1
|
Kukulage DSK, Samarasinghe KTG, Matarage Don NNJ, Shivamadhu MC, Shishikura K, Schiff W, Mashhadi Ramezani F, Padmavathi R, Matthews ML, Ahn YH. Protein phosphatase PP2Cα S-glutathionylation regulates cell migration. J Biol Chem 2024; 300:107784. [PMID: 39303918 PMCID: PMC11530597 DOI: 10.1016/j.jbc.2024.107784] [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: 05/18/2024] [Revised: 08/07/2024] [Accepted: 08/29/2024] [Indexed: 09/22/2024] Open
Abstract
Redox signaling is a fundamental mechanism that controls all major biological processes partly via protein cysteine oxidations, including S-glutathionylation. Despite over 2000 cysteines identified to form S-glutathionylation in databases, the identification of redox cysteines functionally linked to a biological process of interest remains challenging. Here, we demonstrate a strategy combining glutathionylation proteomic database, bioinformatics, and biological screening, which resulted in the identification of S-glutathionylated proteins, including PP2Cα, as redox players of cell migration. We showed that PP2Cα, a prototypical magnesium-dependent serine/threonine phosphatase, is susceptible to S-glutathionylation selectively at nonconserved C314. PP2Cα glutathionylation causes increased migration and invasion of breast cancer cell lines in oxidative stress or upon hydrogen peroxide production. Mechanistically, PP2Cα glutathionylation modulates its protein-protein interactions, activating c-Jun N-terminal kinase and extracellular signal-regulated kinase pathways to elevate migration and invasion. In addition, PP2Cα glutathionylation occurs in response to epidermal growth factor, supporting a serine/threonine phosphatase PP2Cα as a new redox player in growth factor signal transduction.
Collapse
Affiliation(s)
| | | | | | - Madhu C Shivamadhu
- Department of Chemistry, Drexel University, Philadelphia, Pennsylvania, USA
| | - Kyosuke Shishikura
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - William Schiff
- Department of Chemistry, Drexel University, Philadelphia, Pennsylvania, USA
| | | | | | - Megan L Matthews
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Young-Hoon Ahn
- Department of Chemistry, Drexel University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
2
|
Li Z, Chen R, Li Y, Zhou Q, Zhao H, Zeng K, Zhao B, Lu Z. A comprehensive overview of PPM1B: From biological functions to diseases. Eur J Pharmacol 2023; 947:175633. [PMID: 36863552 DOI: 10.1016/j.ejphar.2023.175633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/08/2023] [Accepted: 02/28/2023] [Indexed: 03/04/2023]
Abstract
Reversible phosphorylation of proteins is an important mechanism that regulates cellular processes, which are precisely regulated by protein kinases and phosphatases. PPM1B is a metal ion-dependent serine/threonine protein phosphatase, which regulates multiple biological functions by targeting substrate dephosphorylation, such as cell cycle, energy metabolism, inflammatory responses. In this review, we summarized the occurrent understandings of PPM1B focused on its regulation of signaling pathways, related diseases, and small-molecular inhibitors, which may provide new insights for the identification of PPM1B inhibitors and the treatment of PPM1B-related diseases.
Collapse
Affiliation(s)
- Zhongyao Li
- School of Pharmacy and Pharmaceutical Sciences, Institute of Materia Medica, Shandong First Medical University, Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, 250117, Shandong, China
| | - Ruoyu Chen
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, 250012, Shandong, China
| | - Yanxia Li
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, 250012, Shandong, China
| | - Qian Zhou
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, 250012, Shandong, China
| | - Huanxin Zhao
- School of Pharmacy and Pharmaceutical Sciences, Institute of Materia Medica, Shandong First Medical University, Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, 250117, Shandong, China
| | - Kewu Zeng
- School of Pharmacy and Pharmaceutical Sciences, Institute of Materia Medica, Shandong First Medical University, Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, 250117, Shandong, China.
| | - Baobing Zhao
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, 250012, Shandong, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, 250012, Shandong, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, 250012, Shandong, China.
| | - Zhiyuan Lu
- School of Pharmacy and Pharmaceutical Sciences, Institute of Materia Medica, Shandong First Medical University, Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, 250117, Shandong, China.
| |
Collapse
|
3
|
Nandi N, Zaidi Z, Tracy C, Krämer H. A phospho-switch at Acinus-Serine 437 controls autophagic responses to Cadmium exposure and neurodegenerative stress. eLife 2022; 11:72169. [PMID: 35037620 PMCID: PMC8794470 DOI: 10.7554/elife.72169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 01/14/2022] [Indexed: 12/09/2022] Open
Abstract
Neuronal health depends on quality control functions of autophagy, but mechanisms regulating neuronal autophagy are poorly understood. Previously, we showed that in Drosophila starvation-independent quality control autophagy is regulated by acinus (acn) and the Cdk5-dependent phosphorylation of its serine437 (Nandi et al., 2017). Here, we identify the phosphatase that counterbalances this activity and provides for the dynamic nature of acinus-serine437 (acn-S437) phosphorylation. A genetic screen identified six phosphatases that genetically interacted with an acn gain-of-function model. Among these, loss of function of only one, the PPM-type phosphatase Nil (CG6036), enhanced pS437-acn levels. Cdk5-dependent phosphorylation of acn-S437 in nil1 animals elevates neuronal autophagy and reduces the accumulation of polyQ proteins in a Drosophila Huntington’s disease model. Consistent with previous findings that Cd2+ inhibits PPM-type phosphatases, Cd2+ exposure elevated acn-S437 phosphorylation which was necessary for increased neuronal autophagy and protection against Cd2+-induced cytotoxicity. Together, our data establish the acn-S437 phosphoswitch as critical integrator of multiple stress signals regulating neuronal autophagy.
Collapse
Affiliation(s)
- Nilay Nandi
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Zuhair Zaidi
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Charles Tracy
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Helmut Krämer
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
4
|
Li M, Xu X, Su Y, Shao X, Zhou Y, Yan J. A comprehensive overview of PPM1A: From structure to disease. Exp Biol Med (Maywood) 2021; 247:453-461. [PMID: 34861123 DOI: 10.1177/15353702211061883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
PPM1A (magnesium-dependent phosphatase 1 A, also known as PP2Cα) is a member of the Ser/Thr protein phosphatase family. Protein phosphatases catalyze the removal of phosphate groups from proteins via hydrolysis, thus opposing the role of protein kinases. The PP2C family is generally considered a negative regulator in the eukaryotic stress response pathway. PPM1A can bind and dephosphorylate various proteins and is therefore involved in the regulation of a wide range of physiological processes. It plays a crucial role in transcriptional regulation, cell proliferation, and apoptosis and has been suggested to be closely related to the occurrence and development of cancers of the lung, bladder, and breast, amongst others. Moreover, it is closely related to certain autoimmune diseases and neurodegenerative diseases. In this review, we provide an insight into currently available knowledge of PPM1A, including its structure, biological function, involvement in signaling pathways, and association with diseases. Lastly, we discuss whether PPM1A could be targeted for therapy of certain human conditions.
Collapse
Affiliation(s)
- Mao Li
- Department of Physiology, Guilin Medical University, Guilin 541004, China.,Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin 541004, China
| | - Xingfeng Xu
- Department of Physiology, Guilin Medical University, Guilin 541004, China.,Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin 541004, China
| | - Yan Su
- Department of Physiology, Guilin Medical University, Guilin 541004, China.,Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin 541004, China
| | - Xiaoyun Shao
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin 541004, China
| | - Yali Zhou
- Department of Microbiology, Guilin Medical University, Guilin 541004, China
| | - Jianguo Yan
- Department of Physiology, Guilin Medical University, Guilin 541004, China.,Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin 541004, China
| |
Collapse
|
5
|
Banerjee S, Maity S, Guchhait R, Chatterjee A, Biswas C, Adhikari M, Pramanick K. Toxic effects of cyanotoxins in teleost fish: A comprehensive review. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 240:105971. [PMID: 34560410 DOI: 10.1016/j.aquatox.2021.105971] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 09/03/2021] [Accepted: 09/13/2021] [Indexed: 06/13/2023]
Abstract
The phenomenon of eutrophication leads to the global occurrence of algal blooms. Cyanotoxins as produced by many cyanobacterial species can lead to detrimental effects to the biome due to their stability and potential biomagnification along food webs. Therefore, understanding of the potential risks these toxins pose to the most susceptible organisms is an important prerequisite for ecological risks assessment of cyanobacteria blooms. Fishes are an important component of aquatic ecosystems that are prone to direct exposure to cyanotoxins. However, relatively few investigations have focused on measuring the toxic potentials of cyanotoxins in teleost fishes. This review comprehensively describes the major toxicological impacts (such as hepatotoxicity, neurotoxicity, immune toxicity, reproductive toxicity and cytogenotoxicity) of commonly occurring cyanotoxins in teleost fishes. The present work encompasses recent research progresses with special emphasis on the basic molecular mechanisms by which different cyanotoxins impose their toxicities in teleost fishes. The major research areas, which need to be focused on in future scientific investigations, have also been highlighted. Protein kinase inhibition, transcriptional dysregulation, disruption of redox homeostasis and the induction of apoptotic pathways appear to be the key drivers of the toxicological effects of cyanotoxins in fish. Analyses also showed that the impacts of cyanotoxins on specific reproductive processes are relatively less described in teleosts in comparison to mammalian systems. In fact, as compared to other toxicological effects of cyanotoxins, their reproductive toxicity (such as impacts on oocyte development, maturation and their hormonal regulation) is poorly understood in fish, and thus requires further studies. Furthermore, additonal studies characterizing the molecular mechanisms responsible for the cellular uptake of cyanotoxins need to be investigated.
Collapse
Affiliation(s)
- Sambuddha Banerjee
- Integrative Biology Research Unit, Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata 700073, India
| | - Sukhendu Maity
- Integrative Biology Research Unit, Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata 700073, India
| | - Rajkumar Guchhait
- P.G. Department of Zoology, Mahishadal Raj College, Garkamalpur, Purba Medinipur, India
| | - Ankit Chatterjee
- Integrative Biology Research Unit, Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata 700073, India
| | - Chayan Biswas
- Integrative Biology Research Unit, Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata 700073, India
| | - Madhuchhanda Adhikari
- Integrative Biology Research Unit, Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata 700073, India
| | - Kousik Pramanick
- Integrative Biology Research Unit, Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata 700073, India.
| |
Collapse
|
6
|
Neumann J, Boknik P, Kirchhefer U, Gergs U. The role of PP5 and PP2C in cardiac health and disease. Cell Signal 2021; 85:110035. [PMID: 33964402 DOI: 10.1016/j.cellsig.2021.110035] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/16/2021] [Accepted: 05/03/2021] [Indexed: 02/08/2023]
Abstract
Protein phosphatases are important, for example, as functional antagonists of β-adrenergic stimulation of the mammalian heart. While β-adrenergic stimulations increase the phosphorylation state of regulatory proteins and therefore force of contraction in the heart, these phosphorylations are reversed and thus force is reduced by the activity of protein phosphatases. In this context the role of PP5 and PP2C is starting to unravel. They do not belong to the same family of phosphatases with regard to sequence homology, many similarities with regard to location, activation by lipids and putative substrates have been worked out over the years. We also suggest which pathways for regulation of PP5 and/or PP2C described in other tissues and not yet in the heart might be useful to look for in cardiac tissue. Both phosphatases might play a role in signal transduction of sarcolemmal receptors in the heart. Expression of PP5 and PP2C can be increased by extracellular stimuli in the heart. Because PP5 is overexpressed in failing animal and human hearts, and because overexpression of PP5 or PP2C leads to cardiac hypertrophy and KO of PP5 leads to cardiac hypotrophy, one might argue for a role of PP5 and PP2C in heart failure. Because PP5 and PP2C can reduce, at least in vitro, the phosphorylation state of proteins thought to be relevant for cardiac arrhythmias, a role of these phosphatases for cardiac arrhythmias is also probable. Thus, PP5 and PP2C might be druggable targets to treat important cardiac diseases like heart failure, cardiac hypertrophy and cardiac arrhythmias.
Collapse
Affiliation(s)
- Joachim Neumann
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Magdeburger Str. 4, D-06097 Halle, Germany.
| | - Peter Boknik
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Westfälische Wilhelms-Universität, Domagkstraße 12, D-48149 Münster, Germany.
| | - Uwe Kirchhefer
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Westfälische Wilhelms-Universität, Domagkstraße 12, D-48149 Münster, Germany.
| | - Ulrich Gergs
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Magdeburger Str. 4, D-06097 Halle, Germany.
| |
Collapse
|
7
|
Seumen CHT, Grimm TM, Hauck CR. Protein phosphatases in TLR signaling. Cell Commun Signal 2021; 19:45. [PMID: 33882943 PMCID: PMC8058998 DOI: 10.1186/s12964-021-00722-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptors (TLRs) are critical sensors for the detection of potentially harmful microbes. They are instrumental in initiating innate and adaptive immune responses against pathogenic organisms. However, exaggerated activation of TLR receptor signaling can also be responsible for the onset of autoimmune and inflammatory diseases. While positive regulators of TLR signaling, such as protein serine/threonine kinases, have been studied intensively, only little is known about phosphatases, which counterbalance and limit TLR signaling. In this review, we summarize protein phosphorylation events and their roles in the TLR pathway and highlight the involvement of protein phosphatases as negative regulators at specific steps along the TLR-initiated signaling cascade. Then, we focus on individual phosphatase families, specify the function of individual enzymes in TLR signaling in more detail and give perspectives for future research. A better understanding of phosphatase-mediated regulation of TLR signaling could provide novel access points to mitigate excessive immune activation and to modulate innate immune signaling.![]() Video Abstract
Collapse
Affiliation(s)
- Clovis H T Seumen
- Lehrstuhl Zellbiologie, Universität Konstanz, Universitätsstraße 10, Postablage 621, 78457, Konstanz, Germany
| | - Tanja M Grimm
- Lehrstuhl Zellbiologie, Universität Konstanz, Universitätsstraße 10, Postablage 621, 78457, Konstanz, Germany.,Konstanz Research School Chemical Biology, Universität Konstanz, 78457, Konstanz, Germany
| | - Christof R Hauck
- Lehrstuhl Zellbiologie, Universität Konstanz, Universitätsstraße 10, Postablage 621, 78457, Konstanz, Germany. .,Konstanz Research School Chemical Biology, Universität Konstanz, 78457, Konstanz, Germany.
| |
Collapse
|
8
|
Lee YH, Im E, Hyun M, Park J, Chung KC. Protein phosphatase PPM1B inhibits DYRK1A kinase through dephosphorylation of pS258 and reduces toxic tau aggregation. J Biol Chem 2021; 296:100245. [PMID: 33380426 PMCID: PMC7948726 DOI: 10.1074/jbc.ra120.015574] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/18/2020] [Accepted: 12/30/2020] [Indexed: 11/06/2022] Open
Abstract
Down syndrome (DS) is mainly caused by an extra copy of chromosome 21 (trisomy 21), and patients display a variety of developmental symptoms, including characteristic facial features, physical growth delay, intellectual disability, and neurodegeneration (i.e., Alzheimer's disease; AD). One of the pathological hallmarks of AD is insoluble deposits of neurofibrillary tangles (NFTs) that consist of hyperphosphorylated tau. The human DYRK1A gene is mapped to chromosome 21, and the protein is associated with the formation of inclusion bodies in AD. For example, DYRK1A directly phosphorylates multiple serine and threonine residues of tau, including Thr212. However, the mechanism underpinning DYRK1A involvement in Trisomy 21-related pathological tau aggregation remains unknown. Here, we explored a novel regulatory mechanism of DYRK1A and subsequent tau pathology through a phosphatase. Using LC-MS/MS technology, we analyzed multiple DYRK1A-binding proteins, including PPM1B, a member of the PP2C family of Ser/Thr protein phosphatases, in HEK293 cells. We found that PPM1B dephosphorylates DYRK1A at Ser258, contributing to the inhibition of DYRK1A activity. Moreover, PPM1B-mediated dephosphorylation of DYRK1A reduced tau phosphorylation at Thr212, leading to inhibition of toxic tau oligomerization and aggregation. In conclusion, our study demonstrates that DYRK1A autophosphorylates Ser258, the dephosphorylation target of PPM1B, and PPM1B negatively regulates DYRK1A activity. This finding also suggests that PPM1B reduces the toxic formation of phospho-tau protein via DYRK1A modulation, possibly providing a novel cellular protective mechanism to regulate toxic tau-mediated neuropathology in AD of DS.
Collapse
Affiliation(s)
- Ye Hyung Lee
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Eunju Im
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Minju Hyun
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Joongkyu Park
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Kwang Chul Chung
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea.
| |
Collapse
|
9
|
Berberine Improves Inflammatory Responses of Diabetes Mellitus in Zucker Diabetic Fatty Rats and Insulin-Resistant HepG2 Cells through the PPM1B Pathway. J Immunol Res 2020; 2020:2141508. [PMID: 32908938 PMCID: PMC7450322 DOI: 10.1155/2020/2141508] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/05/2020] [Accepted: 06/23/2020] [Indexed: 12/30/2022] Open
Abstract
Berberine (BBR), a natural compound extracted from a Chinese herb, has been shown to effectively attenuate insulin resistance (IR) and inflammation in the clinic. However, its ameliorative mechanism against IR is not well defined. This study is aimed at investigating the effect of BBR and protein phosphatase, Mg2+/Mn2+-dependent 1B (PPM1B) on IR. Biochemical measurements and liver histopathology were detected using the biochemical analyzer and HE staining in ZDF rats, respectively. Microarray analysis of liver tissues was performed, and differentially expressed gene (DEG) levels were examined by quantitative real-time PCR (qPCR) and Western blot. Additionally, the effect of BBR was also explored in HepG2-IR cells. The glucose oxidase method and the fluorescent glucose analog were used to detect glucose consumption and uptake, respectively. The PKA inhibitor H89, ELISA, qPCR, Western blot, and immunofluorescence staining were employed to estimate the expression levels of related signaling pathways. To evaluate the roles of PPM1B, HepG2-IR cells were stably infected with lentivirus targeting PPM1B. The administration of BBR drastically decreased the body weight, urine volume, blood glucose, blood urea nitrogen (BUN), CHOL, hepatic index levels, and pathologic changes and improved ALB levels in ZDF rats with PPM1B upregulation. Furthermore, BBR effectively improves glucose consumption, uptake, and inflammation in HepG2-IR cells. The knockdown of PPM1B expression aggravated the inflammatory response and glycometabolism disorder in HepG2-IR cells. Mechanistically, a reversal in the expression of cAMP, PKA, PPM1B, PPARγ, LRP1, GLUT4, NF-κB p65, JNK, pIKKβ Ser181, IKKβ, IRS-1 Ser307, IRS-1, IRS-2 Ser731, IRS-2, PI3K p85, and AKT Ser473 contributes to ameliorate IR in HepG2-IR cells with BBR treatment. Altogether, these results suggest that BBR might regulate IR progression through the regulation of the cAMP, PKA, PPM1B, PPARγ, LRP1, GLUT4, NF-κB p65, JNK, pIKKβ Ser181, IKKβ, IRS-1 Ser307, IRS-1, IRS-2 Ser731, IRS-2, PI3K p85, and AKT Ser473 expression in the liver.
Collapse
|
10
|
Ishii N, Homma T, Watanabe R, Kimura N, Ohnishi M, Kobayashi T, Fujii J. A heterozygous deficiency in protein phosphatase Ppm1b results in an altered ovulation number in mice. Mol Med Rep 2019; 19:5353-5360. [PMID: 31059097 DOI: 10.3892/mmr.2019.10194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 04/16/2019] [Indexed: 11/06/2022] Open
Abstract
Ppm1b, a metal‑dependent serine/threonine protein phosphatase, catalyzes the dephosphorylation of a variety of phosphorylated proteins. Ppm1b‑/‑ mouse embryos die at the fertilized oocyte stage, whereas Ppm1b+/‑ mice with a C57BL/6 background exhibit no phenotypic abnormalities. Because the C57BL/6 strain produces a limited number of pups, in an attempt to produce Ppm1b‑/‑ mice, congenic Ppm1b+/‑ mice with an ICR background were established, which are more fertile and gave birth to more pups. As a result, however, no Ppm1b‑/‑ offspring were obtained when pairs of Ppm1b+/‑ ICR mice were bred again. Ppm1b+/‑ male and female ICR mice were analyzed from the viewpoint of fecundity. The Ppm1b haploinsufficiency had no effect on testicular weight or the number of sperm in male mice. Despite the fact that the levels of Ppm1b protein in the ovaries of sexually mature Ppm1b+/‑ mice were decreased compared with those of Ppm1b+/+ mice, there appeared to be no significant difference in the histological appearance of the ovaries, litter sizes or plasma progesterone levels at the estrous stage. When superovulation was induced by stimulation using a hormone treatment, the number of ovulated oocytes were the same for Ppm1b+/‑ and Ppm1b+/+ mice at 4 weeks of age when the estrous cycle did not proceed, however, the number of ovulated oocytes was lower in sexually mature Ppm1b+/‑ mice at 11 weeks of age compared with Ppm1b+/+ mice in the first and the second superovulation cycles. These collective results suggest that follicle development is excessive in Ppm1b+/‑ mice, and that this leads to a partial depletion of matured follicles and a corresponding decrease in the number of ovulated oocytes.
Collapse
Affiliation(s)
- Naoki Ishii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata, Yamagata 990‑9585, Japan
| | - Takujiro Homma
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata, Yamagata 990‑9585, Japan
| | - Ren Watanabe
- Laboratory of Animal Reproduction, Graduate School of Agricultural Sciences, Yamagata University, Tsuruoka, Yamagata 997‑8555, Japan
| | - Naoko Kimura
- Laboratory of Animal Reproduction, Graduate School of Agricultural Sciences, Yamagata University, Tsuruoka, Yamagata 997‑8555, Japan
| | - Motoko Ohnishi
- Department of Biological Chemistry, College of Bioscience and Biotechnology, Chubu University, Kasugai, Aichi 87‑8501, Japan
| | - Takayasu Kobayashi
- Center for Gene Research, Tohoku University, Sendai, Miyagi 980‑8575, Japan
| | - Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata, Yamagata 990‑9585, Japan
| |
Collapse
|
11
|
The Cellular Senescence-Inhibited Gene Is Essential for PPM1A Myristoylation To Modulate Transforming Growth Factor β Signaling. Mol Cell Biol 2018; 38:MCB.00414-18. [PMID: 30201805 DOI: 10.1128/mcb.00414-18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 09/05/2018] [Indexed: 12/16/2022] Open
Abstract
The cellular senescence-inhibited gene (CSIG) is implicated in important biological processes, including cellular senescence and apoptosis. Our work showed that CSIG is involved in the myristoylation of the serine/threonine protein phosphatase PPM1A. Previous research has shown that myristoylation is necessary for PPM1A to dephosphorylate Smad2 and Smad3. However, the control and the biological significance of the myristoylation remain poorly understood. In this study, we found that CSIG knockdown disturbs PPM1A myristoylation and reduces the dephosphorylation by PPM1A of its substrate Smad2. By regulating PPM1A myristoylation, CSIG is involved in modulating the signaling of transforming growth factor β (TGF-β). Further study of the mechanism indicated that CSIG facilitates the interaction between N-myristoyltransferase 1 (NMT1) and PPM1A. Taking the data together, we found that CSIG is a regulator of PPM1A myristoylation and TGF-β signaling. By promoting the myristoylation of PPM1A, CSIG enhanced the phosphatase activity of PPM1A and further inhibited TGF-β signaling. This work not only extends the biological significance of CSIG but also provides new ideas and a reference for the study of the regulatory mechanism of myristoylation.
Collapse
|
12
|
Narla G, Sangodkar J, Ryder CB. The impact of phosphatases on proliferative and survival signaling in cancer. Cell Mol Life Sci 2018; 75:2695-2718. [PMID: 29725697 PMCID: PMC6023766 DOI: 10.1007/s00018-018-2826-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/24/2018] [Accepted: 04/23/2018] [Indexed: 02/06/2023]
Abstract
The dynamic and stringent coordination of kinase and phosphatase activity controls a myriad of physiologic processes. Aberrations that disrupt the balance of this interplay represent the basis of numerous diseases. For a variety of reasons, early work in this area portrayed kinases as the dominant actors in these signaling events with phosphatases playing a secondary role. In oncology, these efforts led to breakthroughs that have dramatically altered the course of certain diseases and directed vast resources toward the development of additional kinase-targeted therapies. Yet, more recent scientific efforts have demonstrated a prominent and sometimes driving role for phosphatases across numerous malignancies. This maturation of the phosphatase field has brought with it the promise of further therapeutic advances in the field of oncology. In this review, we discuss the role of phosphatases in the regulation of cellular proliferation and survival signaling using the examples of the MAPK and PI3K/AKT pathways, c-Myc and the apoptosis machinery. Emphasis is placed on instances where these signaling networks are perturbed by dysregulation of specific phosphatases to favor growth and persistence of human cancer.
Collapse
Affiliation(s)
| | - Jaya Sangodkar
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | |
Collapse
|
13
|
Miller RE, Uwamahoro N, Park JH. PPM1B depletion in U2OS cells supresses cell growth through RB1-E2F1 pathway and stimulates bleomycin-induced cell death. Biochem Biophys Res Commun 2018; 500:391-397. [DOI: 10.1016/j.bbrc.2018.04.084] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 12/28/2022]
|
14
|
Meeusen B, Janssens V. Tumor suppressive protein phosphatases in human cancer: Emerging targets for therapeutic intervention and tumor stratification. Int J Biochem Cell Biol 2017; 96:98-134. [PMID: 29031806 DOI: 10.1016/j.biocel.2017.10.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 10/04/2017] [Accepted: 10/05/2017] [Indexed: 02/06/2023]
Abstract
Aberrant protein phosphorylation is one of the hallmarks of cancer cells, and in many cases a prerequisite to sustain tumor development and progression. Like protein kinases, protein phosphatases are key regulators of cell signaling. However, their contribution to aberrant signaling in cancer cells is overall less well appreciated, and therefore, their clinical potential remains largely unexploited. In this review, we provide an overview of tumor suppressive protein phosphatases in human cancer. Along their mechanisms of inactivation in defined cancer contexts, we give an overview of their functional roles in diverse signaling pathways that contribute to their tumor suppressive abilities. Finally, we discuss their emerging roles as predictive or prognostic markers, their potential as synthetic lethality targets, and the current feasibility of their reactivation with pharmacologic compounds as promising new cancer therapies. We conclude that their inclusion in clinical practice has obvious potential to significantly improve therapeutic outcome in various ways, and should now definitely be pushed forward.
Collapse
Affiliation(s)
- Bob Meeusen
- Laboratory of Protein Phosphorylation & Proteomics, Dept. of Cellular & Molecular Medicine, Faculty of Medicine, KU Leuven & Leuven Cancer Institute (LKI), KU Leuven, Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation & Proteomics, Dept. of Cellular & Molecular Medicine, Faculty of Medicine, KU Leuven & Leuven Cancer Institute (LKI), KU Leuven, Belgium.
| |
Collapse
|
15
|
Salminen A, Kaarniranta K, Kauppinen A. Age-related changes in AMPK activation: Role for AMPK phosphatases and inhibitory phosphorylation by upstream signaling pathways. Ageing Res Rev 2016; 28:15-26. [PMID: 27060201 DOI: 10.1016/j.arr.2016.04.003] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 03/18/2016] [Accepted: 04/05/2016] [Indexed: 02/07/2023]
Abstract
AMP-activated protein kinase (AMPK) is a fundamental regulator of energy metabolism, stress resistance, and cellular proteostasis. AMPK signaling controls an integrated signaling network which is involved in the regulation of healthspan and lifespan e.g. via FoxO, mTOR/ULK1, CRCT-1/CREB, and SIRT1 signaling pathways. Several studies have demonstrated that the activation capacity of AMPK signaling declines with aging, which impairs the maintenance of efficient cellular homeostasis and enhances the aging process. However, it seems that the aging process affects AMPK activation in a context-dependent manner since occasionally, it can also augment AMPK activation, possibly attributable to the type of insult and tissue homeostasis. Three protein phosphatases, PP1, PP2A, and PP2C, inhibit AMPK activation by dephosphorylating the Thr172 residue of AMPKα, required for AMPK activation. In addition, several upstream signaling pathways can phosphorylate Ser/Thr residues in the β/γ interaction domain of the AMPKα subunit that subsequently blocks the activation of AMPK. These inhibitory pathways include the insulin/AKT, cyclic AMP/PKA, and RAS/MEK/ERK pathways. We will examine the evidence whether the efficiency of AMPK responsiveness declines during the aging process. Next, we will review the mechanisms involved in curtailing the activation of AMPK. Finally, we will elucidate the potential age-related changes in the inhibitory regulation of AMPK signaling that might be a part of the aging process.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, P.O. Box 100, FI-70029 KYS, Finland
| | - Anu Kauppinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| |
Collapse
|
16
|
Multi-Compartmentalisation in the MAPK Signalling Pathway Contributes to the Emergence of Oscillatory Behaviour and to Ultrasensitivity. PLoS One 2016; 11:e0156139. [PMID: 27243235 PMCID: PMC4887093 DOI: 10.1371/journal.pone.0156139] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 05/10/2016] [Indexed: 12/20/2022] Open
Abstract
Signal transduction through the Mitogen Activated Protein Kinase (MAPK) pathways is evolutionarily highly conserved. Many cells use these pathways to interpret changes to their environment and respond accordingly. The pathways are central to triggering diverse cellular responses such as survival, apoptosis, differentiation and proliferation. Though the interactions between the different MAPK pathways are complex, nevertheless, they maintain a high level of fidelity and specificity to the original signal. There are numerous theories explaining how fidelity and specificity arise within this complex context; spatio-temporal regulation of the pathways and feedback loops are thought to be very important. This paper presents an agent based computational model addressing multi-compartmentalisation and how this influences the dynamics of MAPK cascade activation. The model suggests that multi-compartmentalisation coupled with periodic MAPK kinase (MAPKK) activation may be critical factors for the emergence of oscillation and ultrasensitivity in the system. Finally, the model also establishes a link between the spatial arrangements of the cascade components and temporal activation mechanisms, and how both contribute to fidelity and specificity of MAPK mediated signalling.
Collapse
|
17
|
Expression and gene doses changes of the p53-regulator PPM1D in meningiomas: a role in meningioma progression? Brain Tumor Pathol 2016; 33:191-9. [DOI: 10.1007/s10014-016-0252-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 02/14/2016] [Indexed: 01/07/2023]
|
18
|
Yokota T, Wang Y. p38 MAP kinases in the heart. Gene 2015; 575:369-376. [PMID: 26390817 DOI: 10.1016/j.gene.2015.09.030] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 07/31/2015] [Accepted: 09/15/2015] [Indexed: 12/28/2022]
Abstract
p38 kinases are members of the mitogen-activated protein kinases (MAPK) with established contribution to a wide range of signaling pathways and different biological processes. The prototypic p38 MAPK, p38α was originally identified as an essential signaling kinase for inflammatory cytokine production Extensive studies have now revealed that p38s have critical roles in many different tissues far beyond immune regulation and inflammatory responses. In this review, we will focus on the structure and molecular biology of p38s, and their specific roles in heart, especially regarding myocyte proliferation, apoptosis, and hypertrophic responses.
Collapse
Affiliation(s)
- Tomohiro Yokota
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Physiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Yibin Wang
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Physiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
19
|
Protein phosphatase magnesium dependent 1A governs the wound healing-inflammation-angiogenesis cross talk on injury. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2936-50. [PMID: 25196308 DOI: 10.1016/j.ajpath.2014.07.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 06/25/2014] [Accepted: 07/18/2014] [Indexed: 01/09/2023]
Abstract
Protein phosphatase magnesium dependent 1A (PPM1A) has been implicated in fibrosis and skin wounding. We generated PPM1A knockout mice to study the role of PPM1A in the wound healing-inflammation-angiogenesis cross talk. The role of PPM1A in these processes was studied using the ocular alkali burn model system. In the injured cornea the absence of PPM1A led to enhanced inflammatory response, stromal keratocyte transactivation, fibrosis, increased p38 mitogen-activated protein kinase phosphorylation, elevated expression of transforming growth factor-β-related genes (including Acta2, TGF-β, Col1, MMP9, and VEGF) and subsequently to neovascularization. Augmented angiogenesis in the absence of PPM1A is a general process occurring in vivo in PPM1A knockout mice upon subcutaneous Matrigel injection and ex vivo in aortic ring Matrigel cultures. Using primary keratocyte cultures and various experimental approaches, we found that phospho-p38 is a favored PPM1A substrate and that by its dephosphorylation PPM1A participates in the regulation of the transforming growth factor-β signaling cascade, the hallmark of inflammation and the angiogenic process. On the whole, the studies presented here position PPM1A as a new player in the wound healing-inflammation-angiogenesis axis in mouse, reveal its crucial role in homeostasis on injury, and highlight its potential as a therapeutic mediator in pathologic conditions, such as inflammation and angiogenesis disorders, including cancer.
Collapse
|
20
|
Park JH, Hale TK, Smith RJ, Yang T. PPM1B depletion induces premature senescence in human IMR-90 fibroblasts. Mech Ageing Dev 2014; 138:45-52. [DOI: 10.1016/j.mad.2014.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 03/10/2014] [Accepted: 03/15/2014] [Indexed: 01/23/2023]
|
21
|
Vanderstraete M, Gouignard N, Cailliau K, Morel M, Hahnel S, Leutner S, Beckmann S, Grevelding CG, Dissous C. Venus kinase receptors control reproduction in the platyhelminth parasite Schistosoma mansoni. PLoS Pathog 2014; 10:e1004138. [PMID: 24875530 PMCID: PMC4038586 DOI: 10.1371/journal.ppat.1004138] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 04/08/2014] [Indexed: 11/25/2022] Open
Abstract
The Venus Kinase Receptor (VKR) is a single transmembrane molecule composed of an intracellular tyrosine kinase domain close to that of insulin receptor and an extracellular Venus Flytrap (VFT) structure similar to the ligand binding domain of many class C G Protein Coupled Receptors. This receptor tyrosine kinase (RTK) was first discovered in the platyhelminth parasite Schistosoma mansoni, then in a large variety of invertebrates. A single vkr gene is found in most genomes, except in S. mansoni in which two genes Smvkr1 and Smvkr2 exist. VKRs form a unique family of RTKs present only in invertebrates and their biological functions are still to be discovered. In this work, we show that SmVKRs are expressed in the reproductive organs of S. mansoni, particularly in the ovaries of female worms. By transcriptional analyses evidence was obtained that both SmVKRs fulfill different roles during oocyte maturation. Suppression of Smvkr expression by RNA interference induced spectacular morphological changes in female worms with a strong disorganization of the ovary, which was dominated by the presence of primary oocytes, and a defect of egg formation. Following expression in Xenopus oocytes, SmVKR1 and SmVKR2 receptors were shown to be activated by distinct ligands which are L-Arginine and calcium ions, respectively. Signalling analysis in Xenopus oocytes revealed the capacity of SmVKRs to activate the PI3K/Akt/p70S6K and Erk MAPK pathways involved in cellular growth and proliferation. Additionally, SmVKR1 induced phosphorylation of JNK (c-Jun N-terminal kinase). Activation of JNK by SmVKR1 was supported by the results of yeast two-hybrid experiments identifying several components of the JNK pathway as specific interacting partners of SmVKR1. In conclusion, these results demonstrate the functions of SmVKR in gametogenesis, and particularly in oogenesis and egg formation. By eliciting signalling pathways potentially involved in oocyte proliferation, growth and migration, these receptors control parasite reproduction and can therefore be considered as potential targets for anti-schistosome therapies. Schistosomiasis is a chronic, debilitating disease affecting more than 200 million people in the world caused by parasitic flatworms of the genus Schistosoma. Pathology is mainly due to massive egg production by parasites and formation of granulomas around the eggs trapped in liver and different organs. Therefore, targeting the molecular processes responsible for gonad development or egg production in schistosomes appears as a valuable strategy to reduce pathogenesis and dissemination of schistosomiasis. In the present study, we investigated the importance of Venus Kinase Receptors (VKRs) which are unusual receptor tyrosine kinases (RTKs) with an extracellular Venus Flytrap (VFT) ligand-binding domain in the control of reproduction of schistosomes. SmVKRs are expressed in female ovaries of Schistosoma mansoni and the knock-down of their expression provoked dramatic alterations of the oocyte content in ovaries and reduction of egg formation. SmVKRs were also shown to activate different signalling pathways potentially involved in oocyte proliferation, growth and migration. Therefore our results demonstrate that VKRs are essential actors of oogenesis and egg formation in S. mansoni. Moreover, their presence in a large variety of invertebrate species including other helminth parasites and insect parasite vectors can open new perspectives in the control of various vector-borne infectious diseases.
Collapse
Affiliation(s)
- Mathieu Vanderstraete
- Center for Infection and Immunity of Lille, Inserm U1019, CNRS-UMR 8204, University Lille Nord de France, Institut Pasteur de Lille, Lille, France
| | - Nadège Gouignard
- Center for Infection and Immunity of Lille, Inserm U1019, CNRS-UMR 8204, University Lille Nord de France, Institut Pasteur de Lille, Lille, France
| | - Katia Cailliau
- EA 4479, IFR 147, Universite Lille 1 Sciences et Technologies, Villeneuve d'Ascq, France
| | - Marion Morel
- Center for Infection and Immunity of Lille, Inserm U1019, CNRS-UMR 8204, University Lille Nord de France, Institut Pasteur de Lille, Lille, France
| | - Steffen Hahnel
- Institute for Parasitology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Silke Leutner
- Institute for Parasitology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Svenja Beckmann
- Institute for Parasitology, Justus-Liebig-University Giessen, Giessen, Germany
| | | | - Colette Dissous
- Center for Infection and Immunity of Lille, Inserm U1019, CNRS-UMR 8204, University Lille Nord de France, Institut Pasteur de Lille, Lille, France
- * E-mail:
| |
Collapse
|
22
|
Yu Y, Li J, Wan Y, Lu J, Gao J, Huang C. GADD45α induction by nickel negatively regulates JNKs/p38 activation via promoting PP2Cα expression. PLoS One 2013; 8:e57185. [PMID: 23536762 PMCID: PMC3594291 DOI: 10.1371/journal.pone.0057185] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Accepted: 01/15/2013] [Indexed: 12/20/2022] Open
Abstract
Growth arrest and DNA damage (GADD) 45α is a member of GADD inducible gene family, and is inducible in cell response to oxidative stress. GADD45α upregulation induces MKK4/JNK activation in some published experimental systems. However, we found here that the depletion of GADD45α (GADD45α−/−) in mouse embryonic fibroblasts (MEFs) resulted in an increase in the phosphorylation of MKK4/7, MKK3/6 and consequently specific up-regulated the activation of JNK/p38 and their downstream transcription factors, such as c-Jun and ATF2, in comparison to those in GADD45α+/+ MEFs cell following nickel exposure. This up-regulation of MKK-JNK/p38 pathway in GADD45α−/− cell could be rescued by the reconstitutional expression of HA-GADD45α in GADD45α−/− MEFs, GADD45α−/−(HA-GADD45α). Subsequent studies indicated that GADD45α deletion repressed expression of PP2Cα, the phosphotase of MKK3/6 and MKK4/7, whereas ectopic expression of HA-PP2Cα in GADD45α−/− cells attenuated activation of MKK3/6-p38 and MKK4/7-JNK pathways. Collectively, our results demonstrate a novel function and mechanism responsible for GADD45α regulation of MKK/MAPK pathway, further provides insight into understanding the big picture of GADD45α in the regulation of cellular responses to oxidative stress and environmental carcinogens.
Collapse
Affiliation(s)
- Yonghui Yu
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, United States of America
- Oversea Laboratory, Center for Medical Research, Wuhan University, Wuhan, Hubei, China
| | - Jingxia Li
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, United States of America
| | - Yu Wan
- Oversea Laboratory, Center for Medical Research, Wuhan University, Wuhan, Hubei, China
| | - Jianyi Lu
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical College, Wenzhou, Zhejiang, China
| | - Jimin Gao
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical College, Wenzhou, Zhejiang, China
- * E-mail: (JG); (CH)
| | - Chuanshu Huang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, United States of America
- * E-mail: (JG); (CH)
| |
Collapse
|
23
|
N-Myristoylation is essential for protein phosphatases PPM1A and PPM1B to dephosphorylate their physiological substrates in cells. Biochem J 2013; 449:741-9. [PMID: 23088624 DOI: 10.1042/bj20121201] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PPM [metal-dependent protein phosphatase, formerly called PP2C (protein phosphatase 2C)] family members play essential roles in regulating a variety of signalling pathways. While searching for protein phosphatase(s) that act on AMPK (AMP-activated protein kinase), we found that PPM1A and PPM1B are N-myristoylated and that this modification is essential for their ability to dephosphorylate the α subunit of AMPK (AMPKα) in cells. N-Myristoylation was also required for two other functions of PPM1A and PPM1B in cells. Although a non-myristoylated mutation (G2A) of PPM1A and PPM1B prevented membrane association, this relocalization did not likely cause the decreased activity towards AMPKα. In in vitro experiments, the G2A mutants exhibited reduced activities towards AMPKα, but much higher specific activity against an artificial substrate, PNPP (p-nitrophenyl phosphate), compared with the wild-type counterparts. Taken together, the results of the present study suggest that N-myristoylation of PPM1A and PPM1B plays a key role in recognition of their physiological substrates in cells.
Collapse
|
24
|
Protein phosphatase 1 inhibits p53 signaling by dephosphorylating and stabilizing Mdmx. Cell Signal 2012; 25:796-804. [PMID: 23277204 DOI: 10.1016/j.cellsig.2012.12.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 12/16/2012] [Accepted: 12/21/2012] [Indexed: 12/17/2022]
Abstract
The activation and stabilization of the p53 protein play a major role in the DNA damage response. Protein levels of p53 are tightly controlled by transcriptional regulation and a number of positive and negative posttranslational modifiers, including kinases, phosphatases, E3 ubiquitin ligases, deubiquitinases, acetylases and deacetylases. One of the primary p53 regulators is Mdmx. Despite its RING domain and structural similarity with Mdm2, Mdmx does not have an intrinsic ligase activity, but inhibits the transcriptional activity of p53. Previous studies reported that Mdmx is phosphorylated and destabilized in response to DNA damage stress. Three phosphorylation sites identified are Ser342, Ser367, and Ser403. In the present study, we identify protein phosphatase 1 (PP1) as a negative regulator in the p53 signaling pathway. PP1 directly interacts with Mdmx and specifically dephosphorylates Mdmx at Ser367. The dephosphorylation of Mdmx increases its stability and thereby inhibits p53 activity. Our results suggest that PP1 is a crucial component in the ATM-Chk2-p53 signaling pathway.
Collapse
|
25
|
Zhao Y, Liang L, Fan Y, Sun S, An L, Shi Z, Cheng J, Jia W, Sun W, Mori-Akiyama Y, Zhang H, Fu S, Yang J. PPM1B negatively regulates antiviral response via dephosphorylating TBK1. Cell Signal 2012; 24:2197-204. [PMID: 22750291 PMCID: PMC3432707 DOI: 10.1016/j.cellsig.2012.06.017] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 06/25/2012] [Indexed: 01/19/2023]
Abstract
The production of type I interferon must be tightly regulated and aberrant production of type I interferon is harmful or even fatal to the host. TBK1 phosphorylation at Ser172 plays an essential role in TBK1-mediated antiviral response. However, how TBK1 activity is negatively regulated remains poorly understood. Using a functional genomics approach, we have identified PPM1B as a TBK1 phosphatase. PPM1B dephosphorylates TBK1 in vivo and in vitro. PPM1B wild-type but not its phosphatase-deficient R179G mutant inhibits TBK1-mediated antiviral response and facilitates VSV replication in the cells. Viral infection induces association of PPM1B with TBK1 in a transient fashion in the cells. Conversely, suppression of PPM1B expression enhances virus-induced IRF3 phosphorylation and IFNβ production. Our study identifies a previously unrecognized role for PPM1B in the negative regulation of antiviral response by acting as a TBK1 phosphatase.
Collapse
Affiliation(s)
- Yanling Zhao
- Laboratory of Medical Genetics, Harbin Medical University, Harbin 150081, China
- Texas Children’s Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Li Liang
- Department of Tumor Chemotherapy and Radiation Sickness in Peking University Third Hospital, Beijing 100191, China
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Yihui Fan
- Texas Children’s Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Surong Sun
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Lei An
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Zhongcheng Shi
- Department of Pathology and Immunology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Jin Cheng
- Texas Children’s Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Microbiology, Peking University Health Science Center, Beijing, 100191, China
| | - Wei Jia
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Pathology, Shihezi University School of Medicine, Shihezi 832002, China
| | - Wenjing Sun
- Laboratory of Medical Genetics, Harbin Medical University, Harbin 150081, China
- Texas Children’s Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Yuko Mori-Akiyama
- Department of Pathology and Immunology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Hong Zhang
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Songbin Fu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin 150081, China
| | - Jianhua Yang
- Texas Children’s Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
26
|
Abstract
Cell death is regulated by a myriad of intracellular molecular pathways, with many involving protein phosphorylation and dephosphorylation. In this review, we will focus on Ser/Thr phosphatases-mediated regulation in cell apoptosis as well as on their potential roles in cell necrosis. The emerging functional importance of Ser/Thr protein phosphatases in cell death regulation adds new dimension to the signaling mechanisms of cellular function, physiology, and diseases.
Collapse
Affiliation(s)
- Haipeng Sun
- Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Department of Pathophysiology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | |
Collapse
|
27
|
Perry DM, Kitatani K, Roddy P, El-Osta M, Hannun YA. Identification and characterization of protein phosphatase 2C activation by ceramide. J Lipid Res 2012; 53:1513-21. [PMID: 22615346 DOI: 10.1194/jlr.m025395] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Ceramide is a bioactive sphingolipid with many associated biological outcomes, yet there is a significant gap in our current understanding of how ceramide mediates these processes. Previously, ceramide has been shown to activate protein phosphatase (PP) 1 and 2A. While continuing this line of work, a late fraction from a Mono-Q column was consistently observed to be activated by ceramide, yet PP1 and PP2A were undetectable in this fraction. Proteomic analysis of this fraction revealed the identity of the phosphatase to be PP2Cγ/PPM1G. This was consistent with our findings that PP2Cγ 1-eluted in a high salt fraction due to its strongly acidic domain, and 2-was insensitive to okadaic acid. Further characterization was performed with PP2Cα, which showed robust activation by C(6)-ceramide. Activation was specific for the erythro conformation of ceramide and the presence of the acyl chain and hydroxyl group at the first carbon. In order to demonstrate more physiological activation of PP2Cα by ceramide, phospho-p38δ was utilized as substrate. Indeed, PP2Cα induced the dephosphorylation of p38δ only in the presence of C(16)-ceramide. Taken together, these results show that the PP2C family of phosphatases is activated by ceramide, which may have important consequences in mediating the biological effects of ceramide.
Collapse
Affiliation(s)
- David M Perry
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | | | |
Collapse
|
28
|
Shohat M, Ben-Meir D, Lavi S. Protein phosphatase magnesium dependent 1A (PPM1A) plays a role in the differentiation and survival processes of nerve cells. PLoS One 2012; 7:e32438. [PMID: 22384250 PMCID: PMC3288098 DOI: 10.1371/journal.pone.0032438] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 01/27/2012] [Indexed: 12/22/2022] Open
Abstract
The serine/threonine phosphatase type 2C (PPM1A) has a broad range of substrates, and its role in regulating stress response is well established. We have investigated the involvement of PPM1A in the survival and differentiation processes of PC6-3 cells, a subclone of the PC12 cell line. This cell line can differentiate into neuron like cells upon exposure to nerve growth factor (NGF). Overexpression of PPM1A in naive PC6-3 cells caused cell cycle arrest at the G2/M phase followed by apoptosis. Interestingly, PPM1A overexpression did not affect fully differentiated cells. Using PPM1A overexpressing cells and PPM1A knockdown cells, we show that this phosphatase affects NGF signaling in PC6-3 cells and is engaged in neurite outgrowth. In addition, the ablation of PPM1A interferes with NGF-induced growth arrest during differentiation of PC6-3 cells.
Collapse
Affiliation(s)
| | | | - Sara Lavi
- Department of Cell Research and Immunology, Tel Aviv University, Tel Aviv, Israel
- * E-mail:
| |
Collapse
|
29
|
Miyagi T, Kikuchi K, Tamura S. Shigeru Tsuiki: a pioneer in the research fields of complex carbohydrates and protein phosphatases. J Biochem 2011; 150:483-90. [PMID: 22039278 DOI: 10.1093/jb/mvr045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Dr Tsuiki made three major contributions during his illustrious career as a biochemist. First, he developed the procedure for mucin isolation from bovine submaxillary glands. His work became the basis for mucin biochemistry. Second, he identified four distinct molecular species of mammalian sialidase. Subsequent studies based on his work led to the discovery that sialidase plays a unique role as an intracellular signalling factor involved in the regulation of a variety of cellular functions. Finally, he established the molecular basis for the diversity of mammalian protein phosphatases through protein purification and molecular cloning. His work prompted the functional studies of protein phosphatases.
Collapse
Affiliation(s)
- Taeko Miyagi
- Department of Cancer Glycosylation Research, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Komatsushima, Aoba-ku, Sendai, Japan
| | | | | |
Collapse
|
30
|
RASSF7 negatively regulates pro-apoptotic JNK signaling by inhibiting the activity of phosphorylated-MKK7. Cell Death Differ 2010; 18:645-55. [PMID: 21278800 DOI: 10.1038/cdd.2010.137] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Members of the Ras-association domain family (RASSF) of proteins influence apoptosis and cell cycling but little is known about the mechanisms. Here, we show that RASSF7 interacts with N-Ras and mitogen-activated protein kinase kinase 7 (MKK7) to negatively regulate c-Jun N-terminal kinase (JNK) signaling. Stress-induced JNK activation and apoptosis were markedly enhanced in cells depleted of RASSF7 or N-Ras by RNAi knockdown. An interaction with RASSF7 promoted the phosphorylated state of MKK7 but inhibited this kinase's ability to activate JNK. RASSF7 required its RA domain for both interaction with GTP-bound N-Ras and the anti-apoptotic response to stress stimuli. Following prolonged stress, however, RASSF7's anti-apoptotic effect was eliminated because of degradation of RASSF7 protein via the ubiquitin-proteasome pathway. Our results indicate that RASSF7 acts in concert with N-Ras to constitute a stress-sensitive temporary mechanism of apoptotic regulation. With initial stress, RASSF7/N-Ras promotes cell survival by inhibiting the MKK7/JNK pathway. However, with prolonged stress, RASSF7 protein undergoes degradation that allows cell death signaling to proceed. Our findings may account for the association of elevated RASSF7 with tumorigenesis.
Collapse
|
31
|
Wang M, Wang D, Lin L, Hong H. Protein profiles in zebrafish (Danio rerio) brains exposed to chronic microcystin-LR. CHEMOSPHERE 2010; 81:716-724. [PMID: 20800265 DOI: 10.1016/j.chemosphere.2010.07.061] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 07/20/2010] [Accepted: 07/28/2010] [Indexed: 05/29/2023]
Abstract
Microcystin-LR (MCLR) is a commonly encountered blue-green algal hepatotoxin and a known inhibitor of cellular protein phosphatase (PP), however, little is known about its neurotoxicity. This study investigated the protein profiles of zebrafish (Danio rerio) brains chronically exposed to MCLR concentrations (2 or 20 μg L(-1)) using the proteomic approach. The results showed that MCLR strikingly enhanced toxin accumulation and the PP activity in zebrafish brains after 30 d exposure. Comparison of two-dimensional electrophoresis protein profiles of MCLR exposed and non-exposed zebrafish brains revealed that the abundance of 30 protein spots was remarkably altered in response to MCLR exposure. These proteins are involved in cytoskeleton assembly, macromolecule metabolism, oxidative stress, signal transduction, and other functions (e.g. transporting, protein degradation, apoptosis and translation), indicating that MCLR toxicity in the fish brain is complex and diverse. The chronic neurotoxicity of MCLR might initiate the PP pathway via an upregulation of PP2C in the zebrafish brain, in addition to the reactive oxygen species pathway. Additionally, the increase of vitellogenin abundance in MCLR exposed zebrafish brains suggested that MCLR might mimic the effects of endocrine disrupting chemicals. This study demonstrated that MCLR causes neurotoxicity in zebrafish at the proteomic level, which provides a new insight into MCLR toxicity in aquatic organisms and human beings.
Collapse
Affiliation(s)
- Minghua Wang
- State Key Laboratory of Marine Environmental Science/Environmental Science Research Center, Xiamen University, Xiamen 361005, People's Republic of China
| | | | | | | |
Collapse
|
32
|
Zhang B, Zhou Z, Lin H, Lv X, Fu J, Lin P, Zhu C, Wang H. Protein phosphatase 1A (PPM1A) is involved in human cytotrophoblast cell invasion and migration. Histochem Cell Biol 2009; 132:169-79. [PMID: 19404668 DOI: 10.1007/s00418-009-0601-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2009] [Indexed: 11/25/2022]
Abstract
Trophoblast invasion is crucial for embryo implantation and placentation. Excessive trophoblast invasion leads to hydatidiform moles and choriocarcinoma. PPM1A is a phosphatase which dephosphorylates and inactivates a broad range of substrates, including TGF-beta, MAP kinases, p38 and JNK kinase cascades, and is involved in tumor suppression. The objective of this study was to investigate the expression of PPM1A in normal and malignant human placenta and its role in trophoblast invasion, which shares many similarities with invasion of tumor cells. By Western blotting and immunocytochemistry, significantly higher expression of PPM1A in human placental villi at term was found as compared with that during the first trimester. Furthermore, the expression level of PPM1A protein in hydatidiform moles was lower compared with that during normal pregnancy. We further investigated the function of PPM1A in extravillous trophoblast cell line HTR8/SVneo. Transwell migration and Matrigel invasion assays demonstrated that PPM1A siRNA significantly promoted the motility and invasiveness of the cells. Gelatin zymography showed that knockdown of PPM1A with siRNA elevated the expression of pro-matrix metalloproteinase pro-(MMP)-9, but down-regulated tissue inhibitors of metalloproteinases (TIMP)-2. The present data indicate that PPM1A plays a critical role in the regulation of normal placentation by inhibiting trophoblast migration and invasion.
Collapse
Affiliation(s)
- Baohua Zhang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang District, 100101, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Xia Y, Ongusaha P, Lee SW, Liou YC. Loss of Wip1 sensitizes cells to stress- and DNA damage-induced apoptosis. J Biol Chem 2009; 284:17428-37. [PMID: 19395378 DOI: 10.1074/jbc.m109.007823] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In response to various environmental stresses, the stress-responsive MAPKs p38 and JNK are activated and phosphorylate ATF2 and c-Jun transcription factors, thereby affecting cell-fate decision. Targeted gene disruption studies have established that JNK-c-Jun signaling plays a vital role in stress-induced apoptosis. The oncogenic phosphatase Wip1 acts as an important regulator in DNA damage pathway by dephosphorylating a spectrum of proteins including p53, p38, Chk1, Chk2, and ATM. In this study we show that Wip1 negatively regulates the activation of MKK4-JNK-c-Jun signaling during stress-induced apoptosis. The loss of Wip1 function sensitizes mouse embryonic fibroblasts to stress-induced apoptosis via the activation of both p38-ATF2 and JNK-c-Jun signaling. Here we reveal that Wip1 has dual roles in alternatively regulating stress- and DNA damage-induced apoptosis through p38/JNK MAPKs and p38/p53-dependent pathways, respectively. Our results point to Wip1 as a general regulator of apoptosis, which further supports its role in tumorigenesis.
Collapse
Affiliation(s)
- Yun Xia
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore 117543, Singapore
| | | | | | | |
Collapse
|
34
|
Krieglstein J, Hufnagel B, Dworak M, Schwarz S, Kewitz T, Reinbold M, Klumpp S. Influence of various fatty acids on the activity of protein phosphatase type 2C and apoptosis of endothelial cells and macrophages. Eur J Pharm Sci 2008; 35:397-403. [DOI: 10.1016/j.ejps.2008.08.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Revised: 08/20/2008] [Accepted: 08/20/2008] [Indexed: 10/21/2022]
|
35
|
Xue T, Wang D, Zhang S, Ehlting J, Ni F, Jakab S, Zheng C, Zhong Y. Genome-wide and expression analysis of protein phosphatase 2C in rice and Arabidopsis. BMC Genomics 2008; 9:550. [PMID: 19021904 PMCID: PMC2612031 DOI: 10.1186/1471-2164-9-550] [Citation(s) in RCA: 220] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2008] [Accepted: 11/20/2008] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The protein phosphatase 2Cs (PP2Cs) from various organisms have been implicated to act as negative modulators of protein kinase pathways involved in diverse environmental stress responses and developmental processes. A genome-wide overview of the PP2C gene family in plants is not yet available. RESULTS A comprehensive computational analysis identified 80 and 78 PP2C genes in Arabidopsis thaliana (AtPP2Cs) and Oryza sativa (OsPP2Cs), respectively, which denotes the PP2C gene family as one of the largest families identified in plants. Phylogenic analysis divided PP2Cs in Arabidopsis and rice into 13 and 11 subfamilies, respectively, which are supported by the analyses of gene structures and protein motifs. Comparative analysis between the PP2C genes in Arabidopsis and rice identified common and lineage-specific subfamilies and potential 'gene birth-and-death' events. Gene duplication analysis reveals that whole genome and chromosomal segment duplications mainly contributed to the expansion of both OsPP2Cs and AtPP2Cs, but tandem or local duplication occurred less frequently in Arabidopsis than rice. Some protein motifs are widespread among the PP2C proteins, whereas some other motifs are specific to only one or two subfamilies. Expression pattern analysis suggests that 1) most PP2C genes play functional roles in multiple tissues in both species, 2) the induced expression of most genes in subfamily A by diverse stimuli indicates their primary role in stress tolerance, especially ABA response, and 3) the expression pattern of subfamily D members suggests that they may constitute positive regulators in ABA-mediated signaling pathways. The analyses of putative upstream regulatory elements by two approaches further support the functions of subfamily A in ABA signaling, and provide insights into the shared and different transcriptional regulation machineries in dicots and monocots. CONCLUSION This comparative genome-wide overview of the PP2C family in Arabidopsis and rice provides insights into the functions and regulatory mechanisms, as well as the evolution and divergence of the PP2C genes in dicots and monocots. Bioinformatics analyses suggest that plant PP2C proteins from different subfamilies participate in distinct signaling pathways. Our results have established a solid foundation for future studies on the functional divergence in different PP2C subfamilies.
Collapse
Affiliation(s)
- Tongtong Xue
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Sun W, Yu Y, Dotti G, Shen T, Tan X, Savoldo B, Pass AK, Chu M, Zhang D, Lu X, Fu S, Lin X, Yang J. PPM1A and PPM1B act as IKKbeta phosphatases to terminate TNFalpha-induced IKKbeta-NF-kappaB activation. Cell Signal 2008; 21:95-102. [PMID: 18930133 DOI: 10.1016/j.cellsig.2008.09.012] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2008] [Accepted: 09/26/2008] [Indexed: 01/21/2023]
Abstract
IKKbeta serves as a central intermediate signaling molecule in the activation of the NF-kappaB pathway. However, the precise mechanism for the termination of IKKbeta activity is still not fully understood. Using a functional genomic approach, we have identified two protein serine/threonine phosphatases, PPM1A and PPM1B, as IKKbeta phosphatases. Overexpression of PPM1A or PPM1B results in dephosphorylation of IKKbeta at Ser177 and Ser181 and termination of IKKbeta-induced NF-kappaB activation. PPM1A and PPM1B associate with the phosphorylated form of IKKbeta, and the interaction between PPM1A/PPM1B and IKKbeta is induced by TNFalpha in a transient fashion in the cells. Furthermore, knockdown of PPM1A and PPM1B expression enhances TNFalpha-induced IKKbeta phosphorylation, NF-kappaB nuclear translocation and NF-kappaB-dependent gene expression. These data suggest that PPM1A and PPM1B play an important role in the termination of TNFalpha-mediated NF-kappaB activation through dephosphorylating and inactivating IKKbeta.
Collapse
Affiliation(s)
- Wenjing Sun
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, United States
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kaur R, Yuan X, Lu ML, Balk SP. Increased PAK6 expression in prostate cancer and identification of PAK6 associated proteins. Prostate 2008; 68:1510-6. [PMID: 18642328 DOI: 10.1002/pros.20787] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND PAK6 is a member of the p21-activated kinase (PAK) family of serine/threonine kinases that was originally cloned from prostate cancer (PCa) cells as an androgen receptor interacting protein, but its cellular distribution and functions have not been established. METHODS An affinity purified rabbit anti-PAK6 antiserum was generated to assess PAK6 protein expression. PAK6 associated proteins were identified by immunopurification of 3xFlag-tagged PAK6 followed by LC/MS/MS. RESULTS We confirmed that PAK6 protein is expressed in prostate and breast cancer cell lines. PAK6 expression in LNCaP PCa cells was not directly androgen regulated, but was markedly increased when the cells were cultured for 6-8 weeks in steroid hormone depleted medium. By immunohistochemistry, PAK6 was weakly expressed in normal prostate epithelium. Its expression was increased in primary and metastatic PCa, and was further increased in tumors that relapsed after androgen deprivation therapy. LC/MS/MS identified IQ motif containing GTPase activating protein 1 (IQGAP1) and protein phosphatase 1B (PP1B) as candidate PAK6 interacting proteins, and these findings were confirmed by coimmunoprecipitation. CONCLUSIONS These results indicate that PAK6 contributes to PCa development and progression after androgen deprivation therapy, and that it may play roles in the regulation of motility and in stress responses.
Collapse
Affiliation(s)
- Ramneet Kaur
- Department of Medicine, Cancer Biology Program, Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | |
Collapse
|
38
|
Lu X, Nguyen TA, Moon SH, Darlington Y, Sommer M, Donehower LA. The type 2C phosphatase Wip1: an oncogenic regulator of tumor suppressor and DNA damage response pathways. Cancer Metastasis Rev 2008; 27:123-35. [PMID: 18265945 PMCID: PMC2362138 DOI: 10.1007/s10555-008-9127-x] [Citation(s) in RCA: 205] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The Wild-type p53-induced phosphatase 1, Wip1 (or PPM1D), is unusual in that it is a serine/threonine phosphatase with oncogenic activity. A member of the type 2C phosphatases (PP2Cδ), Wip1 has been shown to be amplified and overexpressed in multiple human cancer types, including breast and ovarian carcinomas. In rodent primary fibroblast transformation assays, Wip1 cooperates with known oncogenes to induce transformed foci. The recent identification of target proteins that are dephosphorylated by Wip1 has provided mechanistic insights into its oncogenic functions. Wip1 acts as a homeostatic regulator of the DNA damage response by dephosphorylating proteins that are substrates of both ATM and ATR, important DNA damage sensor kinases. Wip1 also suppresses the activity of multiple tumor suppressors, including p53, ATM, p16INK4a and ARF. We present evidence that the suppression of p53, p38 MAP kinase, and ATM/ATR signaling pathways by Wip1 are important components of its oncogenicity when it is amplified and overexpressed in human cancers.
Collapse
Affiliation(s)
- Xiongbin Lu
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
39
|
Chan PM, Lim L, Manser E. PAK is regulated by PI3K, PIX, CDC42, and PP2Calpha and mediates focal adhesion turnover in the hyperosmotic stress-induced p38 pathway. J Biol Chem 2008; 283:24949-61. [PMID: 18586681 DOI: 10.1074/jbc.m801728200] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Fractionation of brain extracts and functional biochemical assays identified PP2Calpha, a serine/threonine phosphatase, as the major biochemical activity inhibiting PAK1. PP2Calpha dephosphorylated PAK1 and p38, both of which were activated upon hyperosmotic shock with the same kinetics. In comparison to growth factors, hyperosmolality was a more potent activator of PAK1. Therefore we characterize the PAK signaling pathway in the hyperosmotic shock response. Endogenous PAKs were recruited to the p38 kinase complex in a phosphorylation-dependent manner. Overexpression of a PAK inhibitory peptide or dominant negative Cdc42 revealed that p38 activation was dependent on PAK and Cdc42 activities. PAK mutants deficient in binding to Cdc42 or PAK-interacting exchange factor were not activated. Using a panel of kinase inhibitors, we identified PI3K acting upstream of PAK, which correlated with PAK repression by pTEN overexpression. RNA interference knockdown of PAK expression reduced stress-induced p38 activation and conversely, PP2Calpha knockdown increased its activation. Hyperosmotic stress-induced PAK translocation away from focal adhesions to the perinuclear compartment and resulted in disassembly of focal adhesions, which are hallmarks of PAK activation. Inhibition of PAK by overexpression of PP2Calpha or the kinase inhibitory domain prevented sorbitol-induced focal adhesion dissolution. Inhibition of MAPK pathways showed that MEK-ERK signaling but not p38 is required for full PAK activation and focal adhesion turnover. We conclude that 1) PAK plays a required role in hyperosmotic signaling through the PI3K/pTEN/Cdc42/PP2Calpha/p38 pathway, and 2) PAK and PP2Calpha modulate the effects of this pathway on focal adhesion dynamics.
Collapse
Affiliation(s)
- Perry M Chan
- GSK-IMCB Group, Institute of Molecular and Cell Biology, Proteos Building, 61 Biopolis Drive, Singapore 138673.
| | | | | |
Collapse
|
40
|
Awano K, Amano K, Nagaura Y, Kanno SI, Echigo S, Tamura S, Kobayashi T. Phosphorylation of Protein Phosphatase 2Cζ by c-Jun NH2-Terminal Kinase at Ser92 Attenuates Its Phosphatase Activity. Biochemistry 2008; 47:7248-55. [DOI: 10.1021/bi800067p] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Kenjiro Awano
- Department of Biochemistry and Department of Molecular Genetics, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan, and Division of Oral Surgery, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Kazutaka Amano
- Department of Biochemistry and Department of Molecular Genetics, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan, and Division of Oral Surgery, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Yuko Nagaura
- Department of Biochemistry and Department of Molecular Genetics, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan, and Division of Oral Surgery, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Shin-ichiro Kanno
- Department of Biochemistry and Department of Molecular Genetics, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan, and Division of Oral Surgery, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Seishi Echigo
- Department of Biochemistry and Department of Molecular Genetics, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan, and Division of Oral Surgery, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Shinri Tamura
- Department of Biochemistry and Department of Molecular Genetics, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan, and Division of Oral Surgery, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Takayasu Kobayashi
- Department of Biochemistry and Department of Molecular Genetics, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan, and Division of Oral Surgery, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| |
Collapse
|
41
|
Lammers T, Lavi S. Role of type 2C protein phosphatases in growth regulation and in cellular stress signaling. Crit Rev Biochem Mol Biol 2008; 42:437-61. [PMID: 18066953 DOI: 10.1080/10409230701693342] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A number of interesting features, phenotypes, and potential clinical applications have recently been ascribed to the type 2C family of protein phosphatases. Thus far, 16 different PP2C genes have been identified in the human genome, encoding (by means of alternative splicing) for at least 22 different isozymes. Virtually ever since their discovery, type 2C phosphatases have been predominantly linked to cell growth and to cellular stress signaling. Here, we provide an overview of the involvement of type 2C phosphatases in these two processes, and we show that four of them (PP2Calpha, PP2Cbeta, ILKAP, and PHLPP) can be expected to function as tumor suppressor proteins, and one as an oncoprotein (PP2Cdelta /Wip1). In addition, we demonstrate that in virtually all cases in which they have been linked to the stress response, PP2Cs act as inhibitors of cellular stress signaling. Based on the vast amount of experimental evidence obtained thus far, it therefore seems justified to conclude that type 2C protein phosphatases are important physiological regulators of cell growth and of cellular stress signaling.
Collapse
Affiliation(s)
- Twan Lammers
- Department of Innovative Cancer Diagnosis and Therapy, German Cancer Research Center, Heidelberg, Germany.
| | | |
Collapse
|
42
|
Lu G, Wang Y. FUNCTIONAL DIVERSITY OF MAMMALIAN TYPE 2C PROTEIN PHOSPHATASE ISOFORMS: NEW TALES FROM AN OLD FAMILY. Clin Exp Pharmacol Physiol 2008; 35:107-12. [DOI: 10.1111/j.1440-1681.2007.04843.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
43
|
Brenchley R, Tariq H, McElhinney H, Szöor B, Huxley-Jones J, Stevens R, Matthews K, Tabernero L. The TriTryp phosphatome: analysis of the protein phosphatase catalytic domains. BMC Genomics 2007; 8:434. [PMID: 18039372 PMCID: PMC2175518 DOI: 10.1186/1471-2164-8-434] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2007] [Accepted: 11/26/2007] [Indexed: 01/21/2023] Open
Abstract
Background The genomes of the three parasitic protozoa Trypanosoma cruzi, Trypanosoma brucei and Leishmania major are the main subject of this study. These parasites are responsible for devastating human diseases known as Chagas disease, African sleeping sickness and cutaneous Leishmaniasis, respectively, that affect millions of people in the developing world. The prevalence of these neglected diseases results from a combination of poverty, inadequate prevention and difficult treatment. Protein phosphorylation is an important mechanism of controlling the development of these kinetoplastids. With the aim to further our knowledge of the biology of these organisms we present a characterisation of the phosphatase complement (phosphatome) of the three parasites. Results An ontology-based scan of the three genomes was used to identify 86 phosphatase catalytic domains in T. cruzi, 78 in T. brucei, and 88 in L. major. We found interesting differences with other eukaryotic genomes, such as the low proportion of tyrosine phosphatases and the expansion of the serine/threonine phosphatase family. Additionally, a large number of atypical protein phosphatases were identified in these species, representing more than one third of the total phosphatase complement. Most of the atypical phosphatases belong to the dual-specificity phosphatase (DSP) family and show considerable divergence from classic DSPs in both the domain organisation and sequence features. Conclusion The analysis of the phosphatome of the three kinetoplastids indicates that they possess orthologues to many of the phosphatases reported in other eukaryotes, including humans. However, novel domain architectures and unusual combinations of accessory domains, suggest distinct functional roles for several of the kinetoplastid phosphatases, which await further experimental exploration. These distinct traits may be exploited in the selection of suitable new targets for drug development to prevent transmission and spread of the diseases, taking advantage of the already extensive knowledge on protein phosphatase inhibitors.
Collapse
Affiliation(s)
- Rachel Brenchley
- Faculty of Life Sciences, Michael Smith, University of Manchester, M13 9PT, UK.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Lammers T, Peschke P, Ehemann V, Debus J, Slobodin B, Lavi S, Huber P. Role of PP2Calpha in cell growth, in radio- and chemosensitivity, and in tumorigenicity. Mol Cancer 2007; 6:65. [PMID: 17941990 PMCID: PMC2100065 DOI: 10.1186/1476-4598-6-65] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2007] [Accepted: 10/17/2007] [Indexed: 12/19/2022] Open
Abstract
Background PP2Cα is the representative member of the type 2C family of protein phosphatases, and it has recently been implicated in the regulation of p53-, TGFβ-, cyclin-dependent kinase- and apoptosis-signaling. To investigate the role of PP2Cα in cell growth and in radio- and chemosensitivity, wild type and PP2Cα siRNA-expressing MCF7 cells were subjected to several different viability and cell cycle analyses, both under basal conditions and upon treatment with radio- and chemotherapy. By comparing the growth of tumors established from both types of cells, we also evaluated the involvement of PP2Cα in tumorigenesis. Results It was found that knockdown of PP2Cα did not affect the proliferation, the clonogenic survival and the membrane integrity of MCF7 cells. In addition, it did not alter their radio- and chemosensitivity. For PP2Cα siRNA-expressing MCF7 cells, the number of cells in the G0/G1 phase of the cell cycle was reduced, the induction of the G1 block was attenuated, the number of cells in G2/M was increased, and the induction of the G2 block was enhanced. The tumorigenic potential of PP2Cα siRNA-expressing MCF7 cells was found to be higher than that of wild type MCF7 cells, and the in vivo proliferation of these cells was found to be increased. Conclusion Based on these findings, we conclude that PP2Cα is not involved in controlling cell growth and radio- and chemosensitivity in vitro. It does, however, play a role in the regulation of the cell cycle, in the induction of cell cycle checkpoints and in tumorigenesis. The latter notion implies that PP2Cα may possess tumor-suppressing properties, and it thereby sets the stage for more elaborate analyses on its involvement in the development and progression of cancer.
Collapse
Affiliation(s)
- Twan Lammers
- Department of Innovative Cancer Diagnosis and Therapy, Clinical Cooperation Unit Radiotherapeutic Oncology, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | | | | | | | | | | | | |
Collapse
|
45
|
Saito JI, Toriumi S, Awano K, Ichijo H, Sasaki K, Kobayashi T, Tamura S. Regulation of apoptosis signal-regulating kinase 1 by protein phosphatase 2Cepsilon. Biochem J 2007; 405:591-6. [PMID: 17456047 PMCID: PMC2267319 DOI: 10.1042/bj20070231] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
ASK1 (apoptosis signal-regulating kinase 1), a MKKK (mitogen-activated protein kinase kinase kinase), is activated in response to cytotoxic stresses, such as H2O2 and TNFalpha (tumour necrosis factor alpha). ASK1 induction initiates a signalling cascade leading to apoptosis. After exposure of cells to H2O2, ASK1 is transiently activated by autophosphorylation at Thr845. The protein then associates with PP5 (protein serine/threonine phosphatase 5), which inactivates ASK1 by dephosphorylation of Thr845. Although this feedback regulation mechanism has been elucidated, it remains unclear how ASK1 is maintained in the dephosphorylated state under non-stressed conditions. In the present study, we have examined the possible role of PP2Cepsilon (protein phosphatase 2Cepsilon), a member of PP2C family, in the regulation of ASK1 signalling. Following expression in HEK-293 cells (human embryonic kidney cells), wild-type PP2Cepsilon inhibited ASK1-induced activation of an AP-1 (activator protein 1) reporter gene. Conversely, a dominant-negative PP2Cepsilon mutant enhanced AP-1 activity. Exogenous PP2Cepsilon associated with exogenous ASK1 in HEK-293 cells under non-stressed conditions, inactivating ASK1 by decreasing Thr845 phosphorylation. The association of endogenous PP2Cepsilon and ASK1 was also observed in mouse brain extracts. PP2Cepsilon directly dephosphorylated ASK1 at Thr845 in vitro. In contrast with PP5, PP2Cepsilon transiently dissociated from ASK1 within cells upon H2O2 treatment. These results suggest that PP2Cepsilon maintains ASK1 in an inactive state by dephosphorylation in quiescent cells, supporting the possibility that PP2Cepsilon and PP5 play different roles in H2O2-induced regulation of ASK1 activity.
Collapse
Affiliation(s)
- Jun-ichi Saito
- *Department of Biochemistry, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
- †Division of Advanced Prosthetic Dentistry, Graduate School of Dentistry, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
| | - Shinnosuke Toriumi
- *Department of Biochemistry, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
- ‡Division of Periodontology and Endodontology, Graduate School of Dentistry, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
| | - Kenjiro Awano
- *Department of Biochemistry, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
- §Division of Oral Surgery, Graduate School of Dentistry, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
| | - Hidenori Ichijo
- ∥Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo 113-0033, Japan
| | - Keiichi Sasaki
- †Division of Advanced Prosthetic Dentistry, Graduate School of Dentistry, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
| | - Takayasu Kobayashi
- *Department of Biochemistry, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
| | - Shinri Tamura
- *Department of Biochemistry, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
- To whom correspondence should be addressed (email )
| |
Collapse
|
46
|
Teng CH, Huang WN, Meng TC. Several dual specificity phosphatases coordinate to control the magnitude and duration of JNK activation in signaling response to oxidative stress. J Biol Chem 2007; 282:28395-28407. [PMID: 17681939 DOI: 10.1074/jbc.m705142200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mitogen-activated protein kinases (MAPKs) are important mediators that integrate signaling from upstream pathways in response to various environmental cues. In order to control appropriate gene expression through phosphorylation of transcription factors, the activity of MAPKs must be tightly regulated by the actions coordinated between protein kinases and phosphatases. In this study, we explore the underlying mechanism through which the oxidative stress-activated c-Jun N-terminal kinases (JNKs), members of MAPKs, are regulated by dual specificity phosphatases (DUSPs). DUSPs are a group of enzymes that belong to the superfamily of protein-tyrosine phosphatases. They are able to recognize phospho-Ser/Thr and phospho-Tyr residues in substrates. Using quantitative real time PCR, we found that stimulation of human embryonic kidney 293T cells with H(2)O(2) or xanthine/xanthine oxidase led to inducible expression of multiple DUSPs. We used RNA interference to characterize the functional role of these DUSPs and found rapid and transient induction of DUSP1 and DUSP10 to be essential for determining the appropriate magnitude of JNK activation in response to oxidative stress. The transcription factor ATF2, which is phosphorylated and activated by JNK, is a critical mediator for inducible expression of DUSP1 and DUSP10 in this signaling pathway. We further demonstrated that DUSP4 and DUSP16, both showing significant late phase induction, dephosphorylate JNK effectively, causing the down-regulation of the signaling cascade. Thus, this study provides new insights into the role of several DUSPs that coordinate with each other to control the magnitude and duration of JNK activity in response to oxidative stress.
Collapse
Affiliation(s)
- Chun-Hung Teng
- Institute of Biological Chemistry, Academia Sinica, 128 Academia Rd., Section 2, Taipei 11529, Taiwan
| | - Wen-Nin Huang
- Institute of Biological Chemistry, Academia Sinica, 128 Academia Rd., Section 2, Taipei 11529, Taiwan
| | - Tzu-Ching Meng
- Institute of Biological Chemistry, Academia Sinica, 128 Academia Rd., Section 2, Taipei 11529, Taiwan.
| |
Collapse
|
47
|
Sasaki M, Ohnishi M, Tashiro F, Niwa H, Suzuki A, Miyazaki JI, Kobayashi T, Tamura S. Disruption of the mouse protein Ser/Thr phosphatase 2Cβ gene leads to early pre-implantation lethality. Mech Dev 2007; 124:489-99. [PMID: 17499977 DOI: 10.1016/j.mod.2007.04.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Revised: 03/17/2007] [Accepted: 04/02/2007] [Indexed: 11/29/2022]
Abstract
Protein phosphatase 2Cbeta (PP2Cbeta) is a member of a family of protein Ser/Thr phosphatases (PP2C) that is composed of at least twelve different gene products. Recent studies have revealed that PP2Cbeta mRNA accumulates in mature sperm, unfertilized metaphase II-arrested oocytes and zygotes, but that the mRNA level then decreases sharply between the early two-cell and eight-cell stages, remaining at low levels during the 16-cell to blastocyst stages of mice. These observations raised the possibility that PP2Cbeta plays a crucial role during gametogenesis, fertilization, and/or early stages of embryonic development. In this study, we employed a gene knockout technique in mice to test this possibility. We found that PP2Cbeta(Delta/wt) mice generate normal mature gametes. However, PP2Cbeta(Delta/Delta) embryos die between the two-cell and eight-cell stages. To our interest, PP2Cbeta(Delta/Delta) ES cells which had been generated by transfecting PP2Cbeta(3lox/3lox) ES cells with Cre-expressing plasmid were viable. In addition, knockdown of PP2Cbeta using siRNA did not affect the proliferation of wild-type ES cells. These observations suggest that relatively high PP2Cbeta expression is specifically required during the early stages of pre-implantation development. The possible mechanisms for the early pre-implantation lethality of PP2Cbeta(Delta/Delta) mice are discussed.
Collapse
Affiliation(s)
- Masato Sasaki
- Department of Biochemistry, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Vahebi S, Ota A, Li M, Warren CM, de Tombe PP, Wang Y, Solaro RJ. p38-MAPK induced dephosphorylation of alpha-tropomyosin is associated with depression of myocardial sarcomeric tension and ATPase activity. Circ Res 2007; 100:408-15. [PMID: 17234967 DOI: 10.1161/01.res.0000258116.60404.ad] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Our objective in work presented here was to understand the mechanisms by which activated p38alpha MAPK depresses myocardial contractility. To test the hypothesis that activation of p38 MAPK directly influences sarcomeric function, we used transgenic mouse models with hearts in which p38 MAPK was constitutively turned on by an upstream activator (MKK6bE). These hearts demonstrated a significant depression in ejection fraction after induction of the transgene. We also studied hearts of mice expressing a dominant negative p38alpha MAPK. Simultaneous determination of tension and ATPase activity of detergent-skinned fiber bundles from left ventricular papillary muscle demonstrated a significant inhibition of both maximum tension and ATPase activity in the transgenic-MKK6bE hearts. Fibers from hearts expressing dominant negative p38alpha MAPK demonstrated no significant change in tension or ATPase activity. There were no significant changes in phosphorylation level of troponin-T3 and troponin-T4, or myosin light chain 2. However, compared with controls, there was a significant depression in levels of phosphorylation of alpha-tropomyosin and troponin I in fiber bundles from transgenic-MKK6bE hearts, but not from dominant negative p38alpha MAPK hearts. Our experiments also showed that p38alpha MAPK colocalizes with alpha-actinin at the Z-disc and complexes with protein phosphatases (PP2alpha, PP2beta). These data are the first to indicate that chronic activation of p38alpha MAPK directly depresses sarcomeric function in association with decreased phosphorylation of alpha-tropomyosin.
Collapse
Affiliation(s)
- Susan Vahebi
- Department of Physiology and Biophysics, Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, 835 S. Wolcott Ave., Chicago, IL 60612-7342, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Klumpp S, Thissen MC, Krieglstein J. Protein phosphatases types 2Cα and 2Cβ in apoptosis. Biochem Soc Trans 2006; 34:1370-5. [PMID: 17073821 DOI: 10.1042/bst0341370] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This mini-review highlights the involvement of PP2C (protein phosphatase type 2C) family members α and β in apoptosis. The activity of these isoenzymes can be stimulated by unsaturated fatty acids with special structural features, e.g. oleic acid. Those fatty acids capable of activating PP2Cα and PP2Cβ in vitro induce apoptosis in various cell types as shown here for neurons and endothelial cells. Using RNA interference to reduce the amount of PP2Cα and PP2Cβ results in cells significantly less susceptible to the apoptotic effect of oleic acid. Increased endothelial cell death is considered to be an initial step of atherogenesis. Thus activation of PP2C by physiological unbound (‘free’) unsaturated fatty acids (liberated from lipoproteins) could represent a crucial mechanism in the development of atherosclerosis.
Collapse
Affiliation(s)
- S Klumpp
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität, Münster, Germany.
| | | | | |
Collapse
|
50
|
Schwarz S, Hufnagel B, Dworak M, Klumpp S, Krieglstein J. Protein phosphatase type 2Calpha and 2Cbeta are involved in fatty acid-induced apoptosis of neuronal and endothelial cells. Apoptosis 2006; 11:1111-9. [PMID: 16699958 DOI: 10.1007/s10495-006-6982-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Unsaturated fatty acids with special structural features have been shown to activate serine/threonine protein phosphatase type 2C (PP2C) isoforms alpha and beta at physiological Mg(2+)-concentrations in vitro. These compounds also induce apoptosis in neuronal and endothelial cells. In this study we further analysed this striking correlation and tried to elucidate whether or not there is a causative relationship between activation of PP2C and induction of apoptosis. We employed RNA interference to simultaneously knock down PP2Calpha and PP2Cbeta in SH-SY5Y cells or HUVECs, respectively. This downregulation was transient. Treatment of SH-SY5Y cells or HUVECs with oleic acid (18:1,cis-Delta(9)) caused apoptosis in a time- and concentration-dependent manner. In both cases, cells with reduced PP2C-levels were less susceptible to oleic acid-induced cell damage. In conclusion, our results demonstrate that PP2C activation by unsaturated fatty acids actually causes apoptosis in neuronal and endothelial cells.
Collapse
Affiliation(s)
- Stephanie Schwarz
- Institut für Pharmakologie und Toxikologie, Philipps-Universität, Ketzerbach 63, D-35032 Marburg, Germany
| | | | | | | | | |
Collapse
|