1
|
Zhang H, Ge G, Zhang W, Sun H, Liang X, Xia Y, Du J, Wu Z, Bai J, Yang H, Yang X, Zhou J, Xu Y, Geng D. PP2Ac Regulates Autophagy via Mediating mTORC1 and ULK1 During Osteoclastogenesis in the Subchondral Bone of Osteoarthritis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404080. [PMID: 39041921 PMCID: PMC11423161 DOI: 10.1002/advs.202404080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/02/2024] [Indexed: 07/24/2024]
Abstract
The molecular mechanism underlying abnormal osteoclastogenesis triggering subchondral bone remodeling in osteoarthritis (OA) is still unclear. Here, single-cell and bulk transcriptomics sequencing analyses are performed on GEO datasets to identify key molecules and validate them using knee joint tissues from OA patients and rat OA models. It is found that the catalytic subunit of protein phosphatase 2A (PP2Ac) is highly expressed during osteoclastogenesis in the early stage of OA and is correlated with autophagy. Knockdown or inhibition of PP2Ac weakened autophagy during osteoclastogenesis. Furthermore, the ULK1 expression of the downstream genes is significantly increased when PP2Ac is knocked down. PP2Ac-mediated autophagy is dependent on ULK1 phosphorylation activity during osteoclastogenesis, which is associated with enhanced dephosphorylation of ULK1 Ser637 residue regulating at the post-translational level. Additionally, mTORC1 inhibition facilitated the expression level of PP2Ac during osteoclastogenesis. In animal OA models, decreasing the expression of PP2Ac ameliorated early OA progression. The findings suggest that PP2Ac is also a promising therapeutic target in early OA.
Collapse
Affiliation(s)
- Haifeng Zhang
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
- Department of Orthopaedic SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
| | - Gaoran Ge
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
| | - Wei Zhang
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
| | - Houyi Sun
- Department of OrthopedicsQilu Hospital of Shandong UniversityJinanShandong250063China
| | - Xiaolong Liang
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
| | - Yu Xia
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
| | - Jiacheng Du
- Department of Biochemistry and Molecular BiologyJeonbuk National University Medical SchoolJeonjuJeonbuk54896South Korea
| | - Zerui Wu
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
- Department of OrthopedicsChangshu Hospital Affiliated to Soochow UniversityChangshuJiangsu215501China
| | - Jiaxiang Bai
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
- Department of Orthopedicsthe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui234000China
| | - Huilin Yang
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
| | - Xing Yang
- Orthopedics and Sports Medicine CenterSuzhou Municipal HospitalNanjing Medical University Affiliated Suzhou Hospital242, Guangji RoadSuzhouJiangsu215008China
| | - Jun Zhou
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
| | - Yaozeng Xu
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
| | - Dechun Geng
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
| |
Collapse
|
2
|
Hong J, Li Y, Chen L, Han D, Li Y, Mi X, Liu K, Wang Q, Song Y, Liu T, Yang N, Liu Y, Li Z, Guo X. A53T α-synuclein mutation increases susceptibility to postoperative delayed neurocognitive recovery via hippocampal Ang-(1-7)/MasR axis. Biochem Pharmacol 2024; 224:116261. [PMID: 38705534 DOI: 10.1016/j.bcp.2024.116261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/28/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Delayed neurocognitive recovery (dNCR) is a common complication in geriatric surgical patients. The impact of anesthesia and surgery on patients with neurodegenerative diseases, such as Parkinson's disease (PD) or prion disease, has not yet been reported. In this study, we aimed to determine the association between a pre-existing A53T genetic background, which involves a PD-related point mutation, and the development of postoperative dNCR. We observed that partial hepatectomy induced hippocampus-dependent cognitive deficits in 5-month-old A53T transgenic mice, a model of early-stage PD without cognitive deficits, unlike in age-matched wild-type (WT) mice. We respectively examined molecular changes at 6 h, 1 day, and 2 days after partial hepatectomy and observed that cognitive changes were accompanied by weakened angiotensin-(1-7)/Mas receptor [Ang-(1-7)/MasR] axis, increased alpha-synuclein (α-syn) expression and phosphorylation, decreased methylated protein phosphatase-2A (Me-PP2A), and prompted microglia M1 polarization and neuronal apoptosis in the hippocampus at 1 day after surgery. Nevertheless, no changes in blood-brain barrier (BBB) integrity or plasma α-syn levels in either A53T or WT mice. Furthermore, intranasal administration of selective MasR agonist AVE 0991, reversed the mentioned cognitive deficits in A53T mice, enhanced MasR expression, reduced α-syn accumulation and phosphorylation, and attenuated microglia activation and apoptotic response. Our findings suggest that individuals with the A53T genetic background may be more susceptible to developing postoperative dNCR. This susceptibility could be linked to central α-syn accumulation mediated by the weakened Ang-(1-7)/MasR/methyl-PP2A signaling pathway in the hippocampus following surgery, independent of plasma α-syn level and BBB.
Collapse
Affiliation(s)
- Jingshu Hong
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yue Li
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Lei Chen
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Dengyang Han
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yitong Li
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Xinning Mi
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Kaixi Liu
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Qian Wang
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yanan Song
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Beijing Center of Quality Control and Improvement on Clinical Anesthesia, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Taotao Liu
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Ning Yang
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yajie Liu
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Zhengqian Li
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Beijing Center of Quality Control and Improvement on Clinical Anesthesia, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Anesthesia and Perioperative Medicine Branch of China International Exchange and Promotive Association for Medical and Health Care (CPAM), No. 49, North Garden Street, Haidian District, Beijing 100191, China.
| | - Xiangyang Guo
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Beijing Center of Quality Control and Improvement on Clinical Anesthesia, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Anesthesia and Perioperative Medicine Branch of China International Exchange and Promotive Association for Medical and Health Care (CPAM), No. 49, North Garden Street, Haidian District, Beijing 100191, China.
| |
Collapse
|
3
|
Hassan M, Yasir M, Shahzadi S, Chun W, Kloczkowski A. Molecular Role of Protein Phosphatases in Alzheimer's and Other Neurodegenerative Diseases. Biomedicines 2024; 12:1097. [PMID: 38791058 PMCID: PMC11117500 DOI: 10.3390/biomedicines12051097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/02/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Alzheimer's disease (AD) is distinguished by the gradual loss of cognitive function, which is associated with neuronal loss and death. Accumulating evidence supports that protein phosphatases (PPs; PP1, PP2A, PP2B, PP4, PP5, PP6, and PP7) are directly linked with amyloid beta (Aβ) as well as the formation of the neurofibrillary tangles (NFTs) causing AD. Published data reported lower PP1 and PP2A activity in both gray and white matters in AD brains than in the controls, which clearly shows that dysfunctional phosphatases play a significant role in AD. Moreover, PP2A is also a major causing factor of AD through the deregulation of the tau protein. Here, we review recent advances on the role of protein phosphatases in the pathology of AD and other neurodegenerative diseases. A better understanding of this problem may lead to the development of phosphatase-targeted therapies for neurodegenerative disorders in the near future.
Collapse
Affiliation(s)
- Mubashir Hassan
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH 43205, USA;
| | - Muhammad Yasir
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (M.Y.); (W.C.)
| | - Saba Shahzadi
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH 43205, USA;
| | - Wanjoo Chun
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (M.Y.); (W.C.)
| | - Andrzej Kloczkowski
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH 43205, USA;
- Department of Pediatrics, The Ohio State University, Columbus, OH 43205, USA
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
4
|
de Haan LR, van Golen RF, Heger M. Molecular Pathways Governing the Termination of Liver Regeneration. Pharmacol Rev 2024; 76:500-558. [PMID: 38697856 DOI: 10.1124/pharmrev.123.000955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/24/2024] [Accepted: 02/08/2024] [Indexed: 05/05/2024] Open
Abstract
The liver has the unique capacity to regenerate, and up to 70% of the liver can be removed without detrimental consequences to the organism. Liver regeneration is a complex process involving multiple signaling networks and organs. Liver regeneration proceeds through three phases: the initiation phase, the growth phase, and the termination phase. Termination of liver regeneration occurs when the liver reaches a liver-to-body weight that is required for homeostasis, the so-called "hepatostat." The initiation and growth phases have been the subject of many studies. The molecular pathways that govern the termination phase, however, remain to be fully elucidated. This review summarizes the pathways and molecules that signal the cessation of liver regrowth after partial hepatectomy and answers the question, "What factors drive the hepatostat?" SIGNIFICANCE STATEMENT: Unraveling the pathways underlying the cessation of liver regeneration enables the identification of druggable targets that will allow us to gain pharmacological control over liver regeneration. For these purposes, it would be useful to understand why the regenerative capacity of the liver is hampered under certain pathological circumstances so as to artificially modulate the regenerative processes (e.g., by blocking the cessation pathways) to improve clinical outcomes and safeguard the patient's life.
Collapse
Affiliation(s)
- Lianne R de Haan
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, China (L.R.d.H., M.H.); Department of Internal Medicine, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands (L.R.d.H.); Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands (R.F.v.G.); Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands (M.H.); and Membrane Biochemistry and Biophysics, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, The Netherlands (M.H.)
| | - Rowan F van Golen
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, China (L.R.d.H., M.H.); Department of Internal Medicine, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands (L.R.d.H.); Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands (R.F.v.G.); Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands (M.H.); and Membrane Biochemistry and Biophysics, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, The Netherlands (M.H.)
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, China (L.R.d.H., M.H.); Department of Internal Medicine, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands (L.R.d.H.); Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands (R.F.v.G.); Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands (M.H.); and Membrane Biochemistry and Biophysics, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, The Netherlands (M.H.)
| |
Collapse
|
5
|
Brewer A, Sathe G, Pflug BE, Clarke RG, Macartney TJ, Sapkota GP. Mapping the substrate landscape of protein phosphatase 2A catalytic subunit PPP2CA. iScience 2024; 27:109302. [PMID: 38450154 PMCID: PMC10915630 DOI: 10.1016/j.isci.2024.109302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/18/2023] [Accepted: 02/16/2024] [Indexed: 03/08/2024] Open
Abstract
Protein phosphatase 2A (PP2A) is an essential Ser/Thr phosphatase. The PP2A holoenzyme complex comprises a scaffolding (A), regulatory (B), and catalytic (C) subunit, with PPP2CA being the principal catalytic subunit. The full scope of PP2A substrates in cells remains to be defined. To address this, we employed dTAG proteolysis-targeting chimeras to efficiently and selectively degrade dTAG-PPP2CA in homozygous knock-in HEK293 cells. Unbiased global phospho-proteomics identified 2,204 proteins with significantly increased phosphorylation upon dTAG-PPP2CA degradation, implicating them as potential PPP2CA substrates. A vast majority of these are novel. Bioinformatic analyses revealed involvement of the potential PPP2CA substrates in spliceosome function, cell cycle, RNA transport, and ubiquitin-mediated proteolysis. We identify a pSP/pTP motif as a predominant target for PPP2CA and confirm some of our phospho-proteomic data with immunoblotting. We provide an in-depth atlas of potential PPP2CA substrates and establish targeted degradation as a robust tool to unveil phosphatase substrates in cells.
Collapse
Affiliation(s)
- Abigail Brewer
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Gajanan Sathe
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Billie E. Pflug
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Rosemary G. Clarke
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Thomas J. Macartney
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Gopal P. Sapkota
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| |
Collapse
|
6
|
Hu Z, Meng G, Zhang L, Zhang M, Cui H, Tang H. Protective effect of Huanglianjiedu Decoction on microcystin-LR induced nerve injury. Comp Biochem Physiol C Toxicol Pharmacol 2023; 272:109698. [PMID: 37442312 DOI: 10.1016/j.cbpc.2023.109698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/26/2023] [Accepted: 07/09/2023] [Indexed: 07/15/2023]
Abstract
Microcystin-LR (MC-LR) presented in eutrophic water has been identified as having the capacity to induce damage to the mammalian nervous system by crossing the blood-brain barrier through organic anion transporting polypeptides. However, the lack of effective preventive and protective strategies remains a concern. Huanglianjiedu Decoction (HLJD), a classical Chinese traditional formula originating from the Tang Dynasty and comprising Rhizoma Coptidis, Radix Scutellariae, Cortex Phellodendri, and Fructus Gardeniae, has exhibited neuroprotective effects attributed to its antioxidant properties. In this study, we investigated the potential of HLJD in counteracting the neurotoxic effects induced by MC-LR. Our findings revealed that MC-LR dose-dependently inhibited the activity of the PP2A enzyme in PC 12 cells and significantly elevated the phosphorylation levels of JNK, ERK1/2, and p38. Moreover, MC-LR administration resulted in synaptic damage in mouse neurons, hyperphosphorylation of the microtubule-related protein Tau, cognitive impairment, and deficits in learning and memory in C57BL/6J mice. Notably, HLJD effectively reversed the cytotoxicity caused by MC-LR in PC 12 cells, and attenuated MC-LR-induced neuronal damage while improving learning ability in mice. These results highlight the potential of HLJD as a promising protective strategy against MC-LR-induced neurological injury.
Collapse
Affiliation(s)
- Zhengqiang Hu
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Guanmin Meng
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada; Department of Clinical Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310013, China
| | - Lejun Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Meng Zhang
- Department of Clinical Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310013, China
| | - Huashun Cui
- Department of Acupuncture and Moxibustion, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China.
| | - Huifang Tang
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
7
|
Ferreira AF, Santiago J, Silva JV, Oliveira PF, Fardilha M. PP1, PP2A and PP2B Interplay in the Regulation of Sperm Motility: Lessons from Protein Phosphatase Inhibitors. Int J Mol Sci 2022; 23:ijms232315235. [PMID: 36499559 PMCID: PMC9737803 DOI: 10.3390/ijms232315235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/11/2022] Open
Abstract
Male fertility relies on the ability of spermatozoa to fertilize the egg in the female reproductive tract (FRT). Spermatozoa acquire activated motility during epididymal maturation; however, to be capable of fertilization, they must achieve hyperactivated motility in the FRT. Extensive research found that three protein phosphatases (PPs) are crucial to sperm motility regulation, the sperm-specific protein phosphatase type 1 (PP1) isoform gamma 2 (PP1γ2), protein phosphatase type 2A (PP2A) and protein phosphatase type 2B (PP2B). Studies have reported that PP activity decreases during epididymal maturation, whereas protein kinase activity increases, which appears to be a requirement for motility acquisition. An interplay between these PPs has been extensively investigated; however, many specific interactions and some inconsistencies remain to be elucidated. The study of PPs significantly advanced following the identification of naturally occurring toxins, including calyculin A, okadaic acid, cyclosporin, endothall and deltamethrin, which are powerful and specific PP inhibitors. This review aims to overview the protein phosphorylation-dependent biochemical pathways underlying sperm motility acquisition and hyperactivation, followed by a discussion of the PP inhibitors that allowed advances in the current knowledge of these pathways. Since male infertility cases still attain alarming numbers, additional research on the topic is required, particularly using other PP inhibitors.
Collapse
Affiliation(s)
- Ana F. Ferreira
- Laboratory of Signal Transduction, Institute for Biomedicine-iBiMED, Medical Sciences Department, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Joana Santiago
- Laboratory of Signal Transduction, Institute for Biomedicine-iBiMED, Medical Sciences Department, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Joana V. Silva
- Laboratory of Signal Transduction, Institute for Biomedicine-iBiMED, Medical Sciences Department, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Pedro F. Oliveira
- QOPNA & LAQV, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Margarida Fardilha
- Laboratory of Signal Transduction, Institute for Biomedicine-iBiMED, Medical Sciences Department, University of Aveiro, 3810-193 Aveiro, Portugal
- Correspondence: ; Tel.: +351-918-143-947
| |
Collapse
|
8
|
Friedrich D, Marintchev A, Arthanari H. The metaphorical swiss army knife: The multitude and diverse roles of HEAT domains in eukaryotic translation initiation. Nucleic Acids Res 2022; 50:5424-5442. [PMID: 35552740 PMCID: PMC9177959 DOI: 10.1093/nar/gkac342] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 11/24/2022] Open
Abstract
Biomolecular associations forged by specific interaction among structural scaffolds are fundamental to the control and regulation of cell processes. One such structural architecture, characterized by HEAT repeats, is involved in a multitude of cellular processes, including intracellular transport, signaling, and protein synthesis. Here, we review the multitude and versatility of HEAT domains in the regulation of mRNA translation initiation. Structural and cellular biology approaches, as well as several biophysical studies, have revealed that a number of HEAT domain-mediated interactions with a host of protein factors and RNAs coordinate translation initiation. We describe the basic structural architecture of HEAT domains and briefly introduce examples of the cellular processes they dictate, including nuclear transport by importin and RNA degradation. We then focus on proteins in the translation initiation system featuring HEAT domains, specifically the HEAT domains of eIF4G, DAP5, eIF5, and eIF2Bϵ. Comparative analysis of their remarkably versatile interactions, including protein-protein and protein-RNA recognition, reveal the functional importance of flexible regions within these HEAT domains. Here we outline how HEAT domains orchestrate fundamental aspects of translation initiation and highlight open mechanistic questions in the area.
Collapse
Affiliation(s)
- Daniel Friedrich
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Assen Marintchev
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA, USA
| | - Haribabu Arthanari
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Maitiabula G, Tian F, Wang P, Zhang L, Gao X, Wan S, Sun H, Yang J, Zhang Y, Gao T, Xue B, Li C, Li J, Wang X. Liver PP2A-Cα Protects From Parenteral Nutrition-associated Hepatic Steatosis. Cell Mol Gastroenterol Hepatol 2022; 14:669-692. [PMID: 35643235 PMCID: PMC9421584 DOI: 10.1016/j.jcmgh.2022.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 05/18/2022] [Accepted: 05/18/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND & AIMS Parenteral nutrition (PN) is a lifesaving therapy for patients with intestinal failure. Hepatic steatosis is a potentially fatal complication of long-term PN, but the involved pathological mechanisms are incompletely unclarified. Herein, we identify the role of protein phosphatase 2A (PP2A) in the pathogenesis of parenteral nutrition-associated hepatic steatosis (PNAHS). METHODS Proteomic/phosphoproteomic analyses of liver samples from patients with PNAHS were applied to identify the mechanism of PNAHS. Total parenteral nutrition (TPN) mice model, in vivo, and in vitro experiments were used to assess the effect of PP2A-Cα on liver fatty acid metabolism. RESULTS Reduced expression of PP2A-Cα (catalytic subunit) enhanced activation of serine/threonine kinase Akt2 and decreased activation of adenosine monophosphate-activated protein kinase (AMPK) were associated with hepatic steatosis in patients with PNAHS. Mice given PN for 14 days developed hepatic steatosis, down-regulation of PP2A-Cα, activation of Akt2, and inhibition of AMPK. Hepatocyte-specific deletion of PP2A-Cα in mice given PN exacerbated Akt2 activation, AMPK inhibition, and hepatic steatosis through an effect on fatty acid degradation, whereas hepatocyte-specific PP2A-Cα overexpression significantly ameliorated hepatic steatosis accompanied with Akt2 suppression and AMPK activation. Additionally, pharmacological activation of Akt2 in mice overexpressing PP2A-Cα led to the aggravation of hepatic steatosis. CONCLUSIONS Our findings demonstrate that hepatic PP2A-Cα serves as a protective factor of PNAHS due to ameliorating hepatic steatosis and improving liver function. Our study provides a strong rationale that PP2A-Cα may be involved in the pathogenesis of PNAHS.
Collapse
Affiliation(s)
- Gulisudumu Maitiabula
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Feng Tian
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Peng Wang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Li Zhang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xuejin Gao
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Songlin Wan
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Haifeng Sun
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jianbo Yang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yupeng Zhang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Tingting Gao
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Bin Xue
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center of the Medical School of Nanjing University, Nanjing, China,Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China,Bin Xue, PhD, LongMian Avenue, Nanjing 211166, China. tel: +86-25-87115542
| | - Chaojun Li
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center of the Medical School of Nanjing University, Nanjing, China,Chaojun Li, PhD, Hankou Road, Nanjing, 210093, China. tel: +86-25-83596289.
| | - Jieshou Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xinying Wang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China,Correspondence Address correspondence to: Xinying Wang, MD, PhD, Department of General Surgery, Jinling Hospital, Medical School of Nanjing University. 305 East Zhongshan Road, Nanjing, 210002, China. tel: +86-25-80861429
| |
Collapse
|
10
|
Protein Extract of Tobacco Expressing Solanum torvum PP5-Encoding Gene Inhibits Verticillium dahliae Proliferation. HORTICULTURAE 2022. [DOI: 10.3390/horticulturae8030252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Verticillium wilt, a soilborne disease caused by Verticillium dahliae (V. dahliae), can severely affect the yields of Solanaceae crops. In a previous study, it was observed in Solanum torvum (S. torvum) that protein phosphatase 5 (PP5) was induced by V. dahliae infection. To elucidate the function of PP5 more clearly, this study cloned an StPP5 cDNA from S. torvum by PCR. The cDNA contained an ORF of 1458 bp long encoding a putative protein of 485 amino acid residues with a predicted molecular mass of 54.63 kDa and a theoretical isoelectric point of 5.66. StPP5 protein contained a conserved PP domain and showed high similarity to other homologous members of the PP5 family from various plant species. The expression of StPP5 gene was upregulated after V. infection and reached its maximum value at 24 h in leaves. In order to clarify the role of StPP5, four transgenic tobacco plants expressing StPP5 were generated through Agrobacterium-mediated transformation and identified by PCR. In vitro culture assay showed that the growth of V. dahliae in PDA medium containing proteins extracted from the leaves of transgenic tobacco line P6 was inhibited, whose inhibition rate was 55.1%, higher than the non-transgenic control. These results indicated that StPP5 might be involved in plant defense against V. dahliae infection.
Collapse
|
11
|
Lin W, Hung TC, Kurobe T, Wang Y, Yang P. Microcystin-Induced Immunotoxicity in Fishes: A Scoping Review. Toxins (Basel) 2021; 13:765. [PMID: 34822549 PMCID: PMC8623247 DOI: 10.3390/toxins13110765] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/26/2022] Open
Abstract
Cyanobacteria (blue-green algae) have been present on Earth for over 2 billion years, and can produce a variety of bioactive molecules, such as cyanotoxins. Microcystins (MCs), the most frequently detected cyanotoxins, pose a threat to the aquatic environment and to human health. The classic toxic mechanism of MCs is the inhibition of the protein phosphatases 1 and 2A (PP1 and PP2A). Immunity is known as one of the most important physiological functions in the neuroendocrine-immune network to prevent infections and maintain internal homoeostasis in fish. The present review aimed to summarize existing papers, elaborate on the MC-induced immunotoxicity in fish, and put forward some suggestions for future research. The immunomodulatory effects of MCs in fish depend on the exposure concentrations, doses, time, and routes of exposure. Previous field and laboratory studies provided strong evidence of the associations between MC-induced immunotoxicity and fish death. In our review, we summarized that the immunotoxicity of MCs is primarily characterized by the inhibition of PP1 and PP2A, oxidative stress, immune cell damage, and inflammation, as well as apoptosis. The advances in fish immunoreaction upon encountering MCs will benefit the monitoring and prediction of fish health, helping to achieve an ecotoxicological goal and to ensure the sustainability of species. Future studies concerning MC-induced immunotoxicity should focus on adaptive immunity, the hormesis phenomenon and the synergistic effects of aquatic microbial pathogens.
Collapse
Affiliation(s)
- Wang Lin
- Hunan Provincial Collaborative Innovation Center for Efficient and Health Production of Fisheries, Hunan Provincial Key Laboratory for Health Aquaculture and Product Processing in Dongting Lake Area, Hunan Provincial Key Laboratory for Molecular Immunity Technology of Aquatic Animal Diseases, Hunan Engineering Research Center of Aquatic Organism Resources and Environmental Ecology, Zoology Key Laboratory of Hunan Higher Education, College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde 415000, China;
- Department of Biological and Agricultural Engineering, University of California, Davis, CA 95616, USA; (T.-C.H.); (Y.W.)
- Department of Fisheries Resources and Environment, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Tien-Chieh Hung
- Department of Biological and Agricultural Engineering, University of California, Davis, CA 95616, USA; (T.-C.H.); (Y.W.)
| | - Tomofumi Kurobe
- Department of Anatomy, Physiology, and Cell Biology, University of California, Davis, CA 95616, USA;
| | - Yi Wang
- Department of Biological and Agricultural Engineering, University of California, Davis, CA 95616, USA; (T.-C.H.); (Y.W.)
| | - Pinhong Yang
- Hunan Provincial Collaborative Innovation Center for Efficient and Health Production of Fisheries, Hunan Provincial Key Laboratory for Health Aquaculture and Product Processing in Dongting Lake Area, Hunan Provincial Key Laboratory for Molecular Immunity Technology of Aquatic Animal Diseases, Hunan Engineering Research Center of Aquatic Organism Resources and Environmental Ecology, Zoology Key Laboratory of Hunan Higher Education, College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde 415000, China;
| |
Collapse
|
12
|
Shao L, Ma Y, Fang Q, Huang Z, Wan S, Wang J, Yang L. Role of protein phosphatase 2A in kidney disease (Review). Exp Ther Med 2021; 22:1236. [PMID: 34539832 PMCID: PMC8438693 DOI: 10.3892/etm.2021.10671] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
Kidney disease affects millions of people worldwide and is a financial burden on the healthcare system. Protein phosphatase 2A (PP2A), which is involved in renal development and the function of ion-transport proteins, aquaporin-2 and podocytes, is likely to serve an important role in renal processes. PP2A is associated with the pathogenesis of a variety of different kidney diseases including podocyte injury, inflammation, tumors and chronic kidney disease. The current review aimed to discuss the structure and function of PP2A subunits in the context of kidney diseases. How dysregulation of PP2A in the kidneys causes podocyte death and the inactivation of PP2A in renal carcinoma tissues is discussed. Inhibition of PP2A activity prevents epithelial-mesenchymal transition and attenuates renal fibrosis, creating a favorable inflammatory microenvironment and promoting the initiation and progression of tumor pathogenesis. The current review also indicates that PP2A serves an important role in protection against renal inflammation. Understanding the detailed mechanisms of PP2A provides information that can be utilized in the design and application of novel therapeutics for the treatment and prevention of renal diseases.
Collapse
Affiliation(s)
- Lishi Shao
- Department of Radiology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Yiqun Ma
- Department of Radiology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Qixiang Fang
- Department of Urology, The First Affiliated Hospital of the Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P.R. China
| | - Ziye Huang
- Department of Urology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Shanshan Wan
- Department of Radiology, Yunnan Kun-Gang Hospital, Anning, Yunnan 650300, P.R. China
| | - Jiaping Wang
- Department of Radiology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Li Yang
- Department of Anatomy, Histology and Embryology, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
13
|
Chu H, Sacharidou A, Nguyen A, Li C, Chambliss KL, Salmon JE, Shen YM, Lo J, Leone GW, Herz J, Hui DY, Marciano DK, Abrahams VM, Natale BV, Montalbano AP, Xiao X, Xu L, Natale DR, Shaul PW, Mineo C. Protein Phosphatase 2A Activation Via ApoER2 in Trophoblasts Drives Preeclampsia in a Mouse Model of the Antiphospholipid Syndrome. Circ Res 2021; 129:735-750. [PMID: 34404233 DOI: 10.1161/circresaha.120.318941] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Haiyan Chu
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Anastasia Sacharidou
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - An Nguyen
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Chun Li
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Ken L Chambliss
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Jane E Salmon
- Medicine, Hospital for Special Surgery, Weill Cornell Medical College, New York (J.E.S.)
| | - Yu-Min Shen
- Internal Medicine (Y.-M.S., D.K.M.), University of Texas Southwestern Medical Center, Dallas
| | - Julie Lo
- Obstetrics and Gynecology (J.L.), University of Texas Southwestern Medical Center, Dallas
| | - Gustavo W Leone
- Froedtert-Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee (G.W.L.)
| | - Joachim Herz
- Molecular Genetics (J.H.), University of Texas Southwestern Medical Center, Dallas
| | - David Y Hui
- Pathology, University of Cincinnati College of Medicine (D.Y.H.)
| | - Denise K Marciano
- Internal Medicine (Y.-M.S., D.K.M.), University of Texas Southwestern Medical Center, Dallas.,Cell Biology (D.K.M., C.M.), University of Texas Southwestern Medical Center, Dallas
| | - Vikki M Abrahams
- Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT (V.M.A.)
| | - Bryony V Natale
- Obstetrics, Gynecology & Reproductive Science, University of California San Diego, La Jolla (B.V.N., D.R.N.).,Obstetrics and Gynaecology, School of Medicine, Queen's University, Ontario, Canada (B.V.N., D.R.N.)
| | - Alina P Montalbano
- Biochemistry and Obstetrics and Gynecology (A.P.M.), University of Texas Southwestern Medical Center, Dallas
| | - Xue Xiao
- Population and Data Sciences and Pediatrics (X.X., L.X.), University of Texas Southwestern Medical Center, Dallas
| | - Lin Xu
- Population and Data Sciences and Pediatrics (X.X., L.X.), University of Texas Southwestern Medical Center, Dallas
| | - David R Natale
- Obstetrics, Gynecology & Reproductive Science, University of California San Diego, La Jolla (B.V.N., D.R.N.).,Obstetrics and Gynaecology, School of Medicine, Queen's University, Ontario, Canada (B.V.N., D.R.N.)
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.).,Cell Biology (D.K.M., C.M.), University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
14
|
Woydziak ZR, Yucel AJ, Chamberlin AR. Tautomycetin Synthetic Analogues: Selective Inhibitors of Protein Phosphatase I. ChemMedChem 2021; 16:839-850. [PMID: 33301228 PMCID: PMC8582298 DOI: 10.1002/cmdc.202000801] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Indexed: 01/21/2023]
Abstract
Ser/Thr protein phosphatases (PPs) regulate a substantial range of cellular processes with protein phosphatases 1 (PP1) and 2 A (PP2A) accounting for over 90 % of the activity within cells. Nevertheless, tools to study PPs are limited as PPs inhibitors, particularly those selective for PP1 inhibition, are relatively scarce. Two examples of PP1-selective inhibitors, which share structural similarities, are tautomycin (TTM) and tautomycetin (TTN). This work describes the development of PP1/PP2A inhibitors that incorporate key structural features of TTM and TTN and are designed to conserve regions known to bind the active site of PP1/PP2A but vary regions that differentially contact the hydrophobic groove of PP1/PP2A. In all 28 TTN analogues were synthetically generated that inhibit PP1/PP2A activity at <250 mM; seven possessed inhibition activity at 100 nM. The IC50 values were determined for the seven most active analogues, which ranged from 34 to 1500 nM (PP1) and 70 to 6800 nM (PP2A). Four of the seven analogues possessed PP1 selectivity, and one demonstrated eightfold selectivity in the nanomolar range (PP1 IC50 =34 nM, PP2A IC50 =270 nM). A rationale is given for the observed differences in selectivity.
Collapse
Affiliation(s)
- Zachary R Woydziak
- Department of Physical and Life Sciences, Nevada State College, 1300, Nevada State Dr., Henderson, NV 89002, USA
| | - A John Yucel
- Department of Pharmaceutical Sciences, University of California, Irvine, 147 Biol. Sci. Admin., Irvine, CA 92697, USA
| | - A Richard Chamberlin
- Department of Pharmaceutical Sciences, University of California, Irvine, 147 Biol. Sci. Admin., Irvine, CA 92697, USA
| |
Collapse
|
15
|
Porzani SJ, Lima ST, Metcalf JS, Nowruzi B. In Vivo and In Vitro Toxicity Testing of Cyanobacterial Toxins: A Mini-Review. REVIEWS OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2021; 258:109-150. [PMID: 34622370 DOI: 10.1007/398_2021_74] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Harmful cyanobacterial blooms are increasing and becoming a worldwide concern as many bloom-forming cyanobacterial species can produce toxic metabolites named cyanotoxins. These include microcystins, saxitoxins, anatoxins, nodularins, and cylindrospermopsins, which can adversely affect humans, animals, and the environment. Different methods to assess these classes of compounds in vitro and in vivo include biological, biochemical, molecular, and physicochemical techniques. Furthermore, toxic effects not attributable to known cyanotoxins can be observed when assessing bloom material. In order to determine exposures to cyanotoxins and to monitor compliance with drinking and bathing water guidelines, it is necessary to have reliable and effective methods for the analysis of these compounds. Many relatively simple low-cost methods can be employed to rapidly evaluate the potential hazard. The main objective of this mini-review is to describe the assessment of toxic cyanobacterial samples using in vitro and in vivo bioassays. Newly emerging cyanotoxins, the toxicity of analogs, or the interaction of cyanobacteria and cyanotoxins with other toxicants, among others, still requires bioassay assessment. This review focuses on some biological and biochemical assays (MTT assay, Immunohistochemistry, Micronucleus Assay, Artemia salina assay, Daphnia magna test, Radionuclide recovery, Neutral red cytotoxicity and Comet assay, Enzyme-Linked Immunosorbent Assay (ELISA), Annexin V-FITC assay and Protein Phosphatase Inhibition Assay (PPIA)) for the detection and measurement of cyanotoxins including microcystins, cylindrospermopsins, anatoxin-a, saxitoxins, and nodularins. Although most bioassay analyses often confirm the presence of cyanotoxins at low concentrations, such bioassays can be used to determine whether some strains or blooms of cyanobacteria may produce other, as yet unknown toxic metabolites. This review also aims to identify research needs and data gaps concerning the toxicity assessment of cyanobacteria.
Collapse
Affiliation(s)
- Samaneh J Porzani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Stella T Lima
- Center for Nuclear Energy in Agriculture, University of Sao Paulo, Piracicaba, Brazil
| | | | - Bahareh Nowruzi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
16
|
Ahmed T, Van der Jeugd A, Caillierez R, Buée L, Blum D, D'Hooge R, Balschun D. Chronic Sodium Selenate Treatment Restores Deficits in Cognition and Synaptic Plasticity in a Murine Model of Tauopathy. Front Mol Neurosci 2020; 13:570223. [PMID: 33132838 PMCID: PMC7578417 DOI: 10.3389/fnmol.2020.570223] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 08/26/2020] [Indexed: 12/18/2022] Open
Abstract
A major goal in diseases is identifying a potential therapeutic agent that is cost-effective and can remedy some, if not all, disease symptoms. In Alzheimer’s disease (AD), aggregation of hyperphosphorylated tau protein is one of the neuropathological hallmarks, and Tau pathology correlates better with cognitive impairments in AD patients than amyloid-β load, supporting a key role of tau-related mechanisms. Selenium is a non-metallic trace element that is incorporated in the brain into selenoproteins. Chronic treatment with sodium selenate, a non-toxic selenium compound, was recently reported to rescue behavioral phenotypes in tau mouse models. Here, we focused on the effects of chronic selenate application on synaptic transmission and synaptic plasticity in THY-Tau22 mice, a transgenic animal model of tauopathies. Three months with a supplement of sodium selenate in the drinking water (12 μg/ml) restored not only impaired neurocognitive functions but also rescued long-term depression (LTD), a major form of synaptic plasticity. Furthermore, selenate reduced the inactive demethylated catalytic subunit of protein phosphatase 2A (PP2A) in THY-Tau22 without affecting total PP2A.Our study provides evidence that chronic dietary selenate rescues functional synaptic deficits of tauopathy and identifies activation of PP2A as the putative mechanism.
Collapse
Affiliation(s)
- Tariq Ahmed
- Brain and Cognition, Faculty of Psychology and Educational Sciences, KU Leuven, Leuven, Belgium.,Leuven Brain Institute, Leuven, Belgium
| | - Ann Van der Jeugd
- Leuven Brain Institute, Leuven, Belgium.,Laboratory of Biological Psychology, Brain and Cognition, Faculty of Psychology and Educational Sciences, KU Leuven, Leuven, Belgium
| | - Raphaëlle Caillierez
- Univ. Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Luc Buée
- Univ. Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - David Blum
- Univ. Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Rudi D'Hooge
- Leuven Brain Institute, Leuven, Belgium.,Laboratory of Biological Psychology, Brain and Cognition, Faculty of Psychology and Educational Sciences, KU Leuven, Leuven, Belgium
| | - Detlef Balschun
- Brain and Cognition, Faculty of Psychology and Educational Sciences, KU Leuven, Leuven, Belgium.,Leuven Brain Institute, Leuven, Belgium
| |
Collapse
|
17
|
Kang JB, Park DJ, Son HK, Koh PO. Decrease of protein phosphatase 2A subunit B by glutamate exposure in the cerebral cortex of neonatal rats. Lab Anim Res 2020; 36:34. [PMID: 32995332 PMCID: PMC7501672 DOI: 10.1186/s42826-020-00064-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/04/2020] [Indexed: 11/29/2022] Open
Abstract
Glutamate induces neurotoxicity during brain development, causing nerve damage. Protein phosphatase 2A (PP2A) is a type of serine/threonine phosphatase that regulates various biological functions. Among the PP2A subunit types, subunit B is abundant in brain tissue and plays an essential role in the nervous system. This study investigated changes in PP2A subunit B expression through glutamate exposure in the cerebral cortex of newborn rats. Sprague-Dawley rat pups (7 days after birth) were injected intraperitoneally with vehicle or glutamate (10 mg/kg). After 4 h of drug treatment, the brain tissue was isolated and fixed for morphological study. In addition, the cerebral cortex was collected for RNA and protein works. We observed severe histopathological changes including swollen neuron and atrophied dendrite in the glutamate exposed cerebral cortex. Glutamate exposure leads to a decrease in PP2A subunit B. Reverse-transcription PCR and Western blot analyses confirmed that glutamate induces a decrease of PP2A subunit B in the cerebral cortex of newborn rats. Moreover, immunohistochemical study showed a decrease in PP2A subunit B positive cells. The reduction of PP2A subunit B expression is considered an indicator of neurodegenerative damage. These results suggest that glutamate exposure causes neuronal damage in the cerebral cortex of new born rats through a decrease in PP2A subunit B.
Collapse
Affiliation(s)
- Ju-Bin Kang
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinju-daero, Jinju, 52828 South Korea
| | - Dong-Ju Park
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinju-daero, Jinju, 52828 South Korea
| | - Hyun-Kyoung Son
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinju-daero, Jinju, 52828 South Korea
| | - Phil-Ok Koh
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinju-daero, Jinju, 52828 South Korea
| |
Collapse
|
18
|
Baldini R, Mascaro M, Meroni G. The MID1 gene product in physiology and disease. Gene 2020; 747:144655. [PMID: 32283114 PMCID: PMC8011326 DOI: 10.1016/j.gene.2020.144655] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/22/2020] [Accepted: 04/06/2020] [Indexed: 12/23/2022]
Abstract
MID1 is an E3 ubiquitin ligase of the Tripartite Motif (TRIM) subfamily of RING-containing proteins, hence also known as TRIM18. MID1 is a microtubule-binding protein found in complex with the catalytic subunit of PP2A (PP2Ac) and its regulatory subunit alpha 4 (α4). To date, several substrates and interactors of MID1 have been described, providing evidence for the involvement of MID1 in a plethora of essential biological processes, especially during embryonic development. Mutations in the MID1 gene are responsible of the X-linked form of Opitz syndrome (XLOS), a multiple congenital disease characterised by defects in the development of midline structures during embryogenesis. Here, we review MID1-related physiological mechanisms as well as the pathological implication of the MID1 gene in XLOS and in other clinical conditions.
Collapse
Affiliation(s)
- Rossella Baldini
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Martina Mascaro
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Germana Meroni
- Department of Life Sciences, University of Trieste, Trieste, Italy.
| |
Collapse
|
19
|
Li X, Terunuma M, Deeb TG, Wiseman S, Pangalos MN, Nairn AC, Moss SJ, Slesinger PA. Direct Interaction of PP2A Phosphatase with GABA B Receptors Alters Functional Signaling. J Neurosci 2020; 40:2808-2816. [PMID: 32111696 PMCID: PMC7117905 DOI: 10.1523/jneurosci.2654-19.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/02/2020] [Accepted: 02/18/2020] [Indexed: 01/16/2023] Open
Abstract
Addictive drugs usurp the brain's intrinsic mechanism for reward, leading to compulsive and destructive behaviors. In the ventral tegmental area (VTA), the center of the brain's reward circuit, GABAergic neurons control the excitability of dopamine (DA) projection neurons and are the site of initial psychostimulant-dependent changes in signaling. Previous work established that cocaine/methamphetamine exposure increases protein phosphatase 2A (PP2A) activity, which dephosphorylates the GABABR2 subunit, promotes internalization of the GABAB receptor (GABABR) and leads to smaller GABABR-activated G-protein-gated inwardly rectifying potassium (GIRK) currents in VTA GABA neurons. How the actions of PP2A become selective for a particular signaling pathway is poorly understood. Here, we demonstrate that PP2A can associate directly with a short peptide sequence in the C terminal domain of the GABABR1 subunit, and that GABABRs and PP2A are in close proximity in rodent neurons (mouse/rat; mixed sexes). We show that this PP2A-GABABR interaction can be regulated by intracellular Ca2+ Finally, a peptide that potentially reduces recruitment of PP2A to GABABRs and thereby limits receptor dephosphorylation increases the magnitude of baclofen-induced GIRK currents. Thus, limiting PP2A-dependent dephosphorylation of GABABRs may be a useful strategy to increase receptor signaling for treating diseases.SIGNIFICANCE STATEMENT Dysregulation of GABAB receptors (GABABRs) underlies altered neurotransmission in many neurological disorders. Protein phosphatase 2A (PP2A) is involved in dephosphorylating and subsequent internalization of GABABRs in models of addiction and depression. Here, we provide new evidence that PP2A B55 regulatory subunit interacts directly with a small region of the C-terminal domain of the GABABR1 subunit, and that this interaction is sensitive to intracellular Ca2+ We demonstrate that a short peptide corresponding to the PP2A interaction site on GABABR1 competes for PP2A binding, enhances phosphorylation GABABR2 S783, and affects functional signaling through GIRK channels. Our study highlights how targeting PP2A dependent dephosphorylation of GABABRs may provide a specific strategy to modulate GABABR signaling in disease conditions.
Collapse
Affiliation(s)
- Xiaofan Li
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Miho Terunuma
- Division of Oral Biochemistry, Graduate School of Medical and Dental Sciences, Niigata University, 951-8514 Japan
| | - Tarek G Deeb
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Shari Wiseman
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | | | - Angus C Nairn
- Department Psychiatry, Yale University School of Medicine, New Haven, Connecticut 065019
| | - Stephen J Moss
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111,
- Department of Physiology, Pharmacology and Neuroscience, University College, London WC1E 6BT, United Kingdom
| | - Paul A Slesinger
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029,
| |
Collapse
|
20
|
Moens U, Macdonald A. Effect of the Large and Small T-Antigens of Human Polyomaviruses on Signaling Pathways. Int J Mol Sci 2019; 20:ijms20163914. [PMID: 31408949 PMCID: PMC6720190 DOI: 10.3390/ijms20163914] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/09/2019] [Accepted: 08/10/2019] [Indexed: 12/12/2022] Open
Abstract
Viruses are intracellular parasites that require a permissive host cell to express the viral genome and to produce new progeny virus particles. However, not all viral infections are productive and some viruses can induce carcinogenesis. Irrespective of the type of infection (productive or neoplastic), viruses hijack the host cell machinery to permit optimal viral replication or to transform the infected cell into a tumor cell. One mechanism viruses employ to reprogram the host cell is through interference with signaling pathways. Polyomaviruses are naked, double-stranded DNA viruses whose genome encodes the regulatory proteins large T-antigen and small t-antigen, and structural proteins that form the capsid. The large T-antigens and small t-antigens can interfere with several host signaling pathways. In this case, we review the interplay between the large T-antigens and small t-antigens with host signaling pathways and the biological consequences of these interactions.
Collapse
Affiliation(s)
- Ugo Moens
- Molecular Inflammation Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, 9019 Tromsø, Norway.
| | - Andrew Macdonald
- School of Molecular and Cellular Biology, Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
21
|
Park DJ, Kang JB, Shah MA, Koh PO. Quercetin alleviates the injury-induced decrease of protein phosphatase 2A subunit B in cerebral ischemic animal model and glutamate-exposed HT22 cells. J Vet Med Sci 2019; 81:1047-1054. [PMID: 31092742 PMCID: PMC6656806 DOI: 10.1292/jvms.19-0094] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Quercetin is a plant flavonoid that has anti-oxidant, anti-inflammatory, anti-cancer, and anti-ischemic properties. Moreover, quercetin exerts neuroprotective effects against focal cerebral
ischemia. Protein phosphatase 2A (PP2A) is a form of serine/threonine phosphatase that modulates various biological functions. Among PP2A subunit types, subunit B exists abundantly in brain
tissue and plays an essential function in nervous system. We previously reported the decrease of PP2A subunit B in focal cerebral animal model. This study explored the change of PP2A subunit
B expression by quercetin treatment in cerebral ischemic animal model and glutamate-treated hippocampal-derived (HT22) cell culture. Quercetin (10 mg/kg) or vehicle was injected
intraperitoneally into male rats before 30 min of middle cerebral artery occlusion (MCAO), and cerebral cortices were isolated 24 hr after MCAO. MCAO induced the neurological behavioral
deficit and increased infarct volume. However, quercetin treatment attenuated the increase of neurological deficit and infarction. We detected the alleviation of MCAO-induced the decrease in
PP2A subunit B by quercetin treatment using a proteomic approach. Reverse-transcription PCR and Western blot analyses confirmed lower PP2A subunit B expression levels in MCAO group with
vehicle. However, quercetin treatment attenuated MCAO-induced this reduction. We also observed the neuroprotective effect of quercetin and the change of PP2A subunit B expression in
glutamate-exposed HT22 cells. Glutamate exposure dramatically reduced cell viability and PP2A subunit B expression, and quercetin treatment significantly improved these decreases. We clearly
showed that quercetin performs a neuroprotective function and modulates down-regulation of PP2A subunit B against MCAO injury and glutamate toxicity. Thus, our finding suggests that the
regulation of PP2A subunit B by quercetin contributes to neuroprotective function in ischemic brain injury.
Collapse
Affiliation(s)
- Dong-Ju Park
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinjudaero, Jinju, 52828, South Korea
| | - Ju-Bin Kang
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinjudaero, Jinju, 52828, South Korea
| | - Murad-Ali Shah
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinjudaero, Jinju, 52828, South Korea
| | - Phil-Ok Koh
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinjudaero, Jinju, 52828, South Korea
| |
Collapse
|
22
|
Ding Y, Yu A, Tsokos GC, Malek TR. CD25 and Protein Phosphatase 2A Cooperate to Enhance IL-2R Signaling in Human Regulatory T Cells. THE JOURNAL OF IMMUNOLOGY 2019; 203:93-104. [PMID: 31085588 DOI: 10.4049/jimmunol.1801570] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 04/20/2019] [Indexed: 11/19/2022]
Abstract
Low-dose IL-2 therapy is a direct approach to boost regulatory T cells (Tregs) and promote immune tolerance in autoimmune patients. However, the mechanisms responsible for selective response of Tregs to low-dose IL-2 is not fully understood. In this study we directly assessed the contribution of CD25 and protein phosphatase 2A (PP2A) in promoting IL-2R signaling in Tregs. IL-2-induced tyrosine phosphorylation of STAT5 (pSTAT5) was proportional to CD25 levels on human CD4+ T cells and YT human NK cell line, directly demonstrating that CD25 promotes IL-2R signaling. Overexpression of the PP2A catalytic subunit (PP2Ac) by lentiviral transduction in human Tregs increased the level of IL-2R subunits and promoted tyrosine phosphorylation of Jak3 and STAT5. Interestingly, increased expression of CD25 only partially accounted for this enhanced activation of pSTAT5, indicating that PP2A promotes IL-2R signaling through multiple mechanisms. Consistent with these findings, knockdown of PP2Ac in human Tregs and impaired PP2Ac activity in mouse Tregs significantly reduced IL-2-dependent STAT5 activation. In contrast, overexpression or knockdown of PP2Ac in human T effector cells did not affect IL-2-dependent pSTAT5 activation. Overexpression of PP2Ac in human Tregs also increased the expressions of proteins related to survival, activation, and immunosuppressive function, and upregulated several IL-2-regulated genes. Collectively, these findings suggest that CD25 and PP2A cooperatively enhance the responsiveness of Tregs to IL-2, which provide potential therapeutic targets for low-dose IL-2 therapy.
Collapse
Affiliation(s)
- Ying Ding
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - Aixin Yu
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215; and
| | - Thomas R Malek
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136; .,Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33136
| |
Collapse
|
23
|
Schuhmacher D, Sontag JM, Sontag E. Protein Phosphatase 2A: More Than a Passenger in the Regulation of Epithelial Cell-Cell Junctions. Front Cell Dev Biol 2019; 7:30. [PMID: 30895176 PMCID: PMC6414416 DOI: 10.3389/fcell.2019.00030] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/22/2019] [Indexed: 12/17/2022] Open
Abstract
Cell–cell adhesion plays a key role in the maintenance of the epithelial barrier and apicobasal cell polarity, which is crucial for homeostasis. Disruption of cell–cell adhesion is a hallmark of numerous pathological conditions, including invasive carcinomas. Adhesion between apposing cells is primarily regulated by three types of junctional structures: desmosomes, adherens junctions, and tight junctions. Cell junctional structures are highly regulated multiprotein complexes that also serve as signaling platforms to control epithelial cell function. The biogenesis, integrity, and stability of cell junctions is controlled by complex regulatory interactions with cytoskeletal and polarity proteins, as well as modulation of key component proteins by phosphorylation/dephosphorylation processes. Not surprisingly, many essential signaling molecules, including protein Ser/Thr phosphatase 2A (PP2A) are associated with intercellular junctions. Here, we examine how major PP2A enzymes regulate epithelial cell–cell junctions, either directly by associating with and dephosphorylating component proteins, or indirectly by affecting signaling pathways that control junctional integrity and cytoskeletal dynamics. PP2A deregulation has severe consequences on the stability and functionality of these structures, and disruption of cell–cell adhesion and cell polarity likely contribute to the link between PP2A dysfunction and human carcinomas.
Collapse
Affiliation(s)
- Diana Schuhmacher
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
| | - Jean-Marie Sontag
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Estelle Sontag
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
24
|
The Anti-Tumor Agent Sodium Selenate Decreases Methylated PP2A, Increases GSK3βY216 Phosphorylation, Including Tau Disease Epitopes and Reduces Neuronal Excitability in SHSY-5Y Neurons. Int J Mol Sci 2019; 20:ijms20040844. [PMID: 30781361 PMCID: PMC6412488 DOI: 10.3390/ijms20040844] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 02/12/2019] [Accepted: 02/12/2019] [Indexed: 12/20/2022] Open
Abstract
Selenium application as sodium selenate was repeatedly shown to have anti-carcinogenic properties by increasing levels of the serine/ threonine protein phosphatase 2A (PP2A) in cancer cells. PP2A has a prominent role in cell development, homeostasis, and in neurons regulates excitability. PP2A, GSK3β and Tau reside together in a complex, which facilitates their interaction and (dys)-function as has been reported for several neurological disorders. In this study we recorded maximum increase in total PP2A at 3 µM sodium selenate in a neuron cell line. In conjunction with these data, whole-cell electrophysiological studies revealed that this concentration had maximum effect on membrane potentials, conductance and currents. Somewhat surprisingly, the catalytically active form, methylated PP2A (mePP2A) was significantly decreased. In close correlation to these data, the phosphorylation state of two substrate proteins, sensitive to PP2A activity, GSK3β and Tau were found to be increased. In summary, our data reveal that sodium selenate enhances PP2A levels, but reduces catalytic activity of PP2A in a dose dependent manner, which fails to reduce Tau and GSK3β phosphorylation under physiological conditions, indicating an alternative route in the rescue of cell pathology in neurological disorders.
Collapse
|
25
|
Ten Broeke T, Honing H, Brandsma AM, Jacobino S, Bakema JE, Kanters D, van der Linden JAM, Bracke M, Koenderman L, Leusen JHW. FcαRI Dynamics Are Regulated by GSK-3 and PKCζ During Cytokine Mediated Inside-Out Signaling. Front Immunol 2019; 9:3191. [PMID: 30766540 PMCID: PMC6365424 DOI: 10.3389/fimmu.2018.03191] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 12/31/2018] [Indexed: 12/03/2022] Open
Abstract
IgA binding to FcαRI (CD89) is rapidly enhanced by cytokine induced inside-out signaling. Dephosphorylation of serine 263 in the intracellular tail of FcαRI by PP2A and PI3K activation are instrumental in this process. To further investigate these signaling pathways, we targeted downstream kinases of PI3K. Our experiments revealed that PI3K activates PKCζ, which subsequently inhibits GSK-3, a constitutively active kinase in resting cells and found here to be associated with FcαRI. We propose that GSK-3 maintains FcαRI in an inactive state at homeostatic conditions. Upon cytokine stimulation, GSK-3 is inactivated through a PI3K-PKCζ pathway, preventing the maintenance of phosphorylated inactive FcαRI. The concomitantly activated PP2A is then able to dephosphorylate and activate FcαRI. Moreover, FRAP and FLIP studies showed that FcαRI activation coincides with an increased mobile fraction of the receptor. This can enhance FcαRI valency and contribute to stronger avidity for IgA immune complexes. This tightly regulated inside-out signaling pathway allows leukocytes to respond rapidly and efficiently to their environment and could be exploited to enhance the efficacy of future IgA therapeutics.
Collapse
Affiliation(s)
- Toine Ten Broeke
- Laboratory of Translational Immunology, University Medical Center, Utrecht, Netherlands
| | - Henk Honing
- Department of Respiratory Medicine, University Medical Center, Utrecht, Netherlands
| | - Arianne M Brandsma
- Laboratory of Translational Immunology, University Medical Center, Utrecht, Netherlands
| | - Shamir Jacobino
- Laboratory of Translational Immunology, University Medical Center, Utrecht, Netherlands
| | - Jantine E Bakema
- Laboratory of Translational Immunology, University Medical Center, Utrecht, Netherlands.,Tumor Biology Section, Department of Otolaryngology, Head-Neck Surgery, VU University Medical Center, Amsterdam, Netherlands
| | - Deon Kanters
- Laboratory of Translational Immunology, University Medical Center, Utrecht, Netherlands.,Department of Respiratory Medicine, University Medical Center, Utrecht, Netherlands
| | - Jan A M van der Linden
- Laboratory of Translational Immunology, University Medical Center, Utrecht, Netherlands.,Department of Respiratory Medicine, University Medical Center, Utrecht, Netherlands
| | - Madelon Bracke
- Department of Pharmacoepidemiology and Pharmacotherapy, University Medical Center, Utrecht, Netherlands
| | - Leo Koenderman
- Laboratory of Translational Immunology, University Medical Center, Utrecht, Netherlands.,Department of Respiratory Medicine, University Medical Center, Utrecht, Netherlands
| | - Jeanette H W Leusen
- Laboratory of Translational Immunology, University Medical Center, Utrecht, Netherlands
| |
Collapse
|
26
|
Architecture, substructures, and dynamic assembly of STRIPAK complexes in Hippo signaling. Cell Discov 2019; 5:3. [PMID: 30622739 PMCID: PMC6323126 DOI: 10.1038/s41421-018-0077-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/08/2018] [Accepted: 12/10/2018] [Indexed: 01/24/2023] Open
Abstract
Striatin-interacting phosphatases and kinases (STRIPAKs) are evolutionarily conserved supramolecular complexes, which have been implicated in the Hippo signaling pathway. Yet the topological structure and dynamic assembly of STRIPAK complexes remain elusive. Here, we report the overall architecture and substructures of a Hippo kinase-containing STRIPAK complex. PP2Aa/c-bound STRN3 directly contacts the Hippo kinase MST2 and also controls the loading of MST2 via two “arms” in a phosphorylation-dependent manner, one arm being STRIP1 and the other SIKE1-SLMAP. A decreased cell density triggered the dissociation of the STRIP1 arm from STRIPAK, reflecting the dynamic assembly of the complex upon sensing upstream signals. Crystallographic studies defined at atomic resolution the interface between STRN3 and SIKE1, and that between SIKE1 and SLMAP. Disrupting the complex assembly abrogated the regulatory effect of STRIPAK towards Hippo signaling. Collectively, our study revealed a “two-arm” assembly of STRIPAK with context-dependent dynamics, offering a framework for further studies on Hippo signaling and biological processes involving MST kinases.
Collapse
|
27
|
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
|
28
|
Gupta MK, Mohan ML, Naga Prasad SV. G Protein-Coupled Receptor Resensitization Paradigms. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 339:63-91. [PMID: 29776605 DOI: 10.1016/bs.ircmb.2018.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cellular responses to extracellular milieu/environment are driven by cell surface receptors that transmit the signal into the cells resulting in a synchronized and measured response. The ability to provide such exquisite responses to changes in external environment is mediated by the tight and yet, deliberate regulation of cell surface receptor function. In this regard, the seven transmembrane G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors that regulate responses like cardiac contractility, vision, and olfaction including platelet activation. GPCRs regulate these plethora of events through GPCR-activation, -desensitization, and -resensitization. External stimuli (ligands or agonists) activate GPCR initiating downstream signals. The activated GPCR undergoes inactivation or desensitization by phosphorylation and binding of β-arrestin resulting in diminution of downstream signals. The desensitized GPCRs are internalized into endosomes, wherein they undergo dephosphorylation or resensitization by protein phosphatase to be recycled back to the cell membrane as naïve GPCR ready for the next wave of stimuli. Despite the knowledge that activation, desensitization, and resensitization shoulder an equal role in maintaining GPCR function, major advances have been made in understanding activation and desensitization compared to resensitization. However, increasing evidence shows that resensitization is exquisitely regulated process, thereby contributing to the dynamic regulation of GPCR function. In recognition of these observations, in this chapter we discuss the key advances on the mechanistic underpinning that drive and regulate GPCR function with a focus on resensitization.
Collapse
Affiliation(s)
- Manveen K Gupta
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Maradumane L Mohan
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Sathyamangla V Naga Prasad
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States.
| |
Collapse
|
29
|
Yu S, Li L, Wu Q, Dou N, Li Y, Gao Y. PPP2R2D, a regulatory subunit of protein phosphatase 2A, promotes gastric cancer growth and metastasis via mechanistic target of rapamycin activation. Int J Oncol 2018; 52:2011-2020. [PMID: 29568966 DOI: 10.3892/ijo.2018.4329] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 03/15/2018] [Indexed: 11/06/2022] Open
Abstract
Protein phosphatase 2A (PP2A) is an essential serine/threonine protein phosphatase that regulates the basic activities of eukaryotes by dephosphorylating its substrates. The function and substrate specificity of PP2A are generally determined by its regulatory subunits. In the present study, the clinical significance and roles of PPP2R2D, one of the regulatory subunits of PP2A, were demonstrated in gastric cancer (GC) carcinogenesis. Through a tissue microarray and quantitative polymerase chain reaction analysis, it was demonstrated that PPP2R2D was commonly upregulated in GC samples. This upregulation was positively correlated with the patients' tumor stage (P<0.01), T classification (P<0.01) and N classification (P=0.01). Furthermore, a high expression of PPP2R2D was closely associated with poor prognosis of patients. Knockdown of PPP2R2D significantly inhibited the proliferation and migration of GC cells in vitro, as well as the tumorigenicity and metastasis in vivo in an animal GC model. By contrast, overexpression of PPP2R2D promoted GC cell proliferation and migration in vitro. The analysis of underlying mechanisms indicated that PPP2R2D silencing decreased the phosphorylation level of mechanistic target of rapamycin (mTOR), thereby implicating that PPP2R2D is involved in the regulation of mTOR activity during tumorigenesis. Thus, the findings of the present study suggested that PPP2R2D may serve as a potential oncogene in GC and as a novel target for therapeutic strategies against this disease.
Collapse
Affiliation(s)
- Shijun Yu
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Li Li
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Qiong Wu
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Ning Dou
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Yandong Li
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Yong Gao
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| |
Collapse
|
30
|
Antiphospholipid antibodies induce thrombosis by PP2A activation via apoER2-Dab2-SHC1 complex formation in endothelium. Blood 2018; 131:2097-2110. [PMID: 29500169 DOI: 10.1182/blood-2017-11-814681] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 02/23/2018] [Indexed: 01/10/2023] Open
Abstract
In the antiphospholipid syndrome (APS), antiphospholipid antibody (aPL) recognition of β2 glycoprotein I promotes thrombosis, and preclinical studies indicate that this is due to endothelial nitric oxide synthase (eNOS) antagonism via apolipoprotein E receptor 2 (apoER2)-dependent processes. How apoER2 molecularly links these events is unknown. Here, we show that, in endothelial cells, the apoER2 cytoplasmic tail serves as a scaffold for aPL-induced assembly and activation of the heterotrimeric protein phosphatase 2A (PP2A). Disabled-2 (Dab2) recruitment to the apoER2 NPXY motif promotes the activating L309 methylation of the PP2A catalytic subunit by leucine methyl transferase-1. Concurrently, Src homology domain-containing transforming protein 1 (SHC1) recruits the PP2A scaffolding subunit to the proline-rich apoER2 C terminus along with 2 distinct regulatory PP2A subunits that mediate inhibitory dephosphorylation of Akt and eNOS. In mice, the coupling of these processes in endothelium is demonstrated to underlie aPL-invoked thrombosis. By elucidating these intricacies in the pathogenesis of APS-related thrombosis, numerous potential new therapeutic targets have been identified.
Collapse
|
31
|
Janghorban M, Langer EM, Wang X, Zachman D, Daniel CJ, Hooper J, Fleming WH, Agarwal A, Sears RC. The tumor suppressor phosphatase PP2A-B56α regulates stemness and promotes the initiation of malignancies in a novel murine model. PLoS One 2017; 12:e0188910. [PMID: 29190822 PMCID: PMC5708644 DOI: 10.1371/journal.pone.0188910] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 11/15/2017] [Indexed: 01/13/2023] Open
Abstract
Protein phosphatase 2A (PP2A) is a ubiquitously expressed Serine-Threonine phosphatase mediating 30–50% of protein phosphatase activity. PP2A functions as a heterotrimeric complex, with the B subunits directing target specificity to regulate the activity of many key pathways that control cellular phenotypes. PP2A-B56α has been shown to play a tumor suppressor role and to negatively control c-MYC stability and activity. Loss of B56α promotes cellular transformation, likely at least in part through its regulation of c-MYC. Here we report generation of a B56α hypomorph mouse with very low B56α expression that we used to study the physiologic activity of the PP2A-B56α phosphatase. The predominant phenotype we observed in mice with B56α deficiency in the whole body was spontaneous skin lesion formation with hyperproliferation of the epidermis, hair follicles and sebaceous glands. Increased levels of c-MYC phosphorylation on Serine62 and c-MYC activity were observed in the skin lesions of the B56αhm/hm mice. B56α deficiency was found to increase the number of skin stem cells, and consistent with this, papilloma initiation was accelerated in a carcinogenesis model. Further analysis of additional tissues revealed increased inflammation in spleen, liver, lung, and intestinal lymph nodes as well as in the skin lesions, resembling elevated extramedullary hematopoiesis phenotypes in the B56αhm/hm mice. We also observed an increase in the clonogenicity of bone marrow stem cells in B56αhm/hm mice. Overall, this model suggests that B56α is important for stem cells to maintain homeostasis and that B56α loss leading to increased activity of important oncogenes, including c-MYC, can result in aberrant cell growth and increased stem cells that can contribute to the initiation of malignancy.
Collapse
Affiliation(s)
- Mahnaz Janghorban
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Ellen M. Langer
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Xiaoyan Wang
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Derek Zachman
- Papé Family Pediatric Research Institute, Oregon Stem Cell Center, Department of Pediatrics, Portland, Oregon, United States of America
| | - Colin J. Daniel
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Jody Hooper
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - William H. Fleming
- Papé Family Pediatric Research Institute, Oregon Stem Cell Center, Department of Pediatrics, Portland, Oregon, United States of America
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Anupriya Agarwal
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Division of Hematology & Medical Oncology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Rosalie C. Sears
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, United States of America
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
32
|
Di Conza G, Trusso Cafarello S, Zheng X, Zhang Q, Mazzone M. PHD2 Targeting Overcomes Breast Cancer Cell Death upon Glucose Starvation in a PP2A/B55α-Mediated Manner. Cell Rep 2017; 18:2836-2844. [PMID: 28329677 PMCID: PMC5378984 DOI: 10.1016/j.celrep.2017.02.081] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 12/21/2016] [Accepted: 02/28/2017] [Indexed: 01/07/2023] Open
Abstract
B55α is a regulatory subunit of the PP2A phosphatase. We have recently found that B55α-associated PP2A promotes partial deactivation of the HIF-prolyl-hydroxylase enzyme PHD2. Here, we show that, in turn, PHD2 triggers degradation of B55α by hydroxylating it at proline 319. In the context of glucose starvation, PHD2 reduces B55α protein levels, which correlates with MDA-MB231 and MCF7 breast cancer cell death. Under these conditions, PHD2 silencing rescues B55α degradation, overcoming apoptosis, whereas in SKBR3 breast cancer cells showing resistance to glucose starvation, B55α knockdown restores cell death and prevents neoplastic growth in vitro. Treatment of MDA-MB231-derived xenografts with the glucose competitor 2-deoxy-glucose leads to tumor regression in the presence of PHD2. Knockdown of PHD2 induces B55α accumulation and treatment resistance by preventing cell apoptosis. Overall, our data unravel B55α as a PHD2 substrate and highlight a role for PHD2-B55α in the response to nutrient deprivation.
Collapse
Affiliation(s)
- Giusy Di Conza
- Laboratory of Tumor Inflammation and Angiogenesis, Vesalius Research Center, VIB, 3000 Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, 3000 Leuven, Belgium.
| | - Sarah Trusso Cafarello
- Laboratory of Tumor Inflammation and Angiogenesis, Vesalius Research Center, VIB, 3000 Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Xingnan Zheng
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 450 West Drive, Chapel Hill, NC 27599, USA
| | - Qing Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 450 West Drive, Chapel Hill, NC 27599, USA
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Vesalius Research Center, VIB, 3000 Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
33
|
Taleski G, Sontag E. Protein phosphatase 2A and tau: an orchestrated 'Pas de Deux'. FEBS Lett 2017; 592:1079-1095. [PMID: 29121398 DOI: 10.1002/1873-3468.12907] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 10/18/2017] [Accepted: 11/02/2017] [Indexed: 12/21/2022]
Abstract
The neuronal microtubule-associated protein tau serves a critical role in regulating axonal microtubule dynamics to support neuronal and synaptic functions. Furthermore, it contributes to glutamatergic regulation and synaptic plasticity. Emerging evidence also suggests that tau serves as a signaling scaffold. Tau function and subcellular localization are tightly regulated, in part, by the orchestrated interplay between phosphorylation and dephosphorylation events. Significantly, protein phosphatase type 2A (PP2A), encompassing the regulatory PPP2R2A (or Bα) subunit, is a major brain heterotrimeric enzyme and the primary tau Ser/Thr phosphatase in vivo. Herein, we closely examine how the intimate and compartmentalized interactions between PP2A and tau regulate tau phosphorylation and function, and play an essential role in neuronal homeostasis. We also review evidence supporting a strong link between deregulation of tau-PP2A functional interactions and the molecular underpinnings of various neurodegenerative diseases collectively called tauopathies. Lastly, we discuss the opportunities and associated challenges in more specifically targeting PP2A-tau interactions for drug development for tauopathies.
Collapse
Affiliation(s)
- Goce Taleski
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, Hunter Medical Research Institute, University of Newcastle, Callaghan, Australia
| | - Estelle Sontag
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, Hunter Medical Research Institute, University of Newcastle, Callaghan, Australia
| |
Collapse
|
34
|
Hayashi T, Hikichi M, Yukitake J, Harada N, Utsumi T. Estradiol suppresses phosphorylation of ERα serine 167 through upregulation of PP2A in breast cancer cells. Oncol Lett 2017; 14:8060-8065. [PMID: 29344249 DOI: 10.3892/ol.2017.7216] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 09/07/2017] [Indexed: 12/23/2022] Open
Abstract
Aromatase inhibitors (AIs) are effective endocrine therapeutics for postmenopausal women with estrogen receptor (ER)α-positive breast cancer. However, the efficacy of the treatment is often limited by the onset of AI resistance, owing to the phosphorylation of ERα serine 167 (Ser167). Previous studies have indicated that hyperactivation of the phosphoinositide-3 kinase/RAC serine/threonine-protein kinase signaling pathway occurs in AI-resistant breast cancer models, which coincides with elevated levels of ERα phosphorylation at Ser167. The tumor suppressor serine/threonine-protein phosphatase 2A (PP2A) regulates the phosphatidylinositol 3-kinase/RAC serine/threonine-protein kinase signaling pathway. A previous study indicated that PP2A inhibition decreased ERα Ser167 phosphorylation and estradiol (E2)-independent cell growth. The present study investigated the potential relevance of PP2A in E2 deprivation-resistant MCF-7 cells. E2 depletion reduced the susceptibility of MCF-7 cells to inhibitors of mechanistic target of rapamycin (mTOR) and significantly increased ERα Ser167 phosphorylation and decreased expression of PP2A. Conversely, long-term E2-deprived (LTED) MCF-7 cells, a model of AI-resistant breast cancer, exhibited decreased ERα Ser167 phosphorylation and further upregulation of PP2A in E2-containing medium. The PP2A activator forskolin (FSK) significantly inhibited LTED cell proliferation by increasing the effect of everolimus (Eve), an mTOR inhibitor. In summary, the present study provides further evidence that PP2A represents a therapeutic target for AI-resistant breast cancer.
Collapse
Affiliation(s)
- Takanori Hayashi
- Department of Biochemistry, School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Masahiro Hikichi
- Department of Breast Surgery, School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Jun Yukitake
- Department of Clinical Immunology, School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Nobuhiro Harada
- Department of Biochemistry, School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Toshiaki Utsumi
- Department of Breast Surgery, School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| |
Collapse
|
35
|
Yeh PA, Chang CJ. A novel function of twins, B subunit of protein phosphatase 2A, in regulating actin polymerization. PLoS One 2017; 12:e0186037. [PMID: 28977036 PMCID: PMC5627941 DOI: 10.1371/journal.pone.0186037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 09/22/2017] [Indexed: 11/18/2022] Open
Abstract
Actin is an important component of the cytoskeleton and its polymerization is delicately regulated by several kinases and phosphatases. Heterotrimeric protein phosphatase 2A (PP2A) is a potent phosphatase that is crucial for cell proliferation, apoptosis, tumorigenesis, signal transduction, cytoskeleton arrangement, and neurodegeneration. To facilitate these varied functions, different regulators determine the different targets of PP2A. Among these regulators of PP2A, the B subunits in particular may be involved in cytoskeleton arrangement. However, little is known about the role of PP2A in actin polymerization in vivo. Using sophisticated fly genetics, we demonstrated a novel function for the fly B subunit, twins, to promote actin polymerization in varied tissue types, suggesting a broad and conserved effect. Furthermore, our genetic data suggest that twins may act upstream of the actin-polymerized-proteins, Moesin and Myosin-light-chain, and downstream of Rho to promote actin polymerization. This work opens a new avenue for exploring the biological functions of a PP2A regulator, twins, in cytoskeleton regulation.
Collapse
Affiliation(s)
- Po-An Yeh
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Chung Li, Taiwan
- * E-mail:
| | - Ching-Jin Chang
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei, Taiwan
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei, Taiwan
| |
Collapse
|
36
|
Mohan ML, Chatterjee A, Ganapathy S, Mukherjee S, Srikanthan S, Jolly GP, Anand RS, Naga Prasad SV. Noncanonical regulation of insulin-mediated ERK activation by phosphoinositide 3-kinase γ. Mol Biol Cell 2017; 28:3112-3122. [PMID: 28877982 PMCID: PMC5662266 DOI: 10.1091/mbc.e16-12-0864] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 08/23/2017] [Accepted: 08/31/2017] [Indexed: 12/17/2022] Open
Abstract
Classically, Class IB phosphoinositide 3-kinase (PI3Kγ) plays a role in ERK activation following G-protein–coupled receptor (GPCR) activation. Here we show that PI3Kγ noncanonically regulates ERK phosphorylation in a kinase-independent mechanism, irrespective of the upstream signals. PI3Kγ sequesters PP2A, allowing sustained ERK function. Classically Class IB phosphoinositide 3-kinase (PI3Kγ) plays a role in extracellular signal–regulated kinase (ERK) activation following G-protein coupled receptor (GPCR) activation. Knock-down of PI3Kγ unexpectedly resulted in loss of ERK activation to receptor tyrosine kinase agonists such as epidermal growth factor or insulin. Mouse embryonic fibroblasts (MEFs) or primary adult cardiac fibroblasts isolated from PI3Kγ knock-out mice (PI3KγKO) showed decreased insulin-stimulated ERK activation. However, expression of kinase-dead PI3Kγ resulted in rescue of insulin-stimulated ERK activation. Mechanistically, PI3Kγ sequesters protein phosphatase 2A (PP2A), disrupting ERK–PP2A interaction, as evidenced by increased ERK–PP2A interaction and associated PP2A activity in PI3KγKO MEFs, resulting in decreased ERK activation. Furthermore, β-blocker carvedilol-mediated β-arrestin-dependent ERK activation is significantly reduced in PI3KγKO MEF, suggesting accelerated dephosphorylation. Thus, instead of classically mediating the kinase arm, PI3Kγ inhibits PP2A by scaffolding and sequestering, playing a key parallel synergistic step in sustaining the function of ERK, a nodal enzyme in multiple cellular processes.
Collapse
Affiliation(s)
- Maradumane L Mohan
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Arunachal Chatterjee
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Swetha Ganapathy
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Sromona Mukherjee
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Sowmya Srikanthan
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - George P Jolly
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Rohit S Anand
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Sathyamangla V Naga Prasad
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| |
Collapse
|
37
|
Park DJ, Koh PO. Hyperglycemia reduces PP2A subunit B expression in a middle cerebral artery occlusion animal model. J Vet Med Sci 2017; 79:1342-1347. [PMID: 28652527 PMCID: PMC5573819 DOI: 10.1292/jvms.17-0135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Diabetes is a metabolic disorder that worsens clinical outcome following cerebral
ischemia. Protein phosphatase 2A (PP2A) is a conserved, heterotrimeric, serine/threonine
phosphatase with various cellular functions. PP2A subunit B is abundant in brain tissue
and modulates PP2A function. The aim of this study was to investigate PP2A subunit B
protein expression in the cerebral cortex of non-diabetic and diabetic animals with middle
cerebral artery occlusion (MCAO) injury. Sprague-Dawley rats were injected with
streptozotocin (40 mg/kg, i.p.) to induce diabetic conditions. After 4 weeks of
streptozotocin treatment, the rats underwent MCAO to induce focal cerebral ischemia. The
cerebral cortex tissue was collected 24 hr after MCAO. Body weight and blood glucose were
measured, and Western blot analysis was performed to elucidate the expression of PP2A
subunit B. We confirmed decreased body weight and increased blood glucose in diabetic
animals. Reverse transcription-PCR and Western blot analyses showed decreased PP2A subunit
B expression in the cerebral cortices of MCAO-injured animals. Moreover, diabetic animals
with MCAO showed more severe decreases in PP2A subunit B protein levels than non-diabetic
animals following MCAO. The decline of PP2A subunit B indicates degradation of neuronal
function. These findings suggest that conspicuous decreases in PP2A subunit B may
exacerbate cerebral ischemia under diabetic conditions following MCAO.
Collapse
Affiliation(s)
- Dong-Ju Park
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 900 Gajwa-dong, Jinju 660-701, South Korea
| | - Phil-Ok Koh
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 900 Gajwa-dong, Jinju 660-701, South Korea
| |
Collapse
|
38
|
Wang Y, Zhou Z, Diao Y, Strappe P, Blanchard C. The potential role of p53 and MAPK pathways in the hepatotoxicity of deep‐fried oil and in resistant starch‐induced protection. EUR J LIPID SCI TECH 2017. [DOI: 10.1002/ejlt.201600296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Yuyang Wang
- Key Laboratory of Food Nutrition and Safety, Ministry of EducationTianjin University of Science and TechnologyTianjinP. R. China
| | - Zhongkai Zhou
- Key Laboratory of Food Nutrition and Safety, Ministry of EducationTianjin University of Science and TechnologyTianjinP. R. China
- ARC Industrial Transformation Training Centre for Functional GrainsCharles Sturt UniversityWagga, WaggaAustralia
| | - Yongjia Diao
- Key Laboratory of Food Nutrition and Safety, Ministry of EducationTianjin University of Science and TechnologyTianjinP. R. China
| | - Padraig Strappe
- ARC Industrial Transformation Training Centre for Functional GrainsCharles Sturt UniversityWagga, WaggaAustralia
| | - Chris Blanchard
- ARC Industrial Transformation Training Centre for Functional GrainsCharles Sturt UniversityWagga, WaggaAustralia
| |
Collapse
|
39
|
Sato T, Shiba-Ishii A, Kim Y, Dai T, Husni RE, Hong J, Kano J, Sakashita S, Iijima T, Noguchi M. miR-3941: A novel microRNA that controls IGBP1 expression and is associated with malignant progression of lung adenocarcinoma. Cancer Sci 2017; 108:536-542. [PMID: 28012229 PMCID: PMC5378261 DOI: 10.1111/cas.13148] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/25/2016] [Accepted: 12/18/2016] [Indexed: 01/06/2023] Open
Abstract
Immunoglobulin (CD79a) binding protein 1 (IGBP1) is universally overexpressed in lung adenocarcinoma and exerts an anti‐apoptotic effect by binding to PP2Ac. However, the molecular mechanism of IGBP1 overexpression is still unclear. In the present study, we used a microRNA (miRNA) array and TargetScan Human software to detect IGBP1‐related miRNAs that regulate IGBP1 expression. The miRNA array analysis revealed more than 100 miRNAs that are dysregulated in early invasive adenocarcinoma. On the other hand, in silico analysis using TargetScan Human revealed 79 miRNAs that are associated with IGBP1 protein expression. Among the miRNAs selected by miRNA array analysis, six (miR‐34b, miR‐138, miR‐374a, miR‐374b, miR‐1909, miR‐3941) were also included among those selected by TargetScan analysis. Real‐time reverse transcription PCR (real‐time RT‐PCR) showed that the six microRNAs were downregulated in invasive adenocarcinoma (IGBP1+) relative to adjacent normal lung tissue (IGBP1−). Among these microRNAs, only miR‐34b and miR‐3941 depressed luciferase activity by targeting 3′UTR‐IGBP1 in the luciferase vector. We transfected miR‐34b and miR‐3941 into lung adenocarcinoma cell lines (A549, PC‐9), and both of them suppressed IGBP1 expression and cell proliferation. Moreover, the transfected miR‐34b and miR‐3941 induced apoptosis of a lung adenocarcinoma cell line, similarly to the effect of siIGBP1 RNA. As well as miR‐34b, we found that miR‐3941 targeted IGBP1 specifically and was able to exclusively downregulate IGBP1 expression. These findings indicate that suppression of miR‐3941 has an important role in the progression of lung adenocarcinoma at an early stage.
Collapse
Affiliation(s)
- Taiki Sato
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba-shi, Ibaraki, Japan
| | - Aya Shiba-Ishii
- Department of Pathology, Faculty of Medicine, University of Tsukuba, Tsukuba-shi, Ibaraki, Japan
| | - Yunjung Kim
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba-shi, Ibaraki, Japan
| | - Tomoko Dai
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba-shi, Ibaraki, Japan
| | - Ryan Edbert Husni
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba-shi, Ibaraki, Japan
| | - JeongMin Hong
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba-shi, Ibaraki, Japan
| | - Junko Kano
- Department of Pathology, Faculty of Medicine, University of Tsukuba, Tsukuba-shi, Ibaraki, Japan
| | - Shingo Sakashita
- Department of Pathology, Faculty of Medicine, University of Tsukuba, Tsukuba-shi, Ibaraki, Japan
| | - Tatsuo Iijima
- Department of Pathology, Ibaraki Prefectural Central Hospital, Kasama-shi, Ibaraki, Japan
| | - Masayuki Noguchi
- Department of Pathology, Faculty of Medicine, University of Tsukuba, Tsukuba-shi, Ibaraki, Japan
| |
Collapse
|
40
|
Liu CY, Hsieh FS, Chu PY, Tsai WC, Huang CT, Yu YB, Huang TT, Ko PS, Hung MH, Wang WL, Shiau CW, Chen KF. Carfilzomib induces leukaemia cell apoptosis via inhibiting ELK1/KIAA1524 (Elk-1/CIP2A) and activating PP2A not related to proteasome inhibition. Br J Haematol 2017; 177:726-740. [PMID: 28340282 DOI: 10.1111/bjh.14620] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 12/22/2016] [Indexed: 01/23/2023]
Abstract
Enhancing the tumour suppressive activity of protein phosphatase 2A (PP2A) has been suggested to be an anti-leukaemic strategy. KIAA1524 (also termed CIP2A), an oncoprotein inhibiting PP2A, is associated with disease progression in chronic myeloid leukaemia and may be prognostic in cytogenetically normal acute myeloid leukaemia. Here we demonstrated that the selective proteasome inhibitor, carfilzomib, induced apoptosis in sensitive primary leukaemia cells and in sensitive leukaemia cell lines, associated with KIAA1524 protein downregulation, increased PP2A activity and decreased p-Akt, but not with the proteasome inhibition effect of carfilzomib. Ectopic expression of KIAA1524, or pretreatment with the PP2A inhibitor, okadaic acid, suppressed carfilzomib-induced apoptosis and KIAA1524 downregulation in sensitive cells, whereas co-treatment with the PP2A agonist, forskolin, enhanced carfilzomib-induced apoptosis in resistant cells. Mechanistically, carfilzomib affected KIAA1524 transcription through disturbing ELK1 (Elk-1) binding to the KIAA1524 promoter. Moreover, the drug sensitivity and mechanism of carfilzomib in xenograft mouse models correlated well with the effects of carfilzomib on KIAA1524 and p-Akt expression, as well as PP2A activity. Our data disclosed a novel drug mechanism of carfilzomib in leukaemia cells and suggests the potential therapeutic implication of KIAA1524 in leukaemia treatment.
Collapse
Affiliation(s)
- Chun-Yu Liu
- Comprehensive Breast Health Centre, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Haematology and Oncology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Feng-Shu Hsieh
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Pei-Yi Chu
- Department of Pathology, Show Chwan Memorial Hospital, Changhua, Taiwan.,School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Wen-Chun Tsai
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chun-Teng Huang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Haematology and Oncology, Department of Medicine, Yang-Ming Branch of Taipei City Hospital, Taipei, Taiwan
| | - Yuan-Bin Yu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Haematology and Oncology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzu-Ting Huang
- Comprehensive Breast Health Centre, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Po-Shen Ko
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Haematology and Oncology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Man-Hsin Hung
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Haematology and Oncology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wan-Lun Wang
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chung-Wai Shiau
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Kuen-Feng Chen
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan.,National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
41
|
Hoffman A, Taleski G, Sontag E. The protein serine/threonine phosphatases PP2A, PP1 and calcineurin: A triple threat in the regulation of the neuronal cytoskeleton. Mol Cell Neurosci 2017; 84:119-131. [PMID: 28126489 DOI: 10.1016/j.mcn.2017.01.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 01/16/2017] [Accepted: 01/21/2017] [Indexed: 01/08/2023] Open
Abstract
The microtubule, F-actin and neurofilament networks play a critical role in neuronal cell morphogenesis, polarity and synaptic plasticity. Significantly, the assembly/disassembly and stability of these cytoskeletal networks is crucially modulated by protein phosphorylation and dephosphorylation events. Herein, we aim to more closely examine the role played by three major neuronal Ser/Thr protein phosphatases, PP2A, PP1 and calcineurin, in the homeostasis of the neuronal cytoskeleton. There is strong evidence that these enzymes interact with and dephosphorylate a variety of cytoskeletal proteins, resulting in major regulation of neuronal cytoskeletal dynamics. Conversely, we also discuss how multi-protein cytoskeletal scaffolds can also influence the regulation of these phosphatases, with important implications for neuronal signalling and homeostasis. Not surprisingly, deregulation of these cytoskeletal scaffolds and phosphatase dysfunction are associated with many neurological diseases.
Collapse
Affiliation(s)
- Alexander Hoffman
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, and Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Goce Taleski
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, and Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Estelle Sontag
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, and Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW 2308, Australia.
| |
Collapse
|
42
|
Ren H, Yang B, Ruiz JA, Efe O, Ilori TO, Sands JM, Klein JD. Phosphatase inhibition increases AQP2 accumulation in the rat IMCD apical plasma membrane. Am J Physiol Renal Physiol 2016; 311:F1189-F1197. [PMID: 27488997 PMCID: PMC5210195 DOI: 10.1152/ajprenal.00150.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 08/01/2016] [Indexed: 01/11/2023] Open
Abstract
Vasopressin triggers the phosphorylation and apical plasma membrane accumulation of aquaporin 2 (AQP2), and it plays an essential role in urine concentration. Vasopressin, acting through protein kinase A, phosphorylates AQP2. However, the phosphorylation state of AQP2 could also be affected by the action of protein phosphatases (PPs). Rat inner medullas (IM) were incubated with calyculin (PP1 and PP2A inhibitor, 50 nM) or tacrolimus (PP2B inhibitor, 100 nM). Calyculin did not affect total AQP2 protein abundance (by Western blot) but did significantly increase the abundances of pS256-AQP2 and pS264-AQP2. It did not change pS261-AQP2 or pS269-AQP2. Calyculin significantly enhanced the membrane accumulation (by biotinylation) of total AQP2, pS256-AQP2, and pS264-AQP2. Likewise, immunohistochemistry showed an increase in the apical plasma membrane association of pS256-AQP2 and pS264-AQP2 in calyculin-treated rat IM. Tacrolimus also did not change total AQP2 abundance but significantly increased the abundances of pS261-AQP2 and pS264-AQP2. In contrast to calyculin, tacrolimus did not change the amount of total AQP2 in the plasma membrane (by biotinylation and immunohistochemistry). Tacrolimus did increase the expression of pS264-AQP2 in the apical plasma membrane (by immunohistochemistry). In conclusion, PP1/PP2A regulates the phosphorylation and apical plasma membrane accumulation of AQP2 differently than PP2B. Serine-264 of AQP2 is a phosphorylation site that is regulated by both PP1/PP2A and PP2B. This dual regulatory pathway may suggest a previously unappreciated role for multiple phosphatases in the regulation of urine concentration.
Collapse
Affiliation(s)
- Huiwen Ren
- Department of Medicine, Renal Division, Emory University School of Medicine, Atlanta, Georgia
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China; and
| | - Baoxue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China; and
| | - Joseph A Ruiz
- Department of Medicine, Renal Division, Emory University School of Medicine, Atlanta, Georgia
| | - Orhan Efe
- Department of Medicine, Renal Division, Emory University School of Medicine, Atlanta, Georgia
| | - Titilayo O Ilori
- Department of Medicine, Renal Division, Emory University School of Medicine, Atlanta, Georgia
| | - Jeff M Sands
- Department of Medicine, Renal Division, Emory University School of Medicine, Atlanta, Georgia
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| | - Janet D Klein
- Department of Medicine, Renal Division, Emory University School of Medicine, Atlanta, Georgia;
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
43
|
Park H, Lee K, Park ES, Oh S, Yan R, Zhang J, Beach TG, Adler CH, Voronkov M, Braithwaite SP, Stock JB, Mouradian MM. Dysregulation of protein phosphatase 2A in parkinson disease and dementia with lewy bodies. Ann Clin Transl Neurol 2016; 3:769-780. [PMID: 27752512 PMCID: PMC5048387 DOI: 10.1002/acn3.337] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/08/2016] [Indexed: 01/29/2023] Open
Abstract
OBJECTIVE Protein phosphatase 2A (PP2A) is a heterotrimeric holoenzyme composed of a catalytic C subunit, a structural A subunit, and one of several regulatory B subunits that confer substrate specificity. The assembly and activity of PP2A are regulated by reversible methylation of the C subunit. α-Synuclein, which aggregates in Parkinson disease (PD) and dementia with Lewy bodies (DLB), is phosphorylated at Ser129, and PP2A containing a B55α subunit is a major phospho-Ser129 phosphatase. The objective of this study was to investigate PP2A in α-synucleinopathies. METHODS We compared the state of PP2A methylation, as well as the expression of its methylating enzyme, leucine carboxyl methyltransferase (LCMT-1), and demethylating enzyme, protein phosphatase methylesterase (PME-1), in postmortem brains from PD and DLB cases as well as age-matched Controls. Immunohistochemical studies and quantitative image analysis were employed. RESULTS LCMT-1 was significantly reduced in the substantia nigra (SN) and frontal cortex in both PD and DLB. PME-1, on the other hand, was elevated in the PD SN. In concert with these changes, the ratio of methylated PP2A to demethylated PP2A was markedly decreased in PD and DLB brains in both SN and frontal cortex. No changes in total PP2A or total B55α subunit were detected. INTERPRETATION These findings support the hypothesis that PP2A dysregulation in α-synucleinopathies may contribute to the accumulation of hyperphosphorylated α-synuclein and to the disease process, raising the possibility that pharmacological means to enhance PP2A phosphatase activity may be a useful disease-modifying therapeutic approach.
Collapse
Affiliation(s)
- Hye‐Jin Park
- Center for Neurodegenerative and Neuroimmunologic DiseasesDepartment of NeurologyRutgers – Robert Wood Johnson Medical SchoolPiscatawayNew Jersey08854
| | - Kang‐Woo Lee
- Center for Neurodegenerative and Neuroimmunologic DiseasesDepartment of NeurologyRutgers – Robert Wood Johnson Medical SchoolPiscatawayNew Jersey08854
- Present address: Department of Biological SciencesKorea Advanced Institute of Science and Technology (KAIST)Daejeon305‐701Republic of Korea
| | - Eun S. Park
- Center for Neurodegenerative and Neuroimmunologic DiseasesDepartment of NeurologyRutgers – Robert Wood Johnson Medical SchoolPiscatawayNew Jersey08854
- Present address: Albert Einstein College of MedicineBronxNew Jersey10461
| | - Stephanie Oh
- Center for Neurodegenerative and Neuroimmunologic DiseasesDepartment of NeurologyRutgers – Robert Wood Johnson Medical SchoolPiscatawayNew Jersey08854
| | - Run Yan
- Center for Neurodegenerative and Neuroimmunologic DiseasesDepartment of NeurologyRutgers – Robert Wood Johnson Medical SchoolPiscatawayNew Jersey08854
| | - Jie Zhang
- Center for Neurodegenerative and Neuroimmunologic DiseasesDepartment of NeurologyRutgers – Robert Wood Johnson Medical SchoolPiscatawayNew Jersey08854
| | | | | | | | - Steven P. Braithwaite
- Signum Biosciences133 Wall StreetPrincetonNew Jersey08540
- Present address: Alkahest75 Shoreway Drive, Suite DSan CarlosCalifornia94070
| | - Jeffry B. Stock
- Signum Biosciences133 Wall StreetPrincetonNew Jersey08540
- Department of Molecular BiologyPrinceton UniversityPrincetonNew Jersey08544
| | - M. Maral Mouradian
- Center for Neurodegenerative and Neuroimmunologic DiseasesDepartment of NeurologyRutgers – Robert Wood Johnson Medical SchoolPiscatawayNew Jersey08854
| |
Collapse
|
44
|
Wu J, Wang J, Zeng X, Chen Y, Xia J, Wang S, Huang Z, Chen W, Shen Z. Protein phosphatase 2A regulatory subunit B56β modulates erythroid differentiation. Biochem Biophys Res Commun 2016; 478:1179-84. [PMID: 27544028 DOI: 10.1016/j.bbrc.2016.08.090] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 08/16/2016] [Indexed: 11/29/2022]
Abstract
Anemia due to attenuated erythroid terminal differentiation is one of the most common hematological disorders occurring at all stages of life. We previously demonstrated that catalytic subunit α of protein phosphatase 2A (PP2Acα) modulates fetal liver erythropoiesis. However the corresponding PP2A regulatory subunit in this process remains unknown. In this study, we report that chemical inhibition of PP2A activity with okadaic acid impairs hemin-induced erythroid differentiation. Interestingly, B56 family member B56β is the only regulatory subunit whose expression is induced by both erythropoietin in fetal liver cells and hemin in erythroleukemia K562 cells. Finally, knockdown of B56β attenuates hemin-induced K562 erythroid differentiation. Collectively, our data identify B56β as the potential functional regulatory subunit of PP2A in erythroid differentiation, shedding light on new target for precise modulation of PP2A activity for treatment of anemia and related diseases.
Collapse
Affiliation(s)
- Jianping Wu
- Orthopedic Department of the Second Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Jun Wang
- Emergency Department of the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xiansheng Zeng
- Department of Cardiology of the First Affiliated Hospital, Soochow University, Suzhou 215006, China
| | - Yueqiu Chen
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Soochow University, Suzhou 215006, China
| | - Jun Xia
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Soochow University, Suzhou 215006, China
| | - Shizhen Wang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Soochow University, Suzhou 215006, China
| | - Zan Huang
- Jiangsu Province Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agriculture University, Nanjing 210000, China.
| | - Weiqian Chen
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Soochow University, Suzhou 215006, China.
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou 215006, China.
| |
Collapse
|
45
|
Kariithi HM, İnce İA, Boeren S, Murungi EK, Meki IK, Otieno EA, Nyanjom SRG, van Oers MM, Vlak JM, Abd-Alla AMM. Comparative Analysis of Salivary Gland Proteomes of Two Glossina Species that Exhibit Differential Hytrosavirus Pathologies. Front Microbiol 2016; 7:89. [PMID: 26903969 PMCID: PMC4746320 DOI: 10.3389/fmicb.2016.00089] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/18/2016] [Indexed: 01/19/2023] Open
Abstract
Glossina pallidipes salivary gland hypertrophy virus (GpSGHV; family Hytrosaviridae) is a dsDNA virus exclusively pathogenic to tsetse flies (Diptera; Glossinidae). The 190 kb GpSGHV genome contains 160 open reading frames and encodes more than 60 confirmed proteins. The asymptomatic GpSGHV infection in flies can convert to symptomatic infection that is characterized by overt salivary gland hypertrophy (SGH). Flies with SGH show reduced general fitness and reproductive dysfunction. Although the occurrence of SGH is an exception rather than the rule, G. pallidipes is thought to be the most susceptible to expression of overt SGH symptoms compared to other Glossina species that are largely asymptomatic. Although Glossina salivary glands (SGs) play an essential role in GpSGHV transmission, the functions of the salivary components during the virus infection are poorly understood. In this study, we used mass spectrometry to study SG proteomes of G. pallidipes and G. m. morsitans, two Glossina model species that exhibit differential GpSGHV pathologies (high and low incidence of SGH, respectively). A total of 540 host proteins were identified, of which 23 and 9 proteins were significantly up- and down-regulated, respectively, in G. pallidipes compared to G. m. morsitans. Whereas 58 GpSGHV proteins were detected in G. pallidipes F1 progenies, only 5 viral proteins were detected in G. m. morsitans. Unlike in G. pallidipes, qPCR assay did not show any significant increase in virus titers in G. m. morsitans F1 progenies, confirming that G. m. morsitans is less susceptible to GpSGHV infection and replication compared to G. pallidipes. Based on our results, we speculate that in the case of G. pallidipes, GpSGHV employs a repertoire of host intracellular signaling pathways for successful infection. In the case of G. m. morsitans, antiviral responses appeared to be dominant. These results are useful for designing additional tools to investigate the Glossina-GpSGHV interactions.
Collapse
Affiliation(s)
- Henry M Kariithi
- Biotechnology Research Institute, Kenya Agricultural and Livestock Research OrganizationNairobi, Kenya; Insect Pest Control Laboratory, Joint FAO/IAEA Programme of Nuclear Techniques in Food and Agriculture, International Atomic Energy AgencyVienna, Austria; Laboratory of Virology, Wageningen UniversityWageningen, Netherlands
| | - İkbal Agah İnce
- Department of Medical Microbiology, Acıbadem University İstanbul, Turkey
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University Wageningen, Netherlands
| | - Edwin K Murungi
- South African National Bioinformatics Institute, University of the Western Cape Cape Town, South Africa
| | - Irene K Meki
- Insect Pest Control Laboratory, Joint FAO/IAEA Programme of Nuclear Techniques in Food and Agriculture, International Atomic Energy AgencyVienna, Austria; Laboratory of Virology, Wageningen UniversityWageningen, Netherlands
| | - Everlyne A Otieno
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology Nairobi, Kenya
| | - Steven R G Nyanjom
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology Nairobi, Kenya
| | | | - Just M Vlak
- Laboratory of Virology, Wageningen University Wageningen, Netherlands
| | - Adly M M Abd-Alla
- Insect Pest Control Laboratory, Joint FAO/IAEA Programme of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency Vienna, Austria
| |
Collapse
|
46
|
Lai SS, Zhao DD, Cao P, Lu K, Luo OY, Chen WB, Liu J, Jiang EZ, Yu ZH, Lee G, Li J, Yu DC, Xu XJ, Zhu MS, Gao X, Li CJ, Xue B. PP2Acα positively regulates the termination of liver regeneration in mice through the AKT/GSK3β/Cyclin D1 pathway. J Hepatol 2016; 64:352-360. [PMID: 26456844 DOI: 10.1016/j.jhep.2015.09.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 09/17/2015] [Accepted: 09/25/2015] [Indexed: 02/09/2023]
Abstract
BACKGROUND & AIMS Liver injury triggers a highly organized and ordered liver regeneration (LR) process. Once regeneration is complete, a stop signal ensures that the regenerated liver is an appropriate functional size. The inhibitors and stop signals that regulate LR are unknown, and only limited information is available about these mechanisms. METHODS A 70% partial hepatectomy (PH) was performed in hepatocyte-specific PP2Acα-deleted (PP2Acα(-/-)) and control (PP2Acα(+/+)) mice. LR was estimated by liver weight, serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels and cell proliferation, and the related cellular signals were analyzed. RESULTS We found that the catalytic subunit of PP2A was markedly upregulated during the late stage of LR. PP2Acα(-/-) mice showed prolonged LR termination, an increased liver size compared to the original mass and lower levels of serum ALT and AST compared with control mice. In these mice, cyclin D1 protein levels, but not mRNA levels, were increased. Mechanistically, AKT activated by the loss of PP2Acα inhibited glycogen synthase kinase 3β (GSK3β) activity, which led to the accumulation of cyclin D1 protein and accelerated hepatocyte proliferation at the termination stage. Treatment with the PI3K inhibitor wortmannin at the termination stage was sufficient to inhibit cyclin D1 accumulation and hepatocyte proliferation. CONCLUSIONS PP2Acα plays an essential role in the proper termination of LR via the AKT/GSK3β/Cyclin D1 pathway. Our findings enrich the understanding of the molecular mechanism that controls the termination of LR and provides a potential therapeutic target for treating liver injury.
Collapse
Affiliation(s)
- Shan-Shan Lai
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center and School of Medicine, Nanjing University, Nanjing 210093, China
| | - Dan-Dan Zhao
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center and School of Medicine, Nanjing University, Nanjing 210093, China
| | - Peng Cao
- Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Institute of Chinese Medicine, Nanjing 210028, China
| | - Ke Lu
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center and School of Medicine, Nanjing University, Nanjing 210093, China
| | - Ou-Yang Luo
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center and School of Medicine, Nanjing University, Nanjing 210093, China
| | - Wei-Bo Chen
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center and School of Medicine, Nanjing University, Nanjing 210093, China; Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210093, China
| | - Jia Liu
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center and School of Medicine, Nanjing University, Nanjing 210093, China
| | - En-Ze Jiang
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center and School of Medicine, Nanjing University, Nanjing 210093, China
| | - Zi-Han Yu
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center and School of Medicine, Nanjing University, Nanjing 210093, China
| | - Gina Lee
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Jing Li
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - De-Cai Yu
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210093, China
| | - Xiao-Jun Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Min-Sheng Zhu
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center and School of Medicine, Nanjing University, Nanjing 210093, China
| | - Xiang Gao
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center and School of Medicine, Nanjing University, Nanjing 210093, China.
| | - Chao-Jun Li
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center and School of Medicine, Nanjing University, Nanjing 210093, China.
| | - Bin Xue
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center and School of Medicine, Nanjing University, Nanjing 210093, China.
| |
Collapse
|
47
|
Lee JO, Kim N, Lee HJ, Moon JW, Lee SK, Kim SJ, Kim JK, Park SH, Kim HS. [6]-Gingerol Affects Glucose Metabolism by Dual Regulation via the AMPKα2-Mediated AS160-Rab5 Pathway and AMPK-Mediated Insulin Sensitizing Effects. J Cell Biochem 2016; 116:1401-10. [PMID: 25694332 DOI: 10.1002/jcb.25100] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 01/23/2015] [Indexed: 12/24/2022]
Abstract
[6]-Gingerol has been used to control diabetes and dyslipidemia; however, its metabolic role is poorly understood. In this study, [6]-gingerol increased adenosine monophosphate (AMP)-activated protein kinase (AMPK) phosphorylation in mouse skeletal muscle C2C12 cells. Stimulation of glucose uptake by [6]-gingerol was dependent on AMPKα2. Moreover, both Inhibition and knockdown of AMPKα2 blocked [6]-gingerol-induced glucose uptake. [6]-Gingerol significantly decreased the activity of protein phosphatase 2A (PP2A). Inhibition of PP2A activity with okadaic acid enhanced the phosphorylation of AMPKα2. Moreover, the interaction between AMPKα2 and PP2A was increased by [6]-gingerol, suggesting that PP2A mediates the effect of [6]-gingerol on AMPK phosphorylation. In addition, [6]-gingerol increased the phosphorylation of Akt-substrate 160 (AS160), which is a Rab GTPase-activating protein. Inhibition of AMPKα2 blocked [6]-gingerol-induced AS160 phosphorylation. [6]-gingerol increased the Rab5, and AMPKα2 knockdown blocked [6]-gingerol-induced expression of Rab5, indicating AMPK play as an upstream of Rab5. It also increased glucose transporter 4 (GLUT4) mRNA and protein expression and stimulated GLUT4 translocation. Furthermore, insulin-mediated glucose uptake and Akt phosphorylation were further potentiated by [6]-gingerol treatment. This potentiation was not observed in the presence of AMPK inhibitor compound C. In summary, our results suggest that [6]-gingerol plays an important role in glucose metabolism via the AMPKα2-mediated AS160-Rab5 pathway and through potentiation of insulin-mediated glucose regulation.
Collapse
Affiliation(s)
- Jung Ok Lee
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea
| | - Nami Kim
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea
| | - Hye Jeong Lee
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea
| | - Ji Wook Moon
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea
| | - Soo Kyung Lee
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea
| | - Su Jin Kim
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea
| | - Joong Kwan Kim
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea
| | - Sun Hwa Park
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea
| | - Hyeon Soo Kim
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
48
|
Zhang Y, Jiang X, Qin C, Cuevas S, Jose PA, Armando I. Dopamine D2 receptors' effects on renal inflammation are mediated by regulation of PP2A function. Am J Physiol Renal Physiol 2016; 310:F128-34. [PMID: 26290374 PMCID: PMC4719046 DOI: 10.1152/ajprenal.00453.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 06/15/2015] [Indexed: 01/11/2023] Open
Abstract
Lack or downregulation of the dopamine D2 receptor (D2R) results in increased renal expression of injury markers and proinflammatory factors that is independent of a blood pressure increase. This study aimed to determine the mechanisms involved in the regulation of renal inflammation by D2Rs. Silencing D2Rs in mouse renal proximal tubule cells increased the expression of the proinflammatory TNF-α, monocyte chemoattractant protein-1 (MCP-1), and IL-6. D2R downregulation also increased Akt phosphorylation and activity, and glycogen synthase kinase-3β (GSK3β) phosphorylation and cyclin D1 expression, downstream targets of Akt; however. phosphatidylinositol 3-kinase (PI3K) activity was not affected. Conversely, D2R stimulation decreased Akt and GSK3β phosphorylation and cyclin D1 expression. Increased phospho-Akt, in the absence of increased PI3K activity, may result from decreased Akt dephosphorylation. Inhibition of protein phosphatase 2A (PP2A) with okadaic acid reproduced the effects of D2R downregulation on Akt, GSK3β, and cyclin D1. The PP2A catalytic subunit and regulatory subunit PPP2R2C coimmunoprecipitated with the D2R. Basal phosphatase activity and the expression of PPP2R2C were decreased by D2R silencing that also blunted the increase in phosphatase activity induced by D2R stimulation. Similarly, silencing PPP2R2C also increased the phosphorylation of Akt and GSK3β. Moreover, downregulation of PPP2R2C resulted in increased expression of TNF-α, MCP-1, and IL-6, indicating that decreased phosphatase activity may be responsible for the D2R effect on inflammatory factors. Indeed, the increase in NF-κB reporter activity induced by D2R silencing was blunted by increasing PP2A activity with protamine. Our results show that D2R controls renal inflammation, at least in part, by modulation of the Akt pathway through effects on PP2A activity/expression.
Collapse
Affiliation(s)
- Yanrong Zhang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P. R. China; and
| | - Xiaoliang Jiang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P. R. China; and
| | - Chuan Qin
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P. R. China; and
| | - Santiago Cuevas
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Pedro A Jose
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ines Armando
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
49
|
Chen W, Wang S, Xia J, Huang Z, Tu X, Shen Z. Protein phosphatase 2A plays an important role in migration of bone marrow stroma cells. Mol Cell Biochem 2015; 412:173-80. [PMID: 26708215 DOI: 10.1007/s11010-015-2624-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 12/08/2015] [Indexed: 01/07/2023]
Abstract
Administration of bone marrow stroma cells (BMSCs) has the potential to ameliorate degenerative disorders and to repair injured sites. The homing of transplanted BMSCs to damaged tissues is a critical property of engraftment. Therefore, it is important to understand signal molecules controlling migration of BMSCs. Here, we demonstrate that serine-threonine protein phosphatase 2A (PP2A) is responsive to migration of BMSCs. Pharmacological Inhibition of PP2A, using okadaic acid (OA), leads to attenuated cell migration in rat primary BMSCs both in the absence or presence of stromal cell-derived factor-1 (SDF-1). Consistent with the above findings, knockdown of the main catalytic subunit PP2Acα using small interfering RNA also attenuates chemotaxis of BMSCs. On the other hand, cell viability of BMSCs remains unchanged with OA treatment or knockdown of PP2Acα subunit. Moreover, we observed an upregulation of PP2A-B55β in transcription level after SDF-1 treatment, indicating their potential role as the functioning regulatory subunit of PP2A phosphatase in BMSCs migration model. Collectively, these data provide first insight into the modulation of BMSCs migration by PP2A phosphatase activity and lay a foundation for exploring PP2A signaling as a modulating target for BMSCs transplantation.
Collapse
Affiliation(s)
- Weiqian Chen
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Shizhen Wang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Jun Xia
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Zan Huang
- Jiangsu Province Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agriculture University, Nanjing, 210095, Jiangsu, China
| | - Xin Tu
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, 210061, Jiangsu, China
| | - Zhenya Shen
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
50
|
Dudiki T, Kadunganattil S, Ferrara JK, Kline DW, Vijayaraghavan S. Changes in Carboxy Methylation and Tyrosine Phosphorylation of Protein Phosphatase PP2A Are Associated with Epididymal Sperm Maturation and Motility. PLoS One 2015; 10:e0141961. [PMID: 26569399 PMCID: PMC4646675 DOI: 10.1371/journal.pone.0141961] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 10/15/2015] [Indexed: 01/21/2023] Open
Abstract
Mammalian sperm contain the serine/threonine phosphatases PP1γ2 and PP2A. The role of sperm PP1γ2 is relatively well studied. Here we confirm the presence of PP2A in sperm and show that it undergoes marked changes in methylation (leucine 309), tyrosine phosphorylation (tyrosine 307) and catalytic activity during epididymal sperm maturation. Spermatozoa isolated from proximal caput, distal caput and caudal regions of the epididymis contain equal immuno-reactive amounts of PP2A. Using demethyl sensitive antibodies we show that PP2A is methylated at its carboxy terminus in sperm from the distal caput and caudal regions but not in sperm from the proximal caput region of the epididymis. The methylation status of PP2A was confirmed by isolation of PP2A with microcystin agarose followed by alkali treatment, which causes hydrolysis of protein carboxy methyl esters. Tyrosine phosphorylation of sperm PP2A varied inversely with methylation. That is, PP2A was tyrosine phosphorylated when it was demethylated but not when methylated. PP2A demethylation and its reciprocal tyrosine phosphorylation were also affected by treatment of sperm with L-homocysteine and adenosine, which are known to elevate intracellular S-adenosylhomocysteine, a feedback inhibitor of methyltransferases. Catalytic activity of PP2A declined during epididymal sperm maturation. Inhibition of PP2A by okadaic acid or by incubation of caudal epididymal spermatozoa with L-homocysteine and adenosine resulted in increase of sperm motility parameters including percent motility, velocity, and lateral head amplitude. Demethylation or pharmacological inhibition of PP2A also leads to an increase in phosphorylation of glycogen synthase kinase-3 (GSK3). Our results show for the first time that changes in PP2A activity due to methylation and tyrosine phosphorylation occur in sperm and that these changes may play an important role in the regulation of sperm function.
Collapse
Affiliation(s)
- Tejasvi Dudiki
- Department of Biological Sciences, Kent State University, Kent, Ohio, United States of America
| | - Suraj Kadunganattil
- Department of Biological Sciences, Kent State University, Kent, Ohio, United States of America
| | - John K. Ferrara
- Department of Biological Sciences, Kent State University, Kent, Ohio, United States of America
| | - Douglas W. Kline
- Department of Biological Sciences, Kent State University, Kent, Ohio, United States of America
| | | |
Collapse
|