiating hemocytes resulted in their excessive proliferation. Furthermore, this PP2A inhibition downregulated the expression of Smoothened (Smo), a crucial component in the Hedgehog pathway, and smo overexpression was able to rescue the phenotypes of PP2A depletion, indicating that Smo functions as a downstream effector of PP2A to restrict the hemocyte proliferation. PDGF/VEGF-receptor (Pvr) overexpression also restored the Smo expression and lymph gland morphology of PP2A silencing, suggesting a PP2A-Pvr-Smo axis to regulate lymph gland growth and hemocyte proliferation. Moreover, inhibiting PP2A activity in the blood progenitor cells promoted their differentiation, but which was independent with Smo. Together, our data suggested that PP2A plays a dual role in the Drosophila lymph gland by preserving the progenitor population and restraining the hemocyte proliferation, to properly regulate the hematopoietic process.
Collapse
Affiliation(s)
- Fang Zhang
- Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
- College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Wang Luo
- Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
- College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Sumin Liu
- Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
- College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Long Zhao
- Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
- Fisheries College, Ocean University of China, Qingdao, China
| | - Ying Su
- Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
- College of Marine Life Sciences, Ocean University of China, Qingdao, China
| |
Collapse
|
3
|
Fujiwara S. Clinical perspectives of rare ovarian tumors: clear cell ovarian cancer. Jpn J Clin Oncol 2023; 53:664-672. [PMID: 37288485 DOI: 10.1093/jjco/hyad057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/16/2023] [Accepted: 05/20/2023] [Indexed: 06/09/2023] Open
Abstract
Ovarian clear cell carcinoma (OCCC) is a rare and distinct histological type of epithelial ovarian carcinoma in terms of its histopathological, clinical and genetic features. Patients with OCCC are younger and diagnosed at earlier stages than those with the most common histological type-high-grade serous carcinoma. Endometriosis is considered a direct precursor of OCCC. Based on preclinical data, the most frequent gene alternations in OCCC are mutations of AT-rich interaction domain 1A and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha. The prognosis of patients with early-stage OCCC is favorable, whereas patients at an advanced stage or who have the recurrent disease have a dismal prognosis due to OCCC's resistance to standard platinum-based chemotherapy. Despite a lower rate of response due to its resistance to standard platinum-based chemotherapy, the treatment strategy for OCCC resembles that of high-grade serous carcinoma, which includes aggressive cytoreductive surgery and adjuvant platinum-based chemotherapy. Alternative treatment strategies, including biological agents based on molecular characteristics specific to OCCC, are urgently needed. Furthermore, due to its rarity, well-designed collaborative international clinical trials are needed to improve oncologic outcomes and the quality of life in patients with OCCC.
Collapse
Affiliation(s)
- Satoe Fujiwara
- Department of Obstetrics and Gynecology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| |
Collapse
|
4
|
Ganaie IA, Malik MZ, Mangangcha IR, Jain SK, Wajid S. Identification of a survival associated gene trio in chemical induced breast cancer. Biochimie 2023; 208:170-179. [PMID: 36621662 DOI: 10.1016/j.biochi.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 12/10/2022] [Accepted: 01/05/2023] [Indexed: 01/07/2023]
Abstract
Sporadic cases of breast cancer being more prevalent than the hereditary cases, can be largely attributed to environmental pollutants like polycyclic aromatic hydrocarbons (PAHs). The aim of the present study was to identify gene dysregulations and the associations in DMBA (a PAH) induced breast cancer. A breast cancer model was developed in Wistar rats (n = 40), using DMBA. Serum proteomics (2D electrophoresis and MALDI-TOF MS) followed by relative gene expression analysis in mammary glands were conducted to reach to the differential gene signatures. The candidate genes were subjected to survival analysis (by GEPIA2 and KM plotter) and infiltration analysis (by ImmuCellAI) for correlating gene expression with patient survival and immune cell infiltration respectively. Further, the regulatory network investigation (by Cytoscape) was performed to find out the transcription factors (TFs) and miRNAs of the concerned genes. A gene trio (ANXA5, MTG1, PPP2R5B), expressing differentially in early mammary carcinogenesis at 4 months (precancerous stage) till full-fledged cancerous stage (post 6 months) was identified. The altered gene expression was associated with less survival among breast cancer patients (n = 4019). The dysregulated expression also has a correlation with enhanced mammary infiltration of immune cells. Moreover, a regulatory network (comprising of 77 transcription factors and 50 miRNAs) involved in the regulation of candidate genes was also deciphered. The deregulated target genes can therefore be explored for reregulation via identified TFs and miRNAs, and survival thereby improved.
Collapse
Affiliation(s)
- Ishfaq Ahmad Ganaie
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Md Zubbair Malik
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | | | - Swatantra Kumar Jain
- Department of Biochemistry, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard, New Delhi, 110 062, India
| | - Saima Wajid
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
5
|
Tingting Gan, Liu X, Chen X, Shi Y, Wang W. Okadaic Acid Inhibits Protein Phosphatases to Suppress Spermatogonial Cell Proliferation. BIOL BULL+ 2022. [DOI: 10.1134/s1062359022140060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
6
|
Investigation of the Effect of Imatinib and Hydroxyurea Combination Therapy on Hematological Parameters and Gene Expression in Chronic Myeloid Leukemia (CML) Patients. J Clin Med 2022; 11:jcm11174954. [PMID: 36078884 PMCID: PMC9456239 DOI: 10.3390/jcm11174954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 12/24/2022] Open
Abstract
(1) Background: Chronic myeloid leukemia is defined as the neoplastic development of mostly myeloid cells in the bone marrow. Several treatments, including chemotherapy, radiation, hormone treatment, and immunological therapy, can be used to control this condition. The therapeutic impact on leukemic individuals varies, and the response to therapy varies between patients due to disease heterogeneity. The primary goal of this study is to compare the effects of single and Imatinib (IM) and Hydroxyurea (HU) combined treatment on hematological parameters and gene expression in CML patients. (2) Methods: This study was conducted on 51 patients, with chronic myeloid leukemia, who were admitted to Al-Basher hospital in Amman, Jordan, for follow-up. Their hematological parameters were checked and gene expression was measured for (BCL2, PP2A, CIP2A, and WT1). (3) Results: The BCL2 gene was found to be less expressed in both IM and (HU + IM) treatments as compared to the HU group alone, while PP2A gene expression was raised. Such a thing indicates that the outcome of the combined therapy method is not ideal, since PP2A activation causes CML cells to move toward the blast crisis stage. Furthermore, CIP2A gene expression revealed that IM and (HU + IM) had the same therapeutic effect and were more successful in CML patients than HU alone. With regards to the treatment effect on hematological parameters, notably in CML patients in later stages, the combination therapy (HU + IM) raised lymphocyte count, indicating a greater response to the treatment. When compared to single medicines, the combination treatment reduced the proportion of neutrophils to normal reference ranges. Platelet counts, on the other hand, dramatically decreased in both IM and (HU + IM). (4) Conclusion: Because the studied genes (BCL2, PP2A, CIP2A, and WT1) are participating in cell proliferation and death, the findings show that the examined genes are significant to understand the efficacy of various therapies. Furthermore, it was found that there was a clear effect of the clinic-based strategic treatment on hematological indicators such as WBCs, lymphocytes, neutrophils, and platelet counts.
Collapse
|
7
|
Hollenstein DM, Gérecová G, Romanov N, Ferrari J, Veis J, Janschitz M, Beyer R, Schüller C, Ogris E, Hartl M, Ammerer G, Reiter W. A phosphatase-centric mechanism drives stress signaling response. EMBO Rep 2021; 22:e52476. [PMID: 34558777 PMCID: PMC8567219 DOI: 10.15252/embr.202152476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 08/27/2021] [Accepted: 09/03/2021] [Indexed: 12/14/2022] Open
Abstract
Changing environmental cues lead to the adjustment of cellular physiology by phosphorylation signaling networks that typically center around kinases as active effectors and phosphatases as antagonistic elements. Here, we report a signaling mechanism that reverses this principle. Using the hyperosmotic stress response in Saccharomyces cerevisiae as a model system, we find that a phosphatase-driven mechanism causes induction of phosphorylation. The key activating step that triggers this phospho-proteomic response is the Endosulfine-mediated inhibition of protein phosphatase 2A-Cdc55 (PP2ACdc55 ), while we do not observe concurrent kinase activation. In fact, many of the stress-induced phosphorylation sites appear to be direct substrates of the phosphatase, rendering PP2ACdc55 the main downstream effector of a signaling response that operates in parallel and independent of the well-established kinase-centric stress signaling pathways. This response affects multiple cellular processes and is required for stress survival. Our results demonstrate how a phosphatase can assume the role of active downstream effectors during signaling and allow re-evaluating the impact of phosphatases on shaping the phosphorylome.
Collapse
Affiliation(s)
- David Maria Hollenstein
- Department of Biochemistry and Cell BiologyMax Perutz LabsVienna BioCenter (VBC)University of ViennaViennaAustria
| | - Gabriela Gérecová
- Department of Biochemistry and Cell BiologyMax Perutz LabsVienna BioCenter (VBC)University of ViennaViennaAustria
| | | | - Jessica Ferrari
- Department of Biochemistry and Cell BiologyMax Perutz LabsVienna BioCenter (VBC)University of ViennaViennaAustria
| | - Jiri Veis
- Department of Biochemistry and Cell BiologyMax Perutz LabsVienna BioCenter (VBC)University of ViennaViennaAustria
- Center for Medical BiochemistryMax Perutz Labs, Vienna BioCenterMedical University of ViennaViennaAustria
| | - Marion Janschitz
- Department of Biochemistry and Cell BiologyMax Perutz LabsVienna BioCenter (VBC)University of ViennaViennaAustria
| | - Reinhard Beyer
- Department of Applied Genetics and Cell Biology (DAGZ)University of Natural Resources and Life Sciences (BOKU)ViennaAustria
- Research Platform Bioactive Microbial Metabolites (BiMM)Tulln a.d. DonauAustria
| | - Christoph Schüller
- Department of Applied Genetics and Cell Biology (DAGZ)University of Natural Resources and Life Sciences (BOKU)ViennaAustria
- Research Platform Bioactive Microbial Metabolites (BiMM)Tulln a.d. DonauAustria
| | - Egon Ogris
- Center for Medical BiochemistryMax Perutz Labs, Vienna BioCenterMedical University of ViennaViennaAustria
| | - Markus Hartl
- Department of Biochemistry and Cell BiologyMax Perutz LabsVienna BioCenter (VBC)University of ViennaViennaAustria
- Mass Spectrometry FacilityMax Perutz Labs, Vienna BioCenterUniversity of ViennaViennaAustria
| | - Gustav Ammerer
- Department of Biochemistry and Cell BiologyMax Perutz LabsVienna BioCenter (VBC)University of ViennaViennaAustria
| | - Wolfgang Reiter
- Department of Biochemistry and Cell BiologyMax Perutz LabsVienna BioCenter (VBC)University of ViennaViennaAustria
- Mass Spectrometry FacilityMax Perutz Labs, Vienna BioCenterUniversity of ViennaViennaAustria
| |
Collapse
|
8
|
Karkache IY, Damodaran JR, Molstad DHH, Bradley EW. Serine/threonine phosphatases in osteoclastogenesis and bone resorption. Gene 2020; 771:145362. [PMID: 33338510 DOI: 10.1016/j.gene.2020.145362] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 11/24/2020] [Accepted: 12/08/2020] [Indexed: 12/27/2022]
Abstract
Maintenance of optimal bone mass is controlled through the concerted functions of several cell types, including bone resorbing osteoclasts. Osteoclasts function to remove calcified tissue during developmental bone modeling, and degrade bone at sites of damage during bone remodeling. Changes to bone homeostasis can arise with alterations in osteoclastogenesis and/or catabolic activity that are not offset by anabolic activity; thus, factors that regulate osteoclastogenesis and bone resorption are of interest to further our understanding of basic bone biology, and as potential targets for therapeutic intervention. Several key cytokines, including RANKL and M-CSF, as well as co-stimulatory factors elicit kinase signaling cascades that promote osteoclastogenesis. These kinase cascades are offset by the action of protein phosphatases, including members of the serine/threonine phosphatase family. Here we review the functions of serine/threonine phosphatases and their control of osteoclast differentiation and function, while highlighting deficiencies in our understanding of this understudied class of proteins within the field.
Collapse
Affiliation(s)
- Ismael Y Karkache
- Department of Orthopedic Surgery, University of Minnesota, Minneapolis, MN 55455, United States
| | - Jeyaram R Damodaran
- Department of Orthopedic Surgery, University of Minnesota, Minneapolis, MN 55455, United States
| | - David H H Molstad
- Department of Orthopedic Surgery, University of Minnesota, Minneapolis, MN 55455, United States
| | - Elizabeth W Bradley
- Department of Orthopedic Surgery, University of Minnesota, Minneapolis, MN 55455, United States; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, United States.
| |
Collapse
|
9
|
Wei H, Zhang HL, Xie JZ, Meng DL, Wang XC, Ke D, Zeng J, Liu R. Protein Phosphatase 2A as a Drug Target in the Treatment of Cancer and Alzheimer's Disease. Curr Med Sci 2020; 40:1-8. [PMID: 32166659 DOI: 10.1007/s11596-020-2140-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 12/10/2019] [Indexed: 01/22/2023]
Abstract
Protein phosphatase 2A (PP2A) is a major serine/threonine phosphatase which participates in the regulation of multiple cellular processes. As a confirmed tumor suppressor, PP2A activity is downregulated in tumors and its re-activation can induce apoptosis of cancer cells. In the brains of Alzheimer's disease (AD) patients, decreased PP2A activity also plays a key role in promoting tau hyperphosphorylation and Aβ generation. In this review, we discussed compounds aiming at modulating PP2A activity in the treatment of cancer or AD. The upstream factors that inactivate PP2A in diseases have not been fully elucidated and further studies are needed. It will help for the refinement and development of novel and clinically tractable PP2A-targeted compounds or therapies for the treatment of tumor and AD.
Collapse
Affiliation(s)
- Hui Wei
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hui-Liang Zhang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jia-Zhao Xie
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dong-Li Meng
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Chuan Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dan Ke
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Ji Zeng
- Department of Clinic Laboratory, Wuhan Fourth Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Rong Liu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
10
|
Hou TY, Zhou Y, Zhu LS, Wang X, Pang P, Wang DQ, Liuyang ZY, Man H, Lu Y, Zhu LQ, Liu D. Correcting abnormalities in miR-124/PTPN1 signaling rescues tau pathology in Alzheimer's disease. J Neurochem 2020; 154:441-457. [PMID: 31951013 DOI: 10.1111/jnc.14961] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 01/11/2020] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
MicroRNAs have been implicated in diverse physiological and pathological processes. We previously reported that aberrant microRNA-124 (miR-124)/non-receptor-type protein phosphatase 1 (PTPN1) signaling plays an important role in the synaptic disorders associated with Alzheimer's disease (AD). In this study, we further investigated the potential role of miR-124/PTPN1 in the tau pathology of AD. We first treated the mice with intra-hippocampal stereotactic injections. Then, we used quantitative real-time reverse transcription PCR (qRT-PCR) to detect the expression of microRNAs. Western blotting was used to measure the level of PTPN1, the level of tau protein, the phosphorylation of tau at AD-related sites, and alterations in the activity of glycogen synthase kinase 3β (GSK-3β) and protein phosphatase 2 (PP2A). Immunohistochemistry was also used to detect changes in tau phosphorylation levels at AD-related sites and somadendritic aggregation. Soluble and insoluble tau protein was separated by 70% formic acid (FA) extraction to examine tau solubility. Finally, behavioral experiments (including the Morris water maze, fear conditioning, and elevated plus maze) were performed to examine learning and memory ability and emotion-related behavior. We found that artificially replicating the abnormalities in miR-124/PTPN1 signaling induced AD-like tau pathology in the hippocampus of wild-type mice, including hyperphosphorylation at multiple sites, insolubility and somadendritic aggregation, as well as learning/memory deficits. We also found that disruption of miR-124/PTPN1 signaling was caused by the loss of RE1-silencing transcription factor protein, which can be initiated by Aβ insults or oxidative stress, as observed in the brains of P301S mice. Correcting the deregulation of miR-124/PTPN1 signaling rescued the tau pathology and learning/memory impairments in the P301S mice. We also found that miR-124/PTPN1 abnormalities induced activation of glycogen synthase kinase 3 (GSK-3) and inactivation of protein phosphatase 2A (PP2A) by promoting tyrosine phosphorylation, implicating an imbalance in tau kinase/phosphatase. Thus, targeting the miR-124/PTPN1 signaling pathway is a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Tong-Yao Hou
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yang Zhou
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Ling-Shuang Zhu
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Xiong Wang
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Pei Pang
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Ding-Qi Wang
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Zhen-Yu Liuyang
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Hengye Man
- Department of Biology, Boston University, Boston, MA, USA
| | - Youming Lu
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Dan Liu
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China.,Department of Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| |
Collapse
|
11
|
Salehi B, Staniak M, Czopek K, Stępień A, Dua K, Wadhwa R, Kumar Chellappan D, Sytar O, Brestic M, Ganesh Bhat N, Venkatesh Anil Kumar N, del Mar Contreras M, Sharopov F, C. Cho W, Sharifi-Rad J. The Therapeutic Potential of the Labdane Diterpenoid Forskolin. APPLIED SCIENCES 2019; 9:4089. [DOI: 10.3390/app9194089] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Forskolin is mainly found in the root of a plant called Coleus forskohlii (Willd.) Briq., which has been used in the traditional medicine of Indian Ayurvedic and Southeast Asia since ancient times. Forskolin is responsible for the pharmacological activity of this species. Forskolin is a labdane diterpenoid with a wide biological effect. Several studies suggested a positive role of forskolin on heart complications, respiratory disorders, high blood pressure, obesity, and asthma. There are numerous clinical and pre-clinical studies representing the effect of forskolin on the above-mentioned disorders but more clinical studies need to be performed to support its efficacy.
Collapse
Affiliation(s)
- Bahare Salehi
- Student Research Committee, School of Medicine, Bam University of Medical Sciences, Bam 44340847, Iran
| | - Mariola Staniak
- Institute of Soil Science and Plant Cultivation—State Research Institute, Czartoryskich Str. 8, 24-100 Puławy, Poland
| | - Katarzyna Czopek
- Institute of Soil Science and Plant Cultivation—State Research Institute, Czartoryskich Str. 8, 24-100 Puławy, Poland
| | - Anna Stępień
- Institute of Soil Science and Plant Cultivation—State Research Institute, Czartoryskich Str. 8, 24-100 Puławy, Poland
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
- Centre for Inflammation, Centenary Institute, University of Newcastle, Callaghan, NSW 2308, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) & School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Ridhima Wadhwa
- Faculty of Life Science and Biotechnology, South Asian University, Akbar Bhawan, Chanakyapuri, New Delhi 110021, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Oksana Sytar
- Department of Plant Biology Department, Taras Shevchenko National University of Kyiv, Institute of Biology, Volodymyrska str., 64, Kyiv 01033, Ukraine
- Department of Plant Physiology, Slovak University of Agriculture, Nitra, A. Hlinku 2, 94976 Nitra, Slovak
| | - Marian Brestic
- Department of Plant Physiology, Slovak University of Agriculture, Nitra, A. Hlinku 2, 94976 Nitra, Slovak
| | - Namrata Ganesh Bhat
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, India
| | | | - María del Mar Contreras
- Department of Chemical, Environmental and Materials Engineering, University of Jaén, 23071 Jaén, Spain
| | - Farukh Sharopov
- Department of Pharmaceutical Technology, Avicenna Tajik State Medical University, Dushanbe 73400, Tajikistan
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, 30 Gascoigne Road, Hong Kong, China
| | - Javad Sharifi-Rad
- Department of Pharmacology, Faculty of Medicine, Jiroft University of Medical Sciences, Jiroft 7861756447, Iran
| |
Collapse
|
12
|
FTY720 Improves Behavior, Increases Brain Derived Neurotrophic Factor Levels and Reduces α-Synuclein Pathology in Parkinsonian GM2+/- Mice. Neuroscience 2019; 411:1-10. [PMID: 31129200 DOI: 10.1016/j.neuroscience.2019.05.029] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/26/2019] [Accepted: 05/14/2019] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is a progressive aging disorder that affects millions worldwide, thus, disease-modifying-therapies are urgently needed. PD pathology includes α-synuclein (aSyn) accumulation as synucleinopathy. Loss of GM1 gangliosides occurs in PD brain, which is modeled in GM2 synthase transgenic mice. GM2+/- mice have low, not absent GM1 and develop age-onset motor deficits, making them an excellent PD drug testing model. FTY720 (fingolimod) reduces synucleinopathy in A53T aSyn mice and motor dysfunction in 6-OHDA and rotenone PD models, but no one has tested FTY720 in mice that develop age-onset PD-like motor problems. We confirmed that GM2+/-mice had equivalent rotarod, hindlimb reflexes, and adhesive removal functions at 9 mo. From 11 mo, GM2+/- mice received oral FTY720 or vehicle 3x/week to 16 mo. As bladder problems occur in PD, we also assessed GM2+/- bladder function. This allowed us to demonstrate improved motor and bladder function in GM2+/- mice treated with FTY720. By immunoblot, FTY720 reduced levels of proNGF, a biomarker of bladder dysfunction. In humans with PD, arm swing becomes abnormal, and brachial plexus modulates arm swing. Ultrastructure of brachial plexus in wild type and GM2 transgenic mice confirmed abnormal myelination and axons in GM2 transgenics. FTY720 treated GM2+/- brachial plexus sustained myelin associated protein levels and reduced aggregated aSyn and PSer129 aSyn levels. FTY720 increases brain derived neurotrophic factor (BDNF) and we noted increased BDNF in GM2+/- brachial plexus and cerebellum, which contribute to rotarod performance. These findings provide further support for testing low dose FTY720 in patients with PD.
Collapse
|
13
|
Sun WG, Song RP, Wang Y, Zhang YH, Wang HX, Ge S, Liu JR, Liu LX. γ-Tocotrienol-Inhibited Cell Proliferation of Human Gastric Cancer by Regulation of Nuclear Factor-κB Activity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:441-451. [PMID: 30562020 DOI: 10.1021/acs.jafc.8b05832] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
γ-Tocotrienol (γ-T3) exhibits the activity of anticancer via regulating cell signaling pathways. Nuclear factor-κB (NF-κB), one of the crucial pro-inflammatory factors, is involved in the regulation of cell proliferation, apoptosis, invasion, and migration of tumor. In the present study, NF-κB activity inhibited by γ-T3 was investigated in gastric cancer cells. Cell proliferation, NF-κB activity, active protein phosphatase type 2A (PP2A), and ataxia-telangiectasia mutated (ATM) protein were explored using 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT), methylene blue, enzyme-linked immunosorbent assay (ELISA), malachite green, luciferase, and Western blotting assays. The effects of γ-T3 on tumor growth and the expression of NF-κB and PP2A proteins were also further examined by implanting human gastric cancer cells in a BALB/c nude mouse model. The results showed that γ-T3 significantly inhibited the cell proliferation and attenuated the NF-κB activity in vitro and in vivo. γ-T3 dramatically increased PP2A activity and protein expression, which suppressed ATM phosphorylation and its translocation to the cytoplasm in gastric cancer cells. Thus, our findings may provide mechanistic insight into effects of γ-T3 on the regulation of NF-κB activity by a PP2A-dependent mechanism and suggest that PP2A may serve as a molecular target for a potential chemopreventive agent.
Collapse
Affiliation(s)
- Wen-Guang Sun
- International Peace Maternity and Child Health Hospital, School of Medicine , Shanghai JiaoTong University , 910 Hengshan Road , Shanghai 200030 , P. R. China
| | - Rui-Peng Song
- Department of General Surgery , The First Affiliated Hospital of University of Science and Technology , 17 LuJiang Road , LuYang District, HeFei 230031 , P. R. China
| | - Yong Wang
- Harbin Center for Disease Control and Prevention , 30 WeiXing Road , DaoWai District, Harbin 150056 , P. R. China
| | - Ya-Hui Zhang
- International Peace Maternity and Child Health Hospital, School of Medicine , Shanghai JiaoTong University , 910 Hengshan Road , Shanghai 200030 , P. R. China
| | - Hai-Xia Wang
- International Peace Maternity and Child Health Hospital, School of Medicine , Shanghai JiaoTong University , 910 Hengshan Road , Shanghai 200030 , P. R. China
| | - Sheng Ge
- International Peace Maternity and Child Health Hospital, School of Medicine , Shanghai JiaoTong University , 910 Hengshan Road , Shanghai 200030 , P. R. China
| | - Jia-Ren Liu
- Department of Clinical Laboratory , The Fourth Affiliated Hospital of Harbin Medical University , 37 YiYuan Street , NanGang District, Harbin 150001 , P. R. China
| | - Lian-Xin Liu
- Department of General Surgery , The First Affiliated Hospital of University of Science and Technology , 17 LuJiang Road , LuYang District, HeFei 230031 , P. R. China
| |
Collapse
|
14
|
Baskaran R, Velmurugan BK. Protein phosphatase 2A as therapeutic targets in various disease models. Life Sci 2018; 210:40-46. [PMID: 30170071 DOI: 10.1016/j.lfs.2018.08.063] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/25/2018] [Accepted: 08/27/2018] [Indexed: 12/24/2022]
Abstract
There are a large number of signalling pathways responsible for transmitting information within the cell. Although cellular signalling is thought to be majorly governed by protein kinases 'cascade effects'; their antagonists protein phosphatases also play a crucial dual role in signal transduction. By dephosphorylating the proteins involved in signalling pathways, phosphatases may lead to their activation and sometimes they may terminate a signal generated by kinases activity. Due to counterbalancing the function of phosphorylation, the protein phosphatases are very important to signal transduction processes and thus the control of phosphatase activity is as significant as kinases, in the regulation of a plethora of cellular processes. In general, the protein phosphatases are comprised of a catalytic subunit with one or more regulatory and/or targeting subunits associated with it. The Protein Phosphatase 2A (PP2A), a member of serine/threonine phosphatases family, is ubiquitously expressed a remarkably conserved enzyme in the cell. Its catalytic activity has been highly regulated and may have enormous therapeutic potential which is still untapped. It has specificities for a number of substrates which witnessed its involvement in various signalling modules of cell cycle regulation, cell morphology and development. Thus it can be an appropriate target for studying different diseases associated with abnormal signal transduction pathways such as neurodegenerative diseases and malignancies. This review will focus on the structure and regulatory pathways of PP2A. The de-regulation of PP2A in some specific pathology such as Cancer, Heart diseases, Neurodegenerative disorders and Diabetes will also be touched upon.
Collapse
Affiliation(s)
- Rathinasamy Baskaran
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Bharath Kumar Velmurugan
- Toxicology and Biomedicine Research Group, Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam.
| |
Collapse
|
15
|
Westermarck J. Targeted therapies don't work for a reason; the neglected tumor suppressor phosphatasePP2A strikes back. FEBS J 2018; 285:4139-4145. [DOI: 10.1111/febs.14617] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 07/26/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Jukka Westermarck
- Turku Centre for Biotechnology University of Turku and Åbo Akademi University Turku Finland
- Institute of Biomedicine University of Turku Finland
| |
Collapse
|
16
|
Shouse G, de Necochea-Campion R, Mirshahidi S, Liu X, Chen CS. Novel B55α-PP2A mutations in AML promote AKT T308 phosphorylation and sensitivity to AKT inhibitor-induced growth arrest. Oncotarget 2018; 7:61081-61092. [PMID: 27531894 PMCID: PMC5308637 DOI: 10.18632/oncotarget.11209] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 07/27/2016] [Indexed: 01/10/2023] Open
Abstract
Activation of the Protein Kinase B (PKB), or AKT pathway has been shown to correlate with acute myeloid leukemia (AML) prognosis. B55α-Protein Phosphatase 2A (PP2A) has been shown to dephosphorylate AKT at Thr-308 rendering it inactive. In fact, low expression of the PP2A regulatory subunit B55α was associated with activated phospho-AKT and correlated with inferior outcomes in AML. Despite this fact, no studies have specifically demonstrated a mechanism whereby B55α expression is regulated in AML. In this study, we demonstrate novel loss of function mutations in the PPP2R2A gene identified in leukemic blasts from three AML patients. These mutations eliminate B55α protein expression thereby allowing constitutive AKT activation. In addition, leukemic blasts with PPP2R2A gene mutation were more sensitive to treatment with the AKT inhibitor MK2206, but less responsive to the PP2A activator FTY720. Using leukemia cell lines, we further demonstrate that B55α expression correlates with AKT Thr-308 phosphorylation and predicts responsiveness to AKT inhibition and PP2A activation. Together our data illustrate the importance of the B55α-PP2A-AKT pathway in leukemogenesis. Screening for disruptions in this pathway at initial AML diagnosis may predict response to targeted therapies against AKT and PP2A.
Collapse
Affiliation(s)
- Geoffrey Shouse
- Division of Hematology/Oncology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Rosalia de Necochea-Campion
- Loma Linda University Cancer Center, Biospecimen Laboratory, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Saied Mirshahidi
- Loma Linda University Cancer Center, Biospecimen Laboratory, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Xuan Liu
- Department of Biochemistry, University of California, Riverside, Riverside, CA, USA
| | - Chien-Shing Chen
- Division of Hematology/Oncology, Loma Linda University School of Medicine, Loma Linda, CA, USA.,Loma Linda University Cancer Center, Biospecimen Laboratory, Loma Linda University School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
17
|
Yang X, Yan T, Gong Y, Liu X, Sun H, Xu W, Wang C, Naren D, Zheng Y. High CFTR expression in Philadelphia chromosome-positive acute leukemia protects and maintains continuous activation of BCR-ABL and related signaling pathways in combination with PP2A. Oncotarget 2018; 8:24437-24448. [PMID: 28445932 PMCID: PMC5421860 DOI: 10.18632/oncotarget.15510] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 02/06/2017] [Indexed: 02/05/2023] Open
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR) is classified as an anion channel transporter of Cl- and HCO3-. Through interactions with its PDZ domain, CFTR is capable of regulating other proteins, such as protein phosphatase 2A (PP2A). The aberrant expression and mutation of CFTR have been observed in several tumor, but not in philadelphia chromosome-positive(Ph+) acute leukemia, including Ph+ B cell acute lymphoblastic leukemia(Ph+ B-ALL) and chronic myelogenous leukemia blast crisis phases (CML-BC). In this study, we demonstrated the mean expression level of CFTR in Ph+ acute leukemia cells was markedly higher than that in Ph- B-ALL and CML-chronic phase cells. CFTRinh-172, a classic CFTR inhibitor, down-regulated the expression of CFTR, p-BCR-ABL and classical Wnt/β-catenin signaling in Ph+ acute leukemia cells, while imatinib had no effect on CFTR. Importantly, reduced efficacy of CFTRinh-172 was closely associated with elevated PP2A phosphatase activity. Furthermore, we confirmed an interaction between CFTR and the PP2AA subunit in K562 cells. In addition, we demonstrated CFTR and PP2AA interact in the cytosol, resulting in PP2A complex inactivation and increased degradation of PP2A substrates via the lysosomal/proteasome pathway. In conclusion, our results showed CFTR was highly expressed in Ph+ acute leukemia, which protected and maintained the continuous activation of BCR-ABL and the canonical Wnt/β-catenin signaling pathway by decreasing PP2A phosphatase activity. According to this working model of the CFTR-PP2A-BCR-ABL axis, targeting the CFTR protein will activate PP2A and may offer a new treatment strategy for Ph+ acute leukemia, especially for patients exhibiting high levels of CFTR expression.
Collapse
Affiliation(s)
- Xi Yang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Tianyou Yan
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuping Gong
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Xuehua Liu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Huaqin Sun
- Sichuan University-The Chinese University of Hong Kong Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Chengdu, China
| | - Wenming Xu
- Sichuan University-The Chinese University of Hong Kong Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Chengdu, China
| | - Chunsen Wang
- Department of Hematology, Sichuan Provincial People's Hospital, Affiliated Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Duolan Naren
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuhuan Zheng
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Richard NP, Pippa R, Cleary MM, Puri A, Tibbitts D, Mahmood S, Christensen DJ, Jeng S, McWeeney S, Look AT, Chang BH, Tyner JW, Vitek MP, Odero MD, Sears R, Agarwal A. Combined targeting of SET and tyrosine kinases provides an effective therapeutic approach in human T-cell acute lymphoblastic leukemia. Oncotarget 2018; 7:84214-84227. [PMID: 27705940 PMCID: PMC5356656 DOI: 10.18632/oncotarget.12394] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 09/24/2016] [Indexed: 12/21/2022] Open
Abstract
Recent evidence suggests that inhibition of protein phosphatase 2A (PP2A) tumor suppressor activity via the SET oncoprotein contributes to the pathogenesis of various cancers. Here we demonstrate that both SET and c-MYC expression are frequently elevated in T-ALL cell lines and primary samples compared to healthy T cells. Treatment of T-ALL cells with the SET antagonist OP449 restored the activity of PP2A and reduced SET interaction with the PP2A catalytic subunit, resulting in a decrease in cell viability and c-MYC expression in a dose-dependent manner. Since a tight balance between phosphatases and kinases is required for the growth of both normal and malignant cells, we sought to identify a kinase inhibitor that would synergize with SET antagonism. We tested various T-ALL cell lines against a small-molecule inhibitor screen of 66 compounds targeting two-thirds of the tyrosine kinome and found that combined treatment of T-ALL cells with dovitinib, an orally active multi-targeted small-molecule receptor tyrosine kinase inhibitor, and OP449 synergistically reduced the viability of all tested T-ALL cell lines. Mechanistically, combined treatment with OP449 and dovitinib decreased total and phospho c-MYC levels and reduced ERK1/2, AKT, and p70S6 kinase activity in both NOTCH-dependent and independent T-ALL cell lines. Overall, these results suggest that combined targeting of tyrosine kinases and activation of serine/threonine phosphatases may offer novel therapeutic strategies for the treatment of T-ALL.
Collapse
Affiliation(s)
- Nameeta P Richard
- Randall Children's Hospital at Legacy Emanuel, Children's Cancer and Blood Disorders Program, Portland, OR 97227, USA.,Division of Pediatric Hematology Oncology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Raffaella Pippa
- Division of Oncology, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
| | - Megan M Cleary
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR 97239, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Alka Puri
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR 97239, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Deanne Tibbitts
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Shawn Mahmood
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR 97239, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Dale J Christensen
- Research and Development, Oncotide Pharmaceuticals, Research Triangle Park, NC 27710, USA .,Spyryx Biosciences, Durham, NC 27713, USA
| | - Sophia Jeng
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health and Science University, Portland, OR 97239, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Shannon McWeeney
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health and Science University, Portland, OR 97239, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - A Thomas Look
- Dana-Farber Cancer Institute, Harvard Cancer Center, Boston, MA 02215, USA
| | - Bill H Chang
- Division of Pediatric Hematology Oncology, Oregon Health and Science University, Portland, OR 97239, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Jeffrey W Tyner
- Department of Cell and Developmental Biology, Oregon Health and Science University, Portland, OR 97239, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Michael P Vitek
- Research and Development, Oncotide Pharmaceuticals, Research Triangle Park, NC 27710, USA
| | - María D Odero
- Division of Oncology, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
| | - Rosalie Sears
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR USA-97239.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA-97239
| | - Anupriya Agarwal
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR, USA-97239.,Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR USA-97239.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA-97239
| |
Collapse
|
19
|
Protein Phosphatase 2A in the Regulation of Wnt Signaling, Stem Cells, and Cancer. Genes (Basel) 2018; 9:genes9030121. [PMID: 29495399 PMCID: PMC5867842 DOI: 10.3390/genes9030121] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/16/2018] [Accepted: 02/21/2018] [Indexed: 12/21/2022] Open
Abstract
Protein phosphorylation is a ubiquitous cellular process that allows for the nuanced and reversible regulation of protein activity. Protein phosphatase 2A (PP2A) is a heterotrimeric serine-threonine phosphatase—composed of a structural, regulatory, and catalytic subunit—that controls a variety of cellular events via protein dephosphorylation. While much is known about PP2A and its basic biochemistry, the diversity of its components—especially the multitude of regulatory subunits—has impeded the determination of PP2A function. As a consequence of this complexity, PP2A has been shown to both positively and negatively regulate signaling networks such as the Wnt pathway. Wnt signaling modulates major developmental processes, and is a dominant mediator of stem cell self-renewal, cell fate, and cancer stem cells. Because PP2A affects Wnt signaling both positively and negatively and at multiple levels, further understanding of this complex dynamic may ultimately provide insight into stem cell biology and how to better treat cancers that result from alterations in Wnt signaling. This review will summarize literature that implicates PP2A as a tumor suppressor, explore PP2A mutations identified in human malignancy, and focus on PP2A in the regulation of Wnt signaling and stem cells so as to better understand how aberrancy in this pathway can contribute to tumorigenesis.
Collapse
|
20
|
White C, Alshaker H, Cooper C, Winkler M, Pchejetski D. The emerging role of FTY720 (Fingolimod) in cancer treatment. Oncotarget 2018; 7:23106-27. [PMID: 27036015 PMCID: PMC5029614 DOI: 10.18632/oncotarget.7145] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 01/19/2016] [Indexed: 02/07/2023] Open
Abstract
FTY720 (Fingolimod) is a clinically approved immunomodulating therapy for multiple sclerosis that sequesters T-cells to lymph nodes through functional antagonism of sphingosine-1-phosphate 1 receptor. FTY720 also demonstrates a proven efficacy in multiple in vitro and in vivo cancer models, suggesting a potential therapeutic role in cancer patients. A potential anticancer mechanism of FTY720 is through the inhibition of sphingosine kinase 1, a proto-oncogene with in vitro and clinical cancer association. In addition, FTY720's anticancer properties may be attributable to actions on several other molecular targets. This study focuses on reviewing the emerging evidence regarding the anticancer properties and molecular targets of FTY720. While the clinical transition of FTY720 is currently limited by its immune suppression effects, studies aiming at FTY720 delivery and release together with identifying its key synergetic combinations and relevant patient subsets may lead to its rapid introduction into the clinic.
Collapse
Affiliation(s)
| | - Heba Alshaker
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan.,School of Medicine, University of East Anglia, Norwich, UK
| | - Colin Cooper
- School of Medicine, University of East Anglia, Norwich, UK
| | - Matthias Winkler
- Department of Surgery and Cancer, Imperial College London, London, UK
| | | |
Collapse
|
21
|
A systematic evaluation of the safety and toxicity of fingolimod for its potential use in the treatment of acute myeloid leukaemia. Anticancer Drugs 2017; 27:560-8. [PMID: 26967515 PMCID: PMC4881728 DOI: 10.1097/cad.0000000000000358] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Treatment of acute myeloid leukaemia (AML) is challenging and emerging treatment options include protein phosphatase 2A (PP2A) activators. Fingolimod is a known PP2A activator that inhibits multiple signalling pathways and has been used extensively in patients with multiple sclerosis and other indications. The initial positive results of PP2A activators in vitro and mouse models of AML are promising; however, its safety for use in AML has not been assessed. From human studies of fingolimod in other indications, it is possible to evaluate whether the safety and toxicity profile of the PP2A activators will allow their use in treating AML. A literature review was carried out to assess safety before the commencement of Phase I trials of the PP2A activator Fingolimod in AML. From human studies of fingolimod in other indications, it is possible to evaluate whether the safety and toxicity profile of the PP2A activators will allow their use in treating AML. A systematic review of published literature in Medline, EMBASE and the Cochrane Library of critical reviews was carried out. International standards for the design and reporting of search strategies were followed. Search terms and medical subject headings used in trials involving PP2A activators as well as a specific search were performed for ‘adverse events’, ‘serious adverse events’, ‘delays in treatment’, ‘ side effects’ and ‘toxicity’ for primary objectives. Database searches were limited to papers published in the last 12 years and available in English. The search yielded 677 articles. A total of 69 journal articles were identified as relevant and included 30 clinical trials, 24 review articles and 15 case reports. The most frequently reported adverse events were nausea, diarrhoea, fatigue, back pain, influenza viral infections, nasopharyngitis and bronchitis. Specific safety concerns include monitoring of the heart rate and conduction at commencement of treatment as cardiotoxicity has been reported. There is little evidence to suggest specific bone marrow toxicity. Lymophopenia is a desired effect in the management of multiple sclerosis, but may have implications in patients with acute leukaemia as it may potentially increase susceptibility to viral infections such as influenza. Fingolimod is a potential treatment option for AML with an acceptable risk to benefit ratio, given its lack of bone marrow toxicity and the relatively low rate of serious side effects. As most patients with AML are elderly, specific monitoring for cardiac toxicity as well as infection would be required.
Collapse
|
22
|
Balliu M, Cellai C, Lulli M, Laurenzana A, Torre E, Vannucchi AM, Paoletti F. HDAC1 controls CIP2A transcription in human colorectal cancer cells. Oncotarget 2017; 7:25862-71. [PMID: 27029072 PMCID: PMC5041950 DOI: 10.18632/oncotarget.8406] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/10/2016] [Indexed: 12/03/2022] Open
Abstract
This work describes the effectiveness of HDAC-inhibitor (S)-2 towards colorectal cancer (CRC) HCT116 cells in vitro by inducing cell cycle arrest and apoptosis, and in vivo by contrasting tumour growth in mice xenografts. Among the multifaceted drug-induced events described herein, an interesting link has emerged between the oncoprotein histone deacetylase HDAC1 and the oncogenic Cancerous Inhibitor of Protein Phosphatase 2A (CIP2A) which is overexpressed in several cancers including CRCs. HDAC1 inhibition by (S)-2 or specific siRNAs downregulates CIP2A transcription in three different CRC cell lines, thus restoring the oncosuppressor phosphatase PP2A activity that is reduced in most cancers. Once re-activated, PP2A dephosphorylates pGSK-3β(ser9) which phosphorylates β-catenin that remains within the cytosol where it undergoes degradation. The decreased amount/activity of β-catenin transcription factor prompts cell growth arrest by diminishing c-Myc and cyclin D1 expression and abrogating the prosurvival Wnt/β-catenin signaling pathway. These results are the first evidence that the inhibition of HDAC1 by (S)-2 downregulates CIP2A transcription and unleashes PP2A activity, thus inducing growth arrest and apoptosis in CRC cells.
Collapse
Affiliation(s)
- Manjola Balliu
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Firenze, Italy
| | - Cristina Cellai
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", 50134 Firenze, Italy
| | - Matteo Lulli
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", 50134 Firenze, Italy
| | - Anna Laurenzana
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", 50134 Firenze, Italy
| | - Eugenio Torre
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", 50134 Firenze, Italy
| | | | - Francesco Paoletti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", 50134 Firenze, Italy
| |
Collapse
|
23
|
Shlomai G, Zelenko Z, Antoniou IM, Stasinopoulos M, Tobin-Hess A, Vitek MP, LeRoith D, Gallagher EJ. OP449 inhibits breast cancer growth without adverse metabolic effects. Endocr Relat Cancer 2017; 24:519-529. [PMID: 28830934 PMCID: PMC5678946 DOI: 10.1530/erc-17-0077] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 07/21/2017] [Indexed: 12/23/2022]
Abstract
Hyperinsulinemia is associated with a decrease in breast cancer recurrence-free survival and overall survival. Inhibition of insulin receptor signaling is associated with glycemic dysregulation. SET is a direct modulator of PP2A, which negatively regulates the PI3K/AKT/mTOR pathway. OP449, a SET inhibitor, decreases AKT/mTOR activation. The effects of OP449 treatment on breast cancer growth in the setting of pre-diabetes, and its metabolic implications are currently unknown. We found that the volumes and weights of human MDA-MB-231 breast cancer xenografts were greater in hyperinsulinemic mice compared with controls (P < 0.05), and IR phosphorylation was 4.5-fold higher in these mice (P < 0.05). Human and murine breast cancer tumors treated with OP449 were 47% and 39% smaller than controls (P < 0.05, for both, respectively). AKT and S6RP phosphorylation were 82% and 34% lower in OP449-treated tumors compared with controls (P < 0.05, P = 0.06, respectively). AKT and S6RP phosphorylation in response to insulin was 30% and 12% lower in cells, pre-treated with OP449, compared with control cells (P < 0.01, P < 0.05, respectively). However, even with decreased AKT/mTOR activation, body weights and composition, blood glucose and plasma insulin, glucose tolerance, serum triglyceride and cholesterol levels were similar between OP449-treated mice and controls. Xenografts and liver tissue from OP449-treated mice showed a 64% and 70% reduction in STAT5 activation, compared with controls (P < 0.01 and P = 0.06, respectively). Our data support an anti-neoplastic effect of OP449 on human breast cancer cells in vitro and in xenografts in the setting of hyperinsulinemia. OP449 led to the inhibition of AKT/mTOR signaling, albeit, not leading to metabolic derangements.
Collapse
Affiliation(s)
- Gadi Shlomai
- Division of EndocrinologyDiabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- The Dr Pinchas Borenstein Talpiot Medical Leadership Program 2013Tel-Hashomer, Israel
| | - Zara Zelenko
- Division of EndocrinologyDiabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Irini Markella Antoniou
- Division of EndocrinologyDiabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Marilyn Stasinopoulos
- Division of EndocrinologyDiabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Aviva Tobin-Hess
- Division of EndocrinologyDiabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Michael P Vitek
- CognosciInc., Durham, North Carolina, USA
- Department of NeurologyDuke University Medical Center, Research Drive, Durham, North Carolina, USA
| | - Derek LeRoith
- Division of EndocrinologyDiabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Emily Jane Gallagher
- Division of EndocrinologyDiabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
24
|
Perrotti D, Silvestri G, Stramucci L, Yu J, Trotta R. Cellular and Molecular Networks in Chronic Myeloid Leukemia: The Leukemic Stem, Progenitor and Stromal Cell Interplay. Curr Drug Targets 2017; 18:377-388. [PMID: 27307150 DOI: 10.2174/1389450117666160615074120] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 06/01/2016] [Accepted: 06/02/2016] [Indexed: 12/13/2022]
Abstract
The use of imatinib, second and third generation ABL tyrosine kinase inhibitors (TKI) (i.e. dasatinib, nilotinib, bosutinib and ponatinib) made CML a clinically manageable and, in a small percentage of cases, a cured disease. TKI therapy also turned CML blastic transformation into a rare event; however, disease progression still occurs in those patients who are refractory, not compliant with TKI therapy or develop resistance to multiple TKIs. In the past few years, it became clear that the BCRABL1 oncogene does not operate alone to drive disease emergence, maintenance and progression. Indeed, it seems that bone marrow (BM) microenvironment-generated signals and cell autonomous BCRABL1 kinase-independent genetic and epigenetic alterations all contribute to: i. persistence of a quiescent leukemic stem cell (LSC) reservoir, ii. innate or acquired resistance to TKIs, and iii. progression into the fatal blast crisis stage. Herein, we review the intricate leukemic network in which aberrant, but finely tuned, survival, mitogenic and self-renewal signals are generated by leukemic progenitors, stromal cells, immune cells and metabolic microenvironmental conditions (e.g. hypoxia) to promote LSC maintenance and blastic transformation.
Collapse
Affiliation(s)
- Danilo Perrotti
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | | | | | | | | |
Collapse
|
25
|
Emerging Paradigms of G Protein-Coupled Receptor Dephosphorylation. Trends Pharmacol Sci 2017; 38:621-636. [DOI: 10.1016/j.tips.2017.04.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/31/2017] [Accepted: 04/06/2017] [Indexed: 12/21/2022]
|
26
|
Tibaldi E, Pagano MA, Frezzato F, Trimarco V, Facco M, Zagotto G, Ribaudo G, Pavan V, Bordin L, Visentin A, Zonta F, Semenzato G, Brunati AM, Trentin L. Targeted activation of the SHP-1/PP2A signaling axis elicits apoptosis of chronic lymphocytic leukemia cells. Haematologica 2017; 102:1401-1412. [PMID: 28619847 PMCID: PMC5541874 DOI: 10.3324/haematol.2016.155747] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 06/14/2017] [Indexed: 01/07/2023] Open
Abstract
Lyn, a member of the Src family of kinases, is a key factor in the dysregulation of survival and apoptotic pathways of malignant B cells in chronic lymphocytic leukemia. One of the effects of Lyn’s action is spatial and functional segregation of the tyrosine phosphatase SHP-1 into two pools, one beneath the plasma membrane in an active state promoting pro-survival signals, the other in the cytosol in an inhibited conformation and unable to counter the elevated level of cytosolic tyrosine phosphorylation. We herein show that SHP-1 activity can be elicited directly by nintedanib, an agent also known as a triple angiokinase inhibitor, circumventing the phospho-S591-dependent inhibition of the phosphatase, leading to the dephosphorylation of pro-apoptotic players such as procaspase-8 and serine/threonine phosphatase 2A, eventually triggering apoptosis. Furthermore, the activation of PP2A by using MP07-66, a novel FTY720 analog, stimulated SHP-1 activity via dephosphorylation of phospho-S591, which unveiled the existence of a positive feedback signaling loop involving the two phosphatases. In addition to providing further insights into the molecular basis of this disease, our findings indicate that the PP2A/SHP-1 axis may emerge as an attractive, novel target for the development of alternative strategies in the treatment of chronic lymphocytic leukemia.
Collapse
Affiliation(s)
- Elena Tibaldi
- Department of Molecular Medicine, University of Padua, Italy
| | - Mario Angelo Pagano
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| | - Federica Frezzato
- Department of Medicine, University of Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), Centro di Eccellenza per la Ricerca Biomedica, Padua, Italy
| | - Valentina Trimarco
- Department of Medicine, University of Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), Centro di Eccellenza per la Ricerca Biomedica, Padua, Italy
| | - Monica Facco
- Department of Medicine, University of Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), Centro di Eccellenza per la Ricerca Biomedica, Padua, Italy
| | - Giuseppe Zagotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| | - Giovanni Ribaudo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| | - Valeria Pavan
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| | - Luciana Bordin
- Department of Molecular Medicine, University of Padua, Italy
| | - Andrea Visentin
- Department of Medicine, University of Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), Centro di Eccellenza per la Ricerca Biomedica, Padua, Italy
| | - Francesca Zonta
- Department of Biomedical Sciences, University of Padua, Italy
| | - Gianpietro Semenzato
- Department of Medicine, University of Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), Centro di Eccellenza per la Ricerca Biomedica, Padua, Italy
| | | | - Livio Trentin
- Department of Medicine, University of Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), Centro di Eccellenza per la Ricerca Biomedica, Padua, Italy
| |
Collapse
|
27
|
Overexpression of phosphoprotein phosphatase 2A predicts worse prognosis in patients with breast cancer: a 15-year follow-up. Hum Pathol 2017; 66:93-100. [PMID: 28603063 DOI: 10.1016/j.humpath.2017.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/26/2017] [Accepted: 06/01/2017] [Indexed: 12/15/2022]
Abstract
Breast cancer subtypes can be stratified by IHC expression of estrogen receptor, progesterone receptor, and human epidermal growth factor 2 (HER2). The signaling pathways mediated by these receptors are the dominant drivers of cell proliferation and survival in most human breast cancers. One of the most frequently overactivated pathways in breast cancer is the AKT signaling cascade. Protein phosphatase 2A (PP2A) acts as a switch to turn off signal transduction in the AKT pathway; however, it is frequently inactivated in many cancers by phosphorylation of Tyr-307 to form phosphoprotein phosphatase 2A (p-PP2A). This study aimed to investigate the clinical significance of p-PP2A and phospho-AKT (p-AKT) expression in 672 patients with breast cancer during a 15-year follow-up. The breast tissue microarray was evaluated for p-PP2A and p-AKT expression using IHC staining and scores. Analysis of IHC staining results revealed that p-PP2A expression was positively correlated with HER2, Ki-67, and p-AKT overexpression (P<.001, P=.003, and P=.001, respectively). At the time of diagnosis, breast cancer patients with higher p-PP2A expression had significantly shorter 15-year OS than patients with lower p-PP2A expression did (P=.017). Multivariate Cox regression analysis revealed that high p-PP2A expression was an independent prognostic factor for shorter OS (hazard ratio, 1.741; P=.012). Our data revealed that high p-PP2A expression is positively associated with HER2, Ki-67, and p-AKT expression. High p-PP2A expression correlates with poor clinical outcomes in breast cancer, especially in patients with TNBC.
Collapse
|
28
|
Miller TR, Beversdorf LJ, Weirich CA, Bartlett SL. Cyanobacterial Toxins of the Laurentian Great Lakes, Their Toxicological Effects, and Numerical Limits in Drinking Water. Mar Drugs 2017; 15:E160. [PMID: 28574457 PMCID: PMC5484110 DOI: 10.3390/md15060160] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 04/22/2017] [Accepted: 05/02/2017] [Indexed: 02/07/2023] Open
Abstract
Cyanobacteria are ubiquitous phototrophic bacteria that inhabit diverse environments across the planet. Seasonally, they dominate many eutrophic lakes impacted by excess nitrogen (N) and phosphorus (P) forming dense accumulations of biomass known as cyanobacterial harmful algal blooms or cyanoHABs. Their dominance in eutrophic lakes is attributed to a variety of unique adaptations including N and P concentrating mechanisms, N₂ fixation, colony formation that inhibits predation, vertical movement via gas vesicles, and the production of toxic or otherwise bioactive molecules. While some of these molecules have been explored for their medicinal benefits, others are potent toxins harmful to humans, animals, and other wildlife known as cyanotoxins. In humans these cyanotoxins affect various tissues, including the liver, central and peripheral nervous system, kidneys, and reproductive organs among others. They induce acute effects at low doses in the parts-per-billion range and some are tumor promoters linked to chronic diseases such as liver and colorectal cancer. The occurrence of cyanoHABs and cyanotoxins in lakes presents challenges for maintaining safe recreational aquatic environments and the production of potable drinking water. CyanoHABs are a growing problem in the North American (Laurentian) Great Lakes basin. This review summarizes information on the occurrence of cyanoHABs in the Great Lakes, toxicological effects of cyanotoxins, and appropriate numerical limits on cyanotoxins in finished drinking water.
Collapse
Affiliation(s)
- Todd R Miller
- Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA.
| | - Lucas J Beversdorf
- Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA.
| | - Chelsea A Weirich
- Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA.
| | - Sarah L Bartlett
- Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA.
| |
Collapse
|
29
|
Ward MP, Spiers JP. Protein phosphatase 2A regulation of markers of extracellular matrix remodelling in hepatocellular carcinoma cells: functional consequences for tumour invasion. Br J Pharmacol 2017; 174:1116-1130. [PMID: 28239848 DOI: 10.1111/bph.13759] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 02/13/2017] [Accepted: 02/16/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND AND PURPOSE A hallmark of tumour invasion is breakdown of the extracellular matrix due to dysregulation of the matrix metalloproteinase (MMP) system. While our understanding of how this is regulated by kinase signalling pathways is well established, its counter-regulation by protein phosphatases (PP) is poorly understood. Therefore, we investigated the effect of PP inhibition on markers of extracellular remodelling and how PP2A activity modulated MMP-9 abundance and function of Hep3B cells. EXPERIMENTAL APPROACH Cells were exposed to okadaic acid (OA), tautomycetin and cyclosporin A, and the expression profile determined using PCR. Effects of OA and a protein inhibitor of PP2A, CIP2A, on MMP-9 abundance, PP2A activity and cell migration were investigated using ELISA, promoter constructs, siRNA knockdown and transwell migration assays. KEY RESULTS OA increased expression and abundance of MMP-9 and the tissue inhibitor of MMP, TIMP-1, without affecting other MMPs, TIMPs and ADAMs. The effect on MMP-9 was mimicked by CIP2A overexpression and knockdown of the PPP2CA catalytic, but not PPP2R1A scaffolding, subunit. Cyclosporin A and PPP1CA silencing did not alter MMP-9 expression, while tautomycetin transiently increased it. Mutation of AP-1, but not NF-κB, binding sites inhibited OA-mediated MMP-9 transcriptional activity. OA and CIP2A decreased PP2A activity and increased cell migration. CONCLUSION AND IMPLICATIONS OA increased MMP-9 by decreasing PP2A activity and PP2Ac, through AP-1 binding sites on the MMP-9 promoter. The functional consequence of this and CIP2A overexpression was increased cell migration. Hence, PP2A inhibition induced a metastatic phenotype through alterations in MMP-9 in Hep3B cells.
Collapse
Affiliation(s)
- M P Ward
- Department of Pharmacology and Therapeutics, Trinity College Dublin, Dublin, Ireland
| | - J P Spiers
- Department of Pharmacology and Therapeutics, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
30
|
Tzima E, Serifi I, Tsikari I, Alzualde A, Leonardos I, Papamarcaki T. Transcriptional and Behavioral Responses of Zebrafish Larvae to Microcystin-LR Exposure. Int J Mol Sci 2017; 18:ijms18020365. [PMID: 28208772 PMCID: PMC5343900 DOI: 10.3390/ijms18020365] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 01/29/2017] [Accepted: 02/02/2017] [Indexed: 12/29/2022] Open
Abstract
Microcystins are cyclic heptapeptides that constitute a diverse group of toxins produced by cyanobacteria. One of the most toxic variants of this family is microcystin-LR (MCLR) which is a potent inhibitor of protein phosphatase 2A (PP2A) and induces cytoskeleton alterations. In this study, zebrafish larvae exposed to 500 μg/L of MCLR for four days exhibited a 40% reduction of PP2A activity compared to the controls, indicating early effects of the toxin. Gene expression profiling of the MCLR-exposed larvae using microarray analysis revealed that keratin 96 (krt96) was the most downregulated gene, consistent with the well-documented effects of MCLR on cytoskeleton structure. In addition, our analysis revealed upregulation in all genes encoding for the enzymes of the retinal visual cycle, including rpe65a (retinal pigment epithelium-specific protein 65a), which is critical for the larval vision. Quantitative real-time PCR (qPCR) analysis confirmed the microarray data, showing that rpe65a was significantly upregulated at 50 μg/L and 500 μg/L MCLR in a dose-dependent manner. Consistent with the microarray data, MCLR-treated larvae displayed behavioral alterations such as weakening response to the sudden darkness and hypoactivity in the dark. Our work reveals new molecular targets for MCLR and provides further insights into the molecular mechanisms of MCLR toxicity during early development.
Collapse
Affiliation(s)
- Eleni Tzima
- Laboratory of Biological Chemistry, Medical School, University of Ioannina, 45110 Ioannina, Greece.
- Division of Biomedical Research, Foundation for Research and Technology-Hellas, Institute of Molecular Biology and Biotechnology, 45110 Ιοannina, Greece.
| | - Iliana Serifi
- Laboratory of Biological Chemistry, Medical School, University of Ioannina, 45110 Ioannina, Greece.
- Division of Biomedical Research, Foundation for Research and Technology-Hellas, Institute of Molecular Biology and Biotechnology, 45110 Ιοannina, Greece.
| | - Ioanna Tsikari
- Laboratory of Biological Chemistry, Medical School, University of Ioannina, 45110 Ioannina, Greece.
| | | | - Ioannis Leonardos
- Laboratory of Zoology, Department of Biological Applications and Technologies, University of Ioannina, 45110 Ioannina, Greece.
| | - Thomais Papamarcaki
- Laboratory of Biological Chemistry, Medical School, University of Ioannina, 45110 Ioannina, Greece.
- Division of Biomedical Research, Foundation for Research and Technology-Hellas, Institute of Molecular Biology and Biotechnology, 45110 Ιοannina, Greece.
| |
Collapse
|
31
|
Trost BM, Knopf JD, Brindle CS. Synthetic Strategies Employed for the Construction of Fostriecin and Related Natural Products. Chem Rev 2016; 116:15035-15088. [PMID: 28027648 PMCID: PMC5720176 DOI: 10.1021/acs.chemrev.6b00488] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Fostriecin and related natural products present a significant challenge for synthetic chemists due to their structural complexity and chemical sensitivity. This review will chronicle the successful efforts of synthetic chemists in the construction of these biologically active molecules. Key carbon-carbon bond forming reactions will be highlighted, as well as the methods used to install the numerous stereocenters present in this class of compounds.
Collapse
Affiliation(s)
- Barry M. Trost
- Department of Chemistry, Stanford University, Stanford, California 94305-5080, United States
| | - Joshua D. Knopf
- Department of Chemistry, Trinity College, 300 Summit Street, Hartford, Connecticut 06106, United States
| | - Cheyenne S. Brindle
- Department of Chemistry, Trinity College, 300 Summit Street, Hartford, Connecticut 06106, United States
| |
Collapse
|
32
|
Enoru JO, Yang B, Krishnamachari S, Villanueva E, DeMaio W, Watanyar A, Chinnasamy R, Arterburn JB, Perez RG. Preclinical Metabolism, Pharmacokinetics and In Vivo Analysis of New Blood-Brain-Barrier Penetrant Fingolimod Analogues: FTY720-C2 and FTY720-Mitoxy. PLoS One 2016; 11:e0162162. [PMID: 27611691 PMCID: PMC5017749 DOI: 10.1371/journal.pone.0162162] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 08/18/2016] [Indexed: 12/19/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative aging disorder in which postmortem PD brain exhibits neuroinflammation, as well as synucleinopathy-associated protein phosphatase 2A (PP2A) enzymatic activity loss. Based on our translational research, we began evaluating the PD-repurposing-potential of an anti-inflammatory, neuroprotective, and PP2A stimulatory oral drug that is FDA-approved for multiple sclerosis, FTY720 (fingolimod, Gilenya®). We also designed two new FTY720 analogues, FTY720-C2 and FTY720-Mitoxy, with modifications that affect drug potency and mitochondrial localization, respectively. Herein, we describe the metabolic stability and metabolic profiling of FTY720-C2 and FTY720-Mitoxy in liver microsomes and hepatocytes. Using mouse, rat, dog, monkey, and human liver microsomes the intrinsic clearance of FTY720-C2 was 22.5, 79.5, 6.0, 20.2 and 18.3 μL/min/mg; and for FTY720-Mitoxy was 1.8, 7.8, 1.4, 135.0 and 17.5 μL/min/mg, respectively. In hepatocytes, both FTY720-C2 and FTY720-Mitoxy were metabolized from the octyl side chain, generating a series of carboxylic acids similar to the parent FTY720, but without phosphorylated metabolites. To assess absorption and distribution, we gave equivalent single intravenous (IV) or oral doses of FTY720-C2 or FTY720-Mitoxy to C57BL/6 mice, with two mice per time point evaluated. After IV delivery, both FTY720-C2 and FTY720-Mitoxy were rapidly detected in plasma and brain; and reached peak concentrations at the first sampling time points. After oral dosing, FTY720-C2 was present in plasma and brain, although FTY720-Mitoxy was not orally bioavailable. Brain-to-plasma ratio of both compounds increased time-dependently, suggesting a preferential partitioning to the brain. PP2A activity in mouse adrenal gland increased ~2-fold after FTY720-C2 or FTY720-Mitoxy, as compared to untreated controls. In summary, FTY720-C2 and FTY720-Mitoxy both (i) crossed the blood-brain-barrier; (ii) produced metabolites similar to FTY720, except without phosphorylated species that cause S1P1-mediated-immunosuppression; and (iii) stimulated in vivo PP2A activity, all of which encourage additional preclinical assessment.
Collapse
Affiliation(s)
- Julius O. Enoru
- In Vitro and Molecular Metabolism Laboratory, Ricerca Biosciences LLC, Concord, Ohio, United States of America
| | - Barbara Yang
- Department of Biomedical Sciences, Graduate School of Biomedical Sciences, Center of Emphasis in Neurosciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, United States of America
| | - Sesha Krishnamachari
- Department of Biomedical Sciences, Graduate School of Biomedical Sciences, Center of Emphasis in Neurosciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, United States of America
| | - Ernesto Villanueva
- Department of Biomedical Sciences, Graduate School of Biomedical Sciences, Center of Emphasis in Neurosciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, United States of America
| | - William DeMaio
- In Vitro and Molecular Metabolism Laboratory, Ricerca Biosciences LLC, Concord, Ohio, United States of America
| | - Adiba Watanyar
- In Vitro and Molecular Metabolism Laboratory, Ricerca Biosciences LLC, Concord, Ohio, United States of America
| | - Ramesh Chinnasamy
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico, United States of America
| | - Jeffrey B. Arterburn
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico, United States of America
| | - Ruth G. Perez
- Department of Biomedical Sciences, Graduate School of Biomedical Sciences, Center of Emphasis in Neurosciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, United States of America
- * E-mail:
| |
Collapse
|
33
|
The broken "Off" switch in cancer signaling: PP2A as a regulator of tumorigenesis, drug resistance, and immune surveillance. BBA CLINICAL 2016; 6:87-99. [PMID: 27556014 PMCID: PMC4986044 DOI: 10.1016/j.bbacli.2016.08.002] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 08/01/2016] [Accepted: 08/02/2016] [Indexed: 12/31/2022]
Abstract
Aberrant activation of signal transduction pathways can transform a normal cell to a malignant one and can impart survival properties that render cancer cells resistant to therapy. A diverse set of cascades have been implicated in various cancers including those mediated by serine/threonine kinases such RAS, PI3K/AKT, and PKC. Signal transduction is a dynamic process involving both "On" and "Off" switches. Activating mutations of RAS or PI3K can be viewed as the switch being stuck in the "On" position resulting in continued signaling by a survival and/or proliferation pathway. On the other hand, inactivation of protein phosphatases such as the PP2A family can be seen as the defective "Off" switch that similarly can activate these pathways. A problem for therapeutic targeting of PP2A is that the enzyme is a hetero-trimer and thus drug targeting involves complex structures. More importantly, since PP2A isoforms generally act as tumor suppressors one would want to activate these enzymes rather than suppress them. The elucidation of the role of cellular inhibitors like SET and CIP2A in cancer suggests that targeting these proteins can have therapeutic efficacy by mechanisms involving PP2A activation. Furthermore, drugs such as FTY-720 can activate PP2A isoforms directly. This review will cover the current state of knowledge of PP2A role as a tumor suppressor in cancer cells and as a mediator of processes that can impact drug resistance and immune surveillance.
Collapse
|
34
|
Grech G, Baldacchino S, Saliba C, Grixti MP, Gauci R, Petroni V, Fenech AG, Scerri C. Deregulation of the protein phosphatase 2A, PP2A in cancer: complexity and therapeutic options. Tumour Biol 2016; 37:11691-11700. [PMID: 27444275 DOI: 10.1007/s13277-016-5145-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 07/11/2016] [Indexed: 01/26/2023] Open
Abstract
The complexity of the phosphatase, PP2A, is being unravelled and current research is increasingly providing information on the association of deregulated PP2A function with cancer initiation and progression. It has been reported that decreased activity of PP2A is a recurrent observation in many types of cancer, including colorectal and breast cancer (Baldacchino et al. EPMA J. 5:3, 2014; Cristobal et al. Mol Cancer Ther. 13:938-947, 2014). Since deregulation of PP2A and its regulatory subunits is a common event in cancer, PP2A is a potential target for therapy (Baldacchino et al. EPMA J. 5:3, 2014). In this review, the structural components of the PP2A complex are described, giving an in depth overview of the diversity of regulatory subunits. Regulation of the active PP2A trimeric complex, through phosphorylation and methylation, can be targeted using known compounds, to reactivate the complex. The endogenous inhibitors of the PP2A complex are highly deregulated in cancer, representing cases that are eligible to PP2A-activating drugs. Pharmacological opportunities to target low PP2A activity are available and preclinical data support the efficacy of these drugs, but clinical trials are lacking. We highlight the importance of PP2A deregulation in cancer and the current trends in targeting the phosphatase.
Collapse
Affiliation(s)
- Godfrey Grech
- Department of Pathology, Faculty of Medicine & Surgery, Medical School, University of Malta, Msida, MSD2090, Malta.
| | - Shawn Baldacchino
- Department of Pathology, Faculty of Medicine & Surgery, Medical School, University of Malta, Msida, MSD2090, Malta
| | - Christian Saliba
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Maria Pia Grixti
- Department of Pathology, Faculty of Medicine & Surgery, Medical School, University of Malta, Msida, MSD2090, Malta
| | - Robert Gauci
- Department of Pathology, Faculty of Medicine & Surgery, Medical School, University of Malta, Msida, MSD2090, Malta
| | - Vanessa Petroni
- Department of Anatomy, Faculty of Medicine & Surgery, University of Malta, Msida, Malta
| | - Anthony G Fenech
- Department of Clinical Pharmacology & Therapeutics, Faculty of Medicine & Surgery, University of Malta, Msida, Malta
| | - Christian Scerri
- Department of Physiology and Biochemistry, Faculty of Medicine & Surgery, University of Malta, Msida, Malta.,Molecular Genetics Clinic, Mater Dei Hospital, Msida, Malta
| |
Collapse
|
35
|
Rincón R, Cristóbal I, Zazo S, Arpí O, Menéndez S, Manso R, Lluch A, Eroles P, Rovira A, Albanell J, García-Foncillas J, Madoz-Gúrpide J, Rojo F. PP2A inhibition determines poor outcome and doxorubicin resistance in early breast cancer and its activation shows promising therapeutic effects. Oncotarget 2015; 6:4299-314. [PMID: 25726524 PMCID: PMC4414191 DOI: 10.18632/oncotarget.3012] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 12/31/2014] [Indexed: 11/26/2022] Open
Abstract
The protein phosphatase 2A (PP2A) is a key tumor suppressor which has emerged as a novel molecular target in some human cancers. Here, we show that PP2A inhibition is a common event in breast cancer and identified PP2A phosphorylation and deregulation SET and CIP2A as molecular contributing mechanisms to inactivate PP2A. Interestingly, restoration of PP2A activity after FTY720 treatment reduced cell growth, induced apoptosis and decreased AKT and ERK activation. Moreover, FTY720 led to PP2A activation then enhancing doxorubicin-induced antitumor effects both in vitro and in vivo. PP2A inhibition (CPscore: PP2A phosphorylation and/or CIP2A overexpression) was detected in 27% of cases (62/230), and associated with grade (p = 0.017), relapse (p < 0.001), negative estrogen (p < 0.001) and progesterone receptor expression (p < 0.001), HER2-positive tumors (p = 0.049), Ki-67 expression (p < 0.001), and higher AKT (p < 0.001) and ERK (p < 0.001) phosphorylation. Moreover, PP2A inhibition determined shorter overall (p = 0.006) and event-free survival (p = 0.003), and multivariate analysis confirmed its independent prognostic impact. Altogether, our results indicate that PP2A is frequently inactivated in breast cancer and determines worse outcome, and its restoration using PP2A activators represents an alternative therapeutic strategy in this disease.
Collapse
Affiliation(s)
- Raúl Rincón
- Translational Oncology Division, Oncohealth Institute, Health Research Institute FJD-UAM, University Hospital "Fundación Jiménez Diaz", Madrid, Spain
| | - Ion Cristóbal
- Translational Oncology Division, Oncohealth Institute, Health Research Institute FJD-UAM, University Hospital "Fundación Jiménez Diaz", Madrid, Spain
| | - Sandra Zazo
- Pathology Department, IIS "Fundación Jiménez Diaz", Madrid, Spain
| | - Oriol Arpí
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Silvia Menéndez
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Rebeca Manso
- Pathology Department, IIS "Fundación Jiménez Diaz", Madrid, Spain
| | - Ana Lluch
- Institute of Health Research INCLIVA, Valencia, Spain
| | - Pilar Eroles
- Institute of Health Research INCLIVA, Valencia, Spain
| | - Ana Rovira
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Joan Albanell
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Jesús García-Foncillas
- Translational Oncology Division, Oncohealth Institute, Health Research Institute FJD-UAM, University Hospital "Fundación Jiménez Diaz", Madrid, Spain
| | | | - Federico Rojo
- Pathology Department, IIS "Fundación Jiménez Diaz", Madrid, Spain.,Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| |
Collapse
|
36
|
Abstract
Chronic inflammatory diseases, such as asthma and chronic obstructive pulmonary disease (COPD), are clinically and socioeconomically important diseases globally. Currently the mainstay of anti-inflammatory therapy in respiratory diseases is corticosteroids. Although corticosteroids have proven clinical efficacy in asthma, many asthmatic inflammatory conditions (e.g., infection, exacerbation, and severe asthma) are not responsive to corticosteroids. Moreover, despite an understanding that COPD progression is driven by inflammation, we currently do not have effective anti-inflammatory strategies to combat this disease. Hence, alternative anti-inflammatory strategies are required. p38 mitogen-activated protein kinase (MAPK) has emerged as an important signaling molecule driving airway inflammation, and pharmacological inhibitors against p38 MAPK may provide potential therapies for chronic respiratory disease. In this review, we discuss some of the recent in vitro and in vivo studies targeting p38 MAPK, but suggest that p38 MAPK inhibitors may prove less effective than originally considered because they may block anti-inflammatory molecules along with proinflammatory responses. We propose that an alternative strategy may be to target an anti-inflammatory molecule farther downstream of p38 MAPK, i.e., tristetraprolin (TTP). TTP is an mRNA-destabilizing, RNA-binding protein that enhances the decay of mRNAs, including those encoding proteins implicated in chronic respiratory diseases. We suggest that understanding the molecular mechanism of TTP expression and its temporal regulation will guide future development of novel anti-inflammatory pharmacotherapeutic approaches to combat respiratory disease.
Collapse
Affiliation(s)
- Pavan Prabhala
- Faculty of Pharmacy, University of Sydney, Sydney, New South Wales, Australia
| | - Alaina J Ammit
- Faculty of Pharmacy, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
37
|
Ruvolo PP. The Interplay between PP2A and microRNAs in Leukemia. Front Oncol 2015; 5:43. [PMID: 25750899 PMCID: PMC4335100 DOI: 10.3389/fonc.2015.00043] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 02/05/2015] [Indexed: 12/19/2022] Open
Abstract
Protein phosphatase 2A (PP2A) is a serine/threonine phosphatase family whose members have been implicated in tumor suppression in many cancer models. In many cancers, loss of PP2A activity has been associated with tumorigenesis and drug resistance. Loss of PP2A results in failure to turn off survival signaling cascades that drive drug resistance such as those regulated by protein kinase B. PP2A is responsible for modulating function and controlling expression of tumor suppressors such as p53 and oncogenes such as BCL2 and MYC. Thus, PP2A has diverse functions regulating cell survival. The importance of microRNAs (miRs) is emerging in cancer biology. A role for miR regulation of PP2A is not well understood; however, recent studies suggest a number of clinically significant miRs such as miR-155 and miR-19 may include PP2A targets. We have recently found that a PP2A B subunit (B55α) can regulate a number of miRs in acute myeloid leukemia cells. The identification of a miR/PP2A axis represents a novel regulatory pathway in cellular homeostasis. The ability of miRs to suppress specific PP2A targets and for PP2A to control such miRs can add an extra level of control in signaling that could be used as a rheostat for many signaling cascades that maintain cellular homeostasis. As such, loss of PP2A or expression of miRs relevant for PP2A function could promote tumorigenesis or at least result in drug resistance. In this review, we will cover the current state of miR regulation of PP2A with a focus on leukemia. We will also briefly discuss what is known of PP2A regulation of miR expression.
Collapse
Affiliation(s)
- Peter P Ruvolo
- Department of Leukemia, University of Texas MD Anderson Cancer Center , Houston, TX , USA
| |
Collapse
|
38
|
Ciccone M, Calin GA, Perrotti D. From the Biology of PP2A to the PADs for Therapy of Hematologic Malignancies. Front Oncol 2015; 5:21. [PMID: 25763353 PMCID: PMC4329809 DOI: 10.3389/fonc.2015.00021] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 01/16/2015] [Indexed: 11/13/2022] Open
Abstract
Over the past decades, an emerging role of phosphatases in the pathogenesis of hematologic malignancies and solid tumors has been established. The tumor-suppressor protein phosphatase 2A (PP2A) belongs to the serine-threonine phosphatases family and accounts for the majority of serine-threonine phosphatase activity in eukaryotic cells. Numerous studies have shown that inhibition of PP2A expression and/or function may contribute to leukemogenesis in several hematological malignancies. Likewise, overexpression or aberrant expression of physiologic PP2A inhibitory molecules (e.g., SET and its associated SETBP1 and CIP2A) may turn off PP2A function and participate to leukemic progression. The discovery of PP2A as tumor suppressor has prompted the evaluation of the safety and the efficacy of new compounds, which can restore PP2A activity in leukemic cells. Although further studies are needed to better understand how PP2A acts in the intricate phosphatases/kinases cancer network, the results reviewed herein strongly support the development on new PP2A-activating drugs and the immediate introduction of those available into clinical protocols for leukemia patients refractory or resistant to current available therapies.
Collapse
Affiliation(s)
- Maria Ciccone
- Department of Experimental Therapeutics, MD Anderson Cancer Center, The University of Texas , Houston, TX , USA
| | - George A Calin
- Department of Experimental Therapeutics, MD Anderson Cancer Center, The University of Texas , Houston, TX , USA
| | - Danilo Perrotti
- Department of Medicine, The Greenebaum Cancer Center, University of Maryland School of Medicine , Baltimore, MD , USA
| |
Collapse
|
39
|
Haesen D, Sents W, Lemaire K, Hoorne Y, Janssens V. The Basic Biology of PP2A in Hematologic Cells and Malignancies. Front Oncol 2014; 4:347. [PMID: 25566494 PMCID: PMC4263090 DOI: 10.3389/fonc.2014.00347] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 11/20/2014] [Indexed: 12/30/2022] Open
Abstract
Reversible protein phosphorylation plays a crucial role in regulating cell signaling. In normal cells, phosphoregulation is tightly controlled by a network of protein kinases counterbalanced by several protein phosphatases. Deregulation of this delicate balance is widely recognized as a central mechanism by which cells escape external and internal self-limiting signals, eventually resulting in malignant transformation. A large fraction of hematologic malignancies is characterized by constitutive or unrestrained activation of oncogenic kinases. This is in part achieved by activating mutations, chromosomal rearrangements, or constitutive activation of upstream kinase regulators, in part by inactivation of their anti-oncogenic phosphatase counterparts. Protein phosphatase 2A (PP2A) represents a large family of cellular serine/threonine phosphatases with suspected tumor suppressive functions. In this review, we highlight our current knowledge about the complex structure and biology of these phosphatases in hematologic cells, thereby providing the rationale behind their diverse signaling functions. Eventually, this basic knowledge is a key to truly understand the tumor suppressive role of PP2A in leukemogenesis and to allow further rational development of therapeutic strategies targeting PP2A.
Collapse
Affiliation(s)
- Dorien Haesen
- Laboratory of Protein Phosphorylation and Proteomics, Department Cellular and Molecular Medicine, University of Leuven , Leuven , Belgium
| | - Ward Sents
- Laboratory of Protein Phosphorylation and Proteomics, Department Cellular and Molecular Medicine, University of Leuven , Leuven , Belgium
| | - Katleen Lemaire
- Gene Expression Unit, Department Cellular and Molecular Medicine, University of Leuven , Leuven , Belgium
| | - Yana Hoorne
- Laboratory of Protein Phosphorylation and Proteomics, Department Cellular and Molecular Medicine, University of Leuven , Leuven , Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation and Proteomics, Department Cellular and Molecular Medicine, University of Leuven , Leuven , Belgium
| |
Collapse
|
40
|
Wearne TA, Mirzaei M, Franklin JL, Goodchild AK, Haynes PA, Cornish JL. Methamphetamine-induced sensitization is associated with alterations to the proteome of the prefrontal cortex: implications for the maintenance of psychotic disorders. J Proteome Res 2014; 14:397-410. [PMID: 25245100 DOI: 10.1021/pr500719f] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Repeat administration of psychostimulants, such as methamphetamine, produces a progressive increase in locomotor activity (behavioral sensitization) in rodents that is believed to represent the underlying neurochemical changes driving psychoses. Alterations to the prefrontal cortex (PFC) are suggested to mediate the etiology and maintenance of these behavioral changes. As such, the aim of the current study was to investigate changes to protein expression in the PFC in male rats sensitized to methamphetamine using quantitative label-free shotgun proteomics. A methamphetamine challenge resulted in a significant sensitized locomotor response in methamphetamine pretreated animals compared to saline controls. Proteomic analysis revealed 96 proteins that were differentially expressed in the PFC of methamphetamine treated rats, with 20% of these being previously implicated in the neurobiology of schizophrenia in the PFC. We identified multiple biological functions in the PFC that appear to be commonly altered across methamphetamine-induced sensitization and schizophrenia, and these include synaptic regulation, protein phosphatase signaling, mitochondrial function, and alterations to the inhibitory GABAergic network. These changes could inform how alterations to the PFC could underlie the cognitive and behavioral dysfunction commonly seen across psychoses and places such biological changes as potential mediators in the maintenance of psychosis vulnerability.
Collapse
Affiliation(s)
- Travis A Wearne
- Department of Psychology, ‡Department of Chemistry and Biomolecular Sciences, §Australian School of Advanced Medicine, Macquarie University , Sydney, New South Wales 2109, Australia
| | | | | | | | | | | |
Collapse
|
41
|
Vargas-Medrano J, Krishnamachari S, Villanueva E, Godfrey WH, Lou H, Chinnasamy R, Arterburn JB, Perez RG. Novel FTY720-Based Compounds Stimulate Neurotrophin Expression and Phosphatase Activity in Dopaminergic Cells. ACS Med Chem Lett 2014; 5:782-6. [PMID: 25050165 DOI: 10.1021/ml500128g] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 05/27/2014] [Indexed: 12/15/2022] Open
Abstract
α-Synuclein is a chaperone-like protein implicated in Parkinson's disease (PD). Among α-synuclein's normal functions is an ability to bind to and stimulate the activity of the protein phosphatase 2A (PP2A) catalytic subunit in vitro and in vivo. PP2A activity is impaired in PD and in dementia with Lewy Bodies in brain regions harboring α-synuclein aggregates. Using PP2A as the readout, we measured PP2A activity in response to α-synuclein, ceramides, and FTY720, and then on the basis of those results, we created new FTY720 compounds. We then measured the effects of those compounds in dopaminergic cells. In addition to stimulating PP2A, all three compounds stimulated the expression of brain derived neurotrophic factor and protected MN9D cells against tumor-necrosis-factor-α-associated cell death. FTY720-C2 appears to be more potent while FTY720-Mitoxy targets mitochondria. Importantly, FTY720 is already FDA approved for treating multiple sclerosis and is used clinically worldwide. Our findings suggest that FTY720 and our new FTY720-based compounds have considerable potential for treating synucleinopathies such as PD.
Collapse
Affiliation(s)
- Javier Vargas-Medrano
- Department
of Biomedical Sciences, Center of Excellence in Neurosciences, Texas Tech University Health Sciences Center at El Paso, Paul L. Foster School of Medicine, El Paso, Texas 79905, United States
| | - Sesha Krishnamachari
- Department
of Biomedical Sciences, Center of Excellence in Neurosciences, Texas Tech University Health Sciences Center at El Paso, Paul L. Foster School of Medicine, El Paso, Texas 79905, United States
| | - Ernesto Villanueva
- Department
of Biomedical Sciences, Center of Excellence in Neurosciences, Texas Tech University Health Sciences Center at El Paso, Paul L. Foster School of Medicine, El Paso, Texas 79905, United States
| | - Wesley H. Godfrey
- Department
of Biomedical Sciences, Center of Excellence in Neurosciences, Texas Tech University Health Sciences Center at El Paso, Paul L. Foster School of Medicine, El Paso, Texas 79905, United States
| | - Haiyan Lou
- Department
of Pharmacology, Shandong University School of Medicine, Jinan, Shandong 250012, P. R. China
| | - Ramesh Chinnasamy
- Department
of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico 88003, United States
| | - Jeffrey B. Arterburn
- Department
of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico 88003, United States
| | - Ruth G. Perez
- Department
of Biomedical Sciences, Center of Excellence in Neurosciences, Texas Tech University Health Sciences Center at El Paso, Paul L. Foster School of Medicine, El Paso, Texas 79905, United States
| |
Collapse
|
42
|
Phosphorylated protein phosphatase 2A determines poor outcome in patients with metastatic colorectal cancer. Br J Cancer 2014; 111:756-62. [PMID: 25003662 PMCID: PMC4134505 DOI: 10.1038/bjc.2014.376] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 05/20/2014] [Accepted: 06/13/2014] [Indexed: 01/26/2023] Open
Abstract
Background: Protein phosphatase 2A (PP2A) is a tumour suppressor frequently inactivated in human cancer and its tyrosine-307 phosphorylation has been reported as a molecular inhibitory mechanism. Methods: Expression of phosphorylated PP2A (p-PP2A) was evaluated in 250 metastatic colorectal cancer (CRC) patients. Chi-square, Kaplan–Meier and Cox analyses were used to determine correlations with clinical and molecular parameters and impact on clinical outcomes. Results: High p-PP2A levels were found in 17.2% cases and were associated with ECOG performance status (P=0.001) and presence of synchronous metastasis at diagnosis (P=0.035). This subgroup showed substantially worse overall survival (OS) (median OS, 6.0 vs 26.2 months, P<0.001) and progression-free survival (PFS) (median PFS, 3.8 vs 13.3 months, P<0.001). The prognostic impact of p-PP2A was particularly evident in patients aged <70 years (P<0.001). Multivariate analysis revealed that p-PP2A retained its prognostic impact for OS (hazard ratio 2.7; 95% confidence interval, 1.8–4.1; P<0.001) and PFS (hazard ratio 3.0; 95% confidence interval, 1.8–5.0; P<0.001). Conclusions: Phosphorylated PP2A is an alteration that determines poor outcome in metastatic CRC and represents a novel potential therapeutic target in this disease, thus enabling to define a subgroup of patients who could benefit from future treatments based on PP2A activators.
Collapse
|
43
|
Cristóbal I, Rincón R, Manso R, Madoz-Gúrpide J, Caramés C, del Puerto-Nevado L, Rojo F, García-Foncillas J. Hyperphosphorylation of PP2A in colorectal cancer and the potential therapeutic value showed by its forskolin-induced dephosphorylation and activation. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1823-9. [PMID: 24997451 DOI: 10.1016/j.bbadis.2014.06.032] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 06/25/2014] [Accepted: 06/26/2014] [Indexed: 12/25/2022]
Abstract
BACKGROUND The tumor suppressor protein phosphatase 2A (PP2A) is frequently inactivated in human cancer and phosphorylation of its catalytic subunit (p-PP2A-C) at tyrosine-307 (Y307) has been described to inhibit this phosphatase. However, its molecular and clinical relevance in colorectal cancer (CRC) remains unclear. METHODS p-PP2A-C Y307 was determined by immunoblotting in 7 CRC cell lines and 35 CRC patients. CRC cells were treated with the PP2A activator forskolin alone or combined with the PP2A inhibitor okadaic acid, 5-fluorouracil and oxaliplatin. We examined cell growth, colonosphere formation, caspase activity and AKT and ERK activation. RESULTS PP2A-C was found hyperphosphorylated in CRC cell lines. Forskolin dephosphorylated and activated PP2A, impairing proliferation and colonosphere formation, and inducing activation of caspase 3/7 and changes in AKT and ERK phosphorylation. Moreover, forskolin showed additive effects with 5-fluorouracil and oxaliplatin treatments. Analysis of p-PP2A-C Y307 in primary tumors confirmed the presence of this alteration in a subgroup of CRC patients. CONCLUSIONS Our data show that PP2A-C hyperphosphorylation is a frequent event that contributes to PP2A inhibition in CRC. Antitumoral effects of forskolin-mediated PP2A activation suggest that the analysis of p-PP2A-C Y307 status could be used to identify a subgroup of patients who would benefit from treatments based on PP2A activators.
Collapse
Affiliation(s)
- Ion Cristóbal
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, University Hospital "Fundacion Jimenez Diaz", Autonomous University of Madrid, E-28040 Madrid, Spain.
| | - Raúl Rincón
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, University Hospital "Fundacion Jimenez Diaz", Autonomous University of Madrid, E-28040 Madrid, Spain
| | - Rebeca Manso
- Pathology Department, University Hospital "Fundacion Jimenez Diaz", Autonomous University of Madrid, E-28040 Madrid, Spain
| | - Juan Madoz-Gúrpide
- Pathology Department, University Hospital "Fundacion Jimenez Diaz", Autonomous University of Madrid, E-28040 Madrid, Spain
| | - Cristina Caramés
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, University Hospital "Fundacion Jimenez Diaz", Autonomous University of Madrid, E-28040 Madrid, Spain
| | - Laura del Puerto-Nevado
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, University Hospital "Fundacion Jimenez Diaz", Autonomous University of Madrid, E-28040 Madrid, Spain
| | - Federico Rojo
- Pathology Department, University Hospital "Fundacion Jimenez Diaz", Autonomous University of Madrid, E-28040 Madrid, Spain
| | - Jesús García-Foncillas
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, University Hospital "Fundacion Jimenez Diaz", Autonomous University of Madrid, E-28040 Madrid, Spain.
| |
Collapse
|
44
|
|
45
|
Kubota D, Yoshida A, Kawai A, Kondo T. Proteomics Identified Overexpression of SET Oncogene Product and Possible Therapeutic Utility of Protein Phosphatase 2A in Alveolar Soft Part Sarcoma. J Proteome Res 2014; 13:2250-61. [DOI: 10.1021/pr400929h] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Daisuke Kubota
- Division
of Pharmacoproteomics, National Cancer Center Research Institute, Tsukiji
5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | | | | | - Tadashi Kondo
- Division
of Pharmacoproteomics, National Cancer Center Research Institute, Tsukiji
5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| |
Collapse
|
46
|
Cristóbal I, Manso R, Rincón R, Caramés C, Senin C, Borrero A, Martínez-Useros J, Rodriguez M, Zazo S, Aguilera O, Madoz-Gúrpide J, Rojo F, García-Foncillas J. PP2A inhibition is a common event in colorectal cancer and its restoration using FTY720 shows promising therapeutic potential. Mol Cancer Ther 2014; 13:938-47. [PMID: 24448818 DOI: 10.1158/1535-7163.mct-13-0150] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Protein phosphatase 2A (PP2A) is a tumor suppressor that regulates many signaling pathways crucial for cell transformation. In fact, decreased activity of PP2A has been reported as a recurrent alteration in many types of cancer. Here, we show that PP2A is frequently inactivated in patients with colorectal cancer, indicating that PP2A represents a potential therapeutic target for this disease. We identified overexpression of the endogenous PP2A inhibitors SET and CIP2A, and downregulation of regulatory PP2A such as PPP2R2A and PPP2R5E, as contributing mechanisms to PP2A inhibition in colorectal cancer. Moreover, we observed that its restoration using FTY720 impairs proliferation and clonogenic potential of colorectal cancer cells, induces caspase-dependent apoptosis, and affects AKT and extracellular signal-regulated kinase-1/2 activation status. Interestingly, treatment with FTY720 showed an additive effect with 5-fluorouracil, SN-38, and oxaliplatin, drugs used in standard chemotherapy in patients with colorectal cancer. These results suggest that PP2A activity is commonly decreased in colorectal cancer cells, and that the use of PP2A activators, such as FTY720, might represent a potential novel therapeutic strategy in colorectal cancer.
Collapse
Affiliation(s)
- Ion Cristóbal
- Authors' Affiliations: Translational Oncology Division, Health Research Institute and Oncology Department, University Hospital "Fundación Jiménez Díaz," Autonomous University of Madrid; Pathology Department, IIS "Fundación Jiménez Díaz," Madrid, Spain; and Medical Oncology Department, University Hospital of Vigo, Vigo, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Agarwal A, MacKenzie RJ, Pippa R, Eide CA, Oddo J, Tyner JW, Sears R, Vitek MP, Odero MD, Christensen DJ, Druker BJ. Antagonism of SET using OP449 enhances the efficacy of tyrosine kinase inhibitors and overcomes drug resistance in myeloid leukemia. Clin Cancer Res 2014; 20:2092-103. [PMID: 24436473 DOI: 10.1158/1078-0432.ccr-13-2575] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The SET oncoprotein, a potent inhibitor of the protein phosphatase 2A (PP2A), is overexpressed in leukemia. We evaluated the efficacy of SET antagonism in chronic myeloid leukemia (CML) and acute myeloid leukemia (AML) cell lines, a murine leukemia model, and primary patient samples using OP449, a specific, cell-penetrating peptide that antagonizes SET's inhibition of PP2A. EXPERIMENTAL DESIGN In vitro cytotoxicity and specificity of OP449 in CML and AML cell lines and primary samples were measured using proliferation, apoptosis, and clonogenic assays. Efficacy of target inhibition by OP449 was evaluated by immunoblotting and PP2A assay. In vivo antitumor efficacy of OP449 was measured in human HL-60 xenografted murine model. RESULTS We observed that OP449 inhibited growth of CML cells including those from patients with blastic phase disease and patients harboring highly drug-resistant BCR-ABL1 mutations. Combined treatment with OP449 and ABL1 tyrosine kinase inhibitors was significantly more cytotoxic to K562 cells and primary CD34(+) CML cells. SET protein levels remained unchanged with OP449 treatment, but BCR-ABL1-mediated downstream signaling was significantly inhibited with the degradation of key signaling molecules such as BCR-ABL1, STAT5, and AKT. Similarly, AML cell lines and primary patient samples with various genetic lesions showed inhibition of cell growth after treatment with OP449 alone or in combination with respective kinase inhibitors. Finally, OP449 reduced the tumor burden of mice xenografted with human leukemia cells. CONCLUSIONS We demonstrate a novel therapeutic paradigm of SET antagonism using OP449 in combination with tyrosine kinase inhibitors for the treatment of CML and AML.
Collapse
Affiliation(s)
- Anupriya Agarwal
- Authors' Affiliations: Knight Cancer Institute; Division of Hematology & Medical Oncology; Departments of Cell and Developmental Biology and Molecular and Medical Genetics, Oregon Health and Science University; Howard Hughes Medical Institute, Portland, Oregon; Oncotide Pharmaceuticals, Research Triangle Park; Duke University Medical Center, Durham, North Carolina; and Division of Oncology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Honarpour N, Rose CM, Brumbaugh J, Anderson J, Graham RLJ, Sweredoski MJ, Hess S, Coon JJ, Deshaies RJ. F-box protein FBXL16 binds PP2A-B55α and regulates differentiation of embryonic stem cells along the FLK1+ lineage. Mol Cell Proteomics 2014; 13:780-91. [PMID: 24390425 PMCID: PMC3945908 DOI: 10.1074/mcp.m113.031765] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The programmed formation of specific tissues from embryonic stem cells is a major goal of regenerative medicine. To identify points of intervention in cardiac tissue formation, we performed an siRNA screen in murine embryonic stem cells to identify ubiquitin system genes that repress cardiovascular tissue formation. Our screen uncovered an F-box protein, Fbxl16, as a repressor of one of the earliest steps in the cardiogenic lineage: FLK1+ progenitor formation. Whereas F-box proteins typically form SCF ubiquitin ligases, shotgun mass spectrometry revealed that FBXL16 instead binds protein phosphatase 2A (PP2A) containing a B55 specificity subunit (PP2A(B55)). Phosphoproteomic analyses indicate that FBXL16 negatively regulates phosphorylation of the established PP2A(B55) substrate, vimentin. We suggest that FBXL16 negatively regulates the activity of B55α-PP2A to modulate the genesis of FLK1+ progenitor cells.
Collapse
|
49
|
Yang R, Yang L, Qiu F, Zhang L, Wang H, Yang X, Deng J, Fang W, Zhou Y, Lu J. Functional genetic polymorphisms in PP2A subunit genes confer increased risks of lung cancer in southern and eastern Chinese. PLoS One 2013; 8:e77285. [PMID: 24204789 PMCID: PMC3812212 DOI: 10.1371/journal.pone.0077285] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Accepted: 09/02/2013] [Indexed: 12/28/2022] Open
Abstract
Protein phosphatase-2A (PP2A) is one of the major cellular serine-threonine phosphatases and functions as a tumor suppressor that negatively regulates the activity of some oncogenic kinases. Recent studies have reported that PP2A expression was suppressed during lung carcinogenesis, we there hypothesized that the single nucleotide polymorphisms (SNPs) in PP2A subunit genes may affect PP2A function and thus contribute to lung cancer susceptibility. In a two-stage case-control study with a total of 1559 lung cancer patients and 1679 controls, we genotyped eight putative functional SNPs and one identified functional SNP (i.e., rs11453459) in seven major PP2A subunits (i.e., PPP2R1A, PPP2R1B, PPP2CA, PPP2R2A, PPP2R2B, PPP2R5C, PPP2R5E) in southern and eastern Chinese. We found that rs11453459G (-G/GG) variant genotypes of PPP2R1A and the rs1255722AA variant genotype of PPP2R5E conferred increased risks of lung cancer (rs11453459, -G/GG vs. –: OR = 1.31, 95% CI = 1.13–1.51; rs1255722, AA vs. AG/GG: OR = 1.27, 95% CI = 1.07–1.51). After combined the two variants, the number of the adverse genotypes was positively associated with lung cancer risk in a dose-response manner (Ptrend = 5.63×10−6). Further functional assay showed that lung cancer tissues carrying rs1255722AA variant genotype had a significantly lower mRNA level of PPP2R5E compared with tissues carrying GG/GA genotypes. However, such effect was not observed for the other SNPs and other combinations. Our findings suggested that the two functional variants in PPP2R1A and PPP2R5E and their combination are associated with lung cancer risk in Chinese, which may be valuable biomarkers to predict risk of lung cancer.
Collapse
Affiliation(s)
- Rongrong Yang
- The Institute for Chemical Carcinogenesis, The State Key Lab of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
PKR negatively regulates leukemia progression in association with PP2A activation, Bcl-2 inhibition and increased apoptosis. Blood Cancer J 2013; 3:e144. [PMID: 24013665 PMCID: PMC3789206 DOI: 10.1038/bcj.2013.42] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 07/21/2013] [Accepted: 07/24/2013] [Indexed: 01/05/2023] Open
Abstract
Reduced expression and activity of the proapoptotic, double-stranded RNA-dependent protein kinase, PKR (protein kinase R) is observed in breast, lung and various leukemias, suggesting that loss of PKR potentiates transformation. Now we report that decreased PKR activity inhibits chemotherapy-induced apoptosis of leukemia cells both in vitro and in vivo. Inhibition of PKR expression or activity reduces protein phosphatase 2A (PP2A) activity, a B-cell lymphoma 2 (Bcl-2) phosphatase, resulting in enhanced Bcl-2 phosphorylation. Thus, inhibition of PKR activity leads to hyperphosphorylation of Bcl-2, stabilization of Bcl-2/Bax interaction and decreased Bax insertion into the outer mitochondrial membrane. Treatment with the PP2A activator, FTY720, restores Bcl-2 dephosphorylation and apoptosis in cells with reduced PKR expression following stress. Significantly, xenografts of REH leukemic cells with reduced PKR display significantly increased tumor volume, increased resistance to doxorubicin treatment and shorter survival. Importantly, FTY720 treatment restores sensitivity to chemotherapy and prolongs overall survival of these mice. Collectively, these findings suggest that PP2A activation is a downstream target of PKR and the PKR/PP2A signaling axis is required for rapid and potent stress-induced apoptosis. Importantly, loss of PKR promotes leukemia progression and may serve as a biomarker for predicting chemosensitivity.
Collapse
|