1
|
Wörn M, Lämmer R, Lucio M, Michalke B, Rühl E, Hohberger B. The influence of trace elements on the therapeutic success of suprachoroidal draining devices. J Trace Elem Med Biol 2023; 78:127195. [PMID: 37207415 DOI: 10.1016/j.jtemb.2023.127195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/17/2023] [Accepted: 05/05/2023] [Indexed: 05/21/2023]
Abstract
PURPOSE The therapeutic success of minimal invasive glaucoma surgery (MIGS) is challenging due to many factors including fibrotic or occlusive events. Recent clinical data show sudden peaks of intraocular pressure (IOP) in the postoperative care of glaucoma patients after suprachoroidal draining stents. Yet, the reasons for the IOP peaks are speculative. As a link between trace elements and fibrosis had been previously observed in systemic disorders, the present study aimed to investigate the impact of trace elements on the therapeutic success of the suprachoroidal draining stents in patients with open-angle glaucoma (OAG). MATERIAL AND METHODS An analysis of a prospective single-center study was done: fifty-five eyes of patients with OAG (29 female, 26 male) underwent Cypass Micro-Stent implantation either as a stand-alone procedure or combined with cataract surgery. All patients underwent pre-operatively an ophthalmological examination which included slit lamp biomicroscopy and fundoscopy. IOP was measured by Goldmann applanation tonometry. Functional and morphometric data were assessed by Octopus G1-perimetry, which included measurement of retinal nerve fiber layer thickness (Spectralis OCT). Data of the patients' follow-ups were recorded during 18 months post-operatively. The therapeutic success of CyPass Micro-Stent was classified as 'success' (IOP reduction ≥20% compared to a pre-operative baseline without any medication), 'qualified success' (IOP reduction ≥20 % with same or lower additional eye medication), and 'failure' (IOP reduction ≤20 % or additional surgical treatment necessary). Aqueous humour was extracted once during surgery for analysis of the level of 14 trace elements: Copper (Cu), Cadmium (Cd), Cobalt (Co), Chromium (Cr), Iron (Fe), Lithium (Li), Magnesium (Mg), Manganese (Mn), Phosphorus (P), Lead (Pb), Titanium (Ti), Uranium (U), Vanadium (V), and Zinc (Zn). Analysis of the trace elements was done using an ELEMENT 2, ICP-sf-MS instrument (Thermo-Fisher Scientific, Bremen, Germany). Analysis of levels of trace elements was done across the patients' groups of the three subclasses of therapeutic success. Statistical investigations for substantial differences were conducted using the method of least squares to fit general linear models and mixed models. The last one for the repeated measurements of IOP. RESULTS Levels of Mg were significantly lower one month postoperatively in the success group (LS-Mean 1.30 mg/L) compared to the qualified success group (LS-Mean 1.22 mg/L; p-value = 0.04). Fe was significantly increased in the failure group (LS-Mean 2.07 µg/L) compared to the qualified success group (LS-Mean 1.64 µg/L; p-value = 0.019) after 3 months of follow-up. Additionally, Fe levels were significantly lower in the success group (LS-Mean 1.47 µg/L) compared to the failure cohort (LS-Mean 2.07 µg/L; p-value = 0.009). After a period of 18 months, significantly higher levels of Mn were observed in the success group (LS-Mean 1.24 µg/L) than in the failure group (LS Mean 0.30 µg/L, p-value = 0.019). CONCLUSION The present data might suggest that trace elements can influence therapeutic success of suprachoroidal draining devices postoperatively and thus offer first hints for potential novel therapeutic options.
Collapse
Affiliation(s)
- Maximilian Wörn
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Robert Lämmer
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Marianna Lucio
- BioGeoChemistry, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Bernhard Michalke
- BioGeoChemistry, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Eva Rühl
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Bettina Hohberger
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany.
| |
Collapse
|
2
|
Biomarkers as predictors of treatment response to tricyclic antidepressants in major depressive disorder: A systematic review. J Psychiatr Res 2022; 150:202-213. [PMID: 35397333 DOI: 10.1016/j.jpsychires.2022.03.057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/14/2022] [Accepted: 03/31/2022] [Indexed: 11/21/2022]
Abstract
Tricyclic antidepressants (TCAs) are frequently prescribed in case of non-response to first-line antidepressants in Major Depressive Disorder (MDD). Treatment of MDD often entails a trial-and-error process of finding a suitable antidepressant and its appropriate dose. Nowadays, a shift is seen towards a more personalized treatment strategy in MDD to increase treatment efficacy. One of these strategies involves the use of biomarkers for the prediction of antidepressant treatment response. We aimed to summarize biomarkers for prediction of TCA specific (i.e. per agent, not for the TCA as a drug class) treatment response in unipolar nonpsychotic MDD. We performed a systematic search in PubMed and MEDLINE. After full-text screening, 36 papers were included. Seven genetic biomarkers were identified for nortriptyline treatment response. For desipramine, we identified two biomarkers; one genetic and one nongenetic. Three nongenetic biomarkers were identified for imipramine. None of these biomarkers were replicated. Quality assessment demonstrated that biomarker studies vary in endpoint definitions and frequently lack power calculations. None of the biomarkers can be confirmed as a predictor for TCA treatment response. Despite the necessity for TCA treatment optimization, biomarker studies reporting drug-specific results for TCAs are limited and adequate replication studies are lacking. Moreover, biomarker studies generally use small sample sizes. To move forward, larger cohorts, pooled data or biomarkers combined with other clinical characteristics should be used to improve predictive power.
Collapse
|
3
|
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
|
4
|
Role of active site arginine residues in substrate recognition by PPM1A. Biochem Biophys Res Commun 2021; 581:1-5. [PMID: 34637963 DOI: 10.1016/j.bbrc.2021.10.001] [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: 08/31/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 11/21/2022]
Abstract
Reversible protein phosphorylation is a key mechanism for regulating numerous cellular events. The metal-dependent protein phosphatases (PPM) are a family of Ser/Thr phosphatases, which uniquely recognize their substrate as a monomeric enzyme. In the case of PPM1A, it has the capacity to dephosphorylate a variety of substrates containing different sequences, but it is not yet fully understood how it recognizes its substrates. Here we analyzed the role of Arg33 and Arg186, two residues near the active site, on the dephosphorylation activity of PPM1A. The results showed that both Arg residues were critical for enzymatic activity and docking-model analysis revealed that Arg186 is positioned to interact with the substrate phosphate group. In addition, our results suggest that which Arg residue plays a more significant role in the catalysis depends directly on the substrate.
Collapse
|
5
|
Wang X, Liu H, Liao X, Qiao L, Zhu L, Wu S, Zhou Y, Zhang Y, Li B, Lin L, Ma J, Gu Q, Shu J. Dissecting the Roles of LncRNAs in the Development of Periventricular White Matter Damage. Front Genet 2021; 12:641526. [PMID: 33995480 PMCID: PMC8120246 DOI: 10.3389/fgene.2021.641526] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/29/2021] [Indexed: 12/22/2022] Open
Abstract
Long non-coding RNA (LncRNA) has high expression in the brain. Animal studies have shown that lncRNA plays an important role in brain functions and mediates the development of many neurological diseases. However, data on the expression of lncRNAs and the clinical significance in prematurely born infants with diseases such as periventricular white matter damage (PWMD) remains scant. Here, we compared the expression of the lncRNAs in whole blood samples obtained from prematurely born infants with PWMD with samples from prematurely born infants without PWMD. Our data demonstrated differential expression of the lncRNAs between the two groups. Further, we showed that the lncRNAs play important roles in the development of PWMD. Our findings give insights into the functions of the lncRNAs in PWMD and provide evidence for the improvement of diagnostic and treatment strategies in infants with PWMD.
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Heng Liu
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Xiaoli Liao
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Lixing Qiao
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Lihua Zhu
- Institute of Clinical, Jiangsu Health Vocational College, Nanjing, China
| | - Shun Wu
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Yan Zhou
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Yi Zhang
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Bangbang Li
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Lili Lin
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Jingjing Ma
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Qianying Gu
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Jiaping Shu
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| |
Collapse
|
6
|
Wang L, Ge S, Zhou F. MicroRNA-487a-3p inhibits the growth and invasiveness of oral squamous cell carcinoma by targeting PPM1A. Bioengineered 2021; 12:937-947. [PMID: 33724144 PMCID: PMC8291853 DOI: 10.1080/21655979.2021.1884396] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) forms the majority of the entire cancerous tumors which occur in the mouth. Current treatment advances, such as surgical resection, chemotherapy, and radiotherapy, have significantly helped reduce OSCC. However, the overall patient survival rate remains relatively low. MiRNAs, a non-coding RNA group, are essential for multiple biological functions, which are essential for the progression of cancer, including survival of the cell, migration, multiplication, differentiation, and apoptosis. The study aimed to explore the existing association between miR-487a-3p and PPM1A and elucidating their role in modulation of proliferation in OSCC cell lines. In this study, we used CAL-27 and TCA-8113 OSCC cell lines and human samples to validate our results. The manifestation of miR-487a-3p and PPM1A was checked using quantitative real-time PCR. The miR-487a-3p and PPM1A binding was investigated through western blot assay and dual-luciferase reporter gene. Functional experiments, including colony formation, CCK-8, and transwell experimentations, were undertaken to validate cells' growth and invasion activities. According to the results, the expression of miR-487a-3p is regulated in the OSCC cell lines compared to normal cells. Moreover, the mimicking of miR-487a-3p significantly reduces the OSCC cell growth and invasion, and PPM1A overexpression exerts oncogenic effects and hinders the anti-oncogenic effects of miR-487a-3p. In conclusion, the study demonstrated that miR-487a-3p might act as a tumor suppressor by inhibiting the growth and invasion of OSCC via regulating PPM1A expression.
Collapse
Affiliation(s)
- Lishan Wang
- Department of Maxillofacial Surgery, Weifang City People's Hospital, Weifang City, Shandong Province, China
| | - Shuqing Ge
- Department of Maxillofacial Surgery, Weifang City People's Hospital, Weifang City, Shandong Province, China
| | - Futing Zhou
- Department of Maxillofacial Surgery, Weifang City People's Hospital, Weifang City, Shandong Province, China
| |
Collapse
|
7
|
Flannery PC, Abbott KL, Pondugula SR. Correlation of PPM1A Downregulation with CYP3A4 Repression in the Tumor Liver Tissue of Hepatocellular Carcinoma Patients. Eur J Drug Metab Pharmacokinet 2020; 45:297-304. [PMID: 31792727 DOI: 10.1007/s13318-019-00595-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND OBJECTIVE In many patients with hepatocellular carcinoma (HCC), cytochrome P450 3A4 (CYP3A4) expression has been reported to be significantly reduced in the tumor liver tissue. Moreover, this CYP3A4 repression is associated with decreased CYP3A4-mediated drug metabolism in the tumor liver tissue. However, the underlying mechanisms of CYP3A4 repression are not fully understood. We have previously shown that Mg2+/Mn2+-dependent phosphatase 1A (PPM1A) positively regulates human pregnane X receptor (hPXR)-mediated CYP3A4 expression in a PPM1A expression-dependent manner. We sought to determine whether PPM1A expression is downregulated and whether PPM1A downregulation is correlated with CYP3A4 repression in the tumor liver tissue of HCC patients. METHODS Quantitative RT-PCR and western blot analyses were performed to study mRNA and protein expression, respectively. Cell-based reporter gene assays were conducted to examine the hPXR transactivation of CYP3A4 promoter activity. RESULTS Arginase-1 and glypican-3 gene expression studies confirmed that the tumor samples used in our study originate from HCC livers but not non-hepatocellular tumors. mRNA and protein expression of PPM1A and CYP3A4 was found to be significantly repressed in the tumor liver tissues compared to the matched non-tumor liver tissues. In the reporter gene assays, elevated PPM1A levels counteracted the inhibition of hPXR-mediated CYP3A4 promoter activity by signaling pathways that are upregulated in HCC, suggesting that decreased PPM1A levels in HCC could not fully counteract the hPXR-inhibiting signaling pathways. CONCLUSIONS Together, these results are consistent with the conclusion that PPM1A downregulation in the tumor liver tissue of HCC patients correlates with CYP3A4 repression. Downregulation of PPM1A levels in the tumor liver tissue may contribute to reduced hPXR-mediated CYP3A4 expression, and provide a novel mechanism of CYP3A4 repression in the tumor liver tissue of HCC patients.
Collapse
Affiliation(s)
- Patrick C Flannery
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, 109 Greene Hall, Auburn, AL, 36849, USA.,Auburn University Research Initiative in Cancer, Auburn University, Auburn, AL, 36849, USA
| | - Kodye L Abbott
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, 109 Greene Hall, Auburn, AL, 36849, USA.,Auburn University Research Initiative in Cancer, Auburn University, Auburn, AL, 36849, USA
| | - Satyanarayana R Pondugula
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, 109 Greene Hall, Auburn, AL, 36849, USA. .,Auburn University Research Initiative in Cancer, Auburn University, Auburn, AL, 36849, USA.
| |
Collapse
|
8
|
Wong HL, Poon SHL, Bu Y, Lo ACY, Jhanji V, Chan YK, Shih KC. A Systematic Review on Cornea Epithelial-Stromal Homeostasis. Ophthalmic Res 2020; 64:178-191. [PMID: 32474566 DOI: 10.1159/000509030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/29/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION This review aims to summarise the role of different cells, genes, proteins and lipid in regulating cornea epithelial-stromal homeostasis. METHODS We performed an Entrez PubMed literature search using keywords "human," "cornea," "epithelial," "stromal," "homeostasis," "fibrosis response," and "pathogenesis" on 24th of September 2019, resulting in 35 papers, of which 18 were chosen after filtering for "English language" and "published within 10 years" as well as curation for relevance by the authors. RESULTS The 18 selected papers showed that corneal epithelial cells, fibroblasts and telocytes, together with genes such as Klf4, Pax6 and Id found in the cells, play important roles in achieving homeostasis to maintain corneal integrity and transparency. Proteins classified as pro-fibrotic ligands and anti-fibrotic ligands are responsible for regulating cornea stromal fibrosis and extracellular matrix deposition, thus regulators of scar formation during wound healing. Anti-inflammatory ligands and wound repairing ligands are critical in eliciting protective inflammation and promoting epithelial healing, respectively. Protein receptors located on cellular membrane play a role in maintaining intercellular connections as well as corneal hydration. DISCUSSION/CONCLUSION These studies prompt development of novel therapeutic strategies such as tear drops or ointments that target certain proteins to maintain corneal homeostasis. However, more in vitro and in vivo studies are required to prove the effectiveness of exogenous administration of molecules in improving healing outcome. Hence, future investigations of the molecular pathways highlighted in this review will reveal novel therapeutic tools such as gene or cell therapy to treat corneal diseases.
Collapse
Affiliation(s)
- Ho Lam Wong
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong, China
| | - Stephanie Hiu Ling Poon
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong, China
| | - Yashan Bu
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong, China
| | - Amy Cheuk Yin Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong, China
| | - Vishal Jhanji
- Department of Ophthalmology, University of Pittsburgh Medical Centre, Pittsburgh, Pennsylvania, USA
| | - Yau Kei Chan
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong, China
| | - Kendrick Co Shih
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong, China,
| |
Collapse
|
9
|
Wang F, Wang H, Sun L, Niu C, Xu J. TRIM59 inhibits PPM1A through ubiquitination and activates TGF-β/Smad signaling to promote the invasion of ectopic endometrial stromal cells in endometriosis. Am J Physiol Cell Physiol 2020; 319:C392-C401. [PMID: 32348176 DOI: 10.1152/ajpcell.00127.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This study was conducted to define the underlying molecular mechanism of tripartite motif (TRIM) 59-induced invasion of ectopic endometrial stromal cells in endometriosis. Primary endometriosis ectopic endometrial stromal cells and normal endometrial cells were isolated and purified. Western blot was used to detect the expression of TRIM59, protein phosphatase Mg2+/Mn2+-dependent 1A (PPM1A), smad2/3, and phosphorylated (p)-smad2/3. Lentiviral vector-mediated TRIM59 interference and overexpression were established. Cell Counting Kit-8 assay was used to detect cell proliferation, and the Transwell migration assay was used to detect cell invasion. Matrix metalloproteinase (MMP-2), MMP9, smad2/3, and p-smad2/3 expressions were also detected using Western blot analysis; degradation of PPM1A was verified to be through ubiquitination. We found that TRIM59 expression levels in the endometriosis group was significantly higher compared with the normal group (P < 0.05), whereas the expression levels of PPM1A in the endometriosis group were significantly lower (P < 0.05). Endometriosis did not alter smad2/3 (P > 0.05) expression. However, after activating smad2/3 by phosphorylation, the expression of p-smad2/3 in the endometriosis group was significantly higher compared with the normal group (P < 0.05). The content of PPM1A in the TRIM59 overexpression group was significantly lower than that in the control group (P < 0.001), whereas the content of PPM1A in the siTRIM59 group was significantly higher than that in the control group (P < 0.001). In addition, there were no significant differences in the mRNA levels of PPM1A among the five groups, indicating that TRIM59 affects the expression of PPM1A at the posttranslational level (P < 0.05). Overexpression of TRIM59 significantly promoted the ubiquitination of PPM1A. We conclude that TRIM59 inhibits PPM1A through ubiquitination and activates the transforming growth factor-β/Smad pathway to promote the invasion of ectopic endometrial stromal cells in endometriosis.
Collapse
Affiliation(s)
- Fengyu Wang
- Henan Provincial Research Institute for Population and Family Planning, Key Laboratory of Birth Defects Prevention, National Health Commission, and Key Laboratory of Population Defects Intervention Technology of Henan Province, Zhengzhou, China
| | - Haili Wang
- Henan Provincial Research Institute for Population and Family Planning, Key Laboratory of Birth Defects Prevention, National Health Commission, and Key Laboratory of Population Defects Intervention Technology of Henan Province, Zhengzhou, China
| | - Lei Sun
- Translational Medical Center, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, China
| | - Chengling Niu
- Henan Provincial Research Institute for Population and Family Planning, Key Laboratory of Birth Defects Prevention, National Health Commission, and Key Laboratory of Population Defects Intervention Technology of Henan Province, Zhengzhou, China
| | - Jie Xu
- Department of Gynecology and Obstetrics, Yancheng Third People's Hospital, Yancheng, China
| |
Collapse
|
10
|
Xu X, Liu M. miR-522 stimulates TGF-β/Smad signaling pathway and promotes osteosarcoma tumorigenesis by targeting PPM1A. J Cell Biochem 2019; 120:18425-18434. [PMID: 31190351 DOI: 10.1002/jcb.29160] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 05/18/2019] [Accepted: 05/23/2019] [Indexed: 12/19/2022]
Abstract
Osteosarcoma (OS) is identified as an aggressive malignancy of the skeletal system and normally occurs among young people. It is well accepted that microRNAs are implicated in biological activities of diverse tumors. Although miR-522 has been proved to elicit oncogenic properties in a wide range of human cancers, the physiological function and latent mechanism of miR-522 in OS tumorigenesis remain largely to be probed. In the current study, we certified that miR-522 was highly expressed in OS cells and presented carcinogenic function by contributing to cell proliferation, migration, and EMT progression whereas dampening cell apoptosis. In addition, miR-522 provoked TGF-β/Smad pathway through targeting PPM1A. Finally, the results of mechanism experiments elucidated that miR-522 stimulated TGF-β/Smad pathway to induce the development of OS via targeting PPM1A, which exposed that miR-522 may become a promising curative target for OS patients.
Collapse
Affiliation(s)
- Xiqiang Xu
- Department of Spine Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Mengmeng Liu
- Department of Anesthesiology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| |
Collapse
|
11
|
Tong Y, Song Y, Deng S. Combined analysis and validation for DNA methylation and gene expression profiles associated with prostate cancer. Cancer Cell Int 2019; 19:50. [PMID: 30867653 PMCID: PMC6399908 DOI: 10.1186/s12935-019-0753-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 02/08/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Prostate cancer (PCa) is a malignancy cause of cancer deaths and frequently diagnosed in male. This study aimed to identify tumor suppressor genes, hub genes and their pathways by combined bioinformatics analysis. METHODS A combined analysis method was used for two types of microarray datasets (DNA methylation and gene expression profiles) from the Gene Expression Omnibus (GEO). Differentially methylated genes (DMGs) were identified by the R package minfi and differentially expressed genes (DEGs) were screened out via the R package limma. A total of 4451 DMGs and 1509 DEGs, identified with nine overlaps between DMGs, DEGs and tumor suppressor genes, were screened for candidate tumor suppressor genes. All these nine candidate tumor suppressor genes were validated by TCGA (The Cancer Genome Atlas) database and Oncomine database. And then, the gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) enrichment analyses were performed by DAVID (Database for Annotation, Visualization and Integrated Discovery) database. Protein-protein interaction (PPI) network was constructed by STRING and visualized in Cytoscape. At last, Kaplan-Meier analysis was performed to validate these genes. RESULTS The candidate tumor suppressor genes were IKZF1, PPM1A, FBP1, SMCHD1, ALPL, CASP5, PYHIN1, DAPK1 and CASP8. By validation in TCGA database, PPM1A, DAPK1, FBP1, PYHIN1, ALPL and SMCHD1 were significant. The hub genes were FGFR1, FGF13 and CCND1. These hub genes were identified from the PPI network, and sub-networks revealed by these genes were involved in significant pathways. CONCLUSION In summary, the study indicated that the combined analysis for identifying target genes with PCa by bioinformatics tools promote our understanding of the molecular mechanisms and underlying the development of PCa. And the hub genes might serve as molecular targets and diagnostic biomarkers for precise diagnosis and treatment of PCa.
Collapse
Affiliation(s)
- Yanqiu Tong
- Laboratory of Forensic Medicine and Biomedical Informatics, Chongqing Medical University, Chongqing, 400016 People’s Republic of China
- School of Humanity, Chongqing Jiaotong University, Chongqing, 400074 People’s Republic of China
| | - Yang Song
- Department of Device, Chongqing Medical University, Chongqing, 400016 People’s Republic of China
| | - Shixiong Deng
- Laboratory of Forensic Medicine and Biomedical Informatics, Chongqing Medical University, Chongqing, 400016 People’s Republic of China
| |
Collapse
|
12
|
Coates RF, Gardner JA, Gao Y, Cortright VM, Mitchell JM, Ashikaga T, Skelly J, Yang MX. Significance of positive and inhibitory regulators in the TGF-β signaling pathway in colorectal cancers. Hum Pathol 2017; 66:34-39. [PMID: 28601657 DOI: 10.1016/j.humpath.2017.05.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/25/2017] [Accepted: 05/26/2017] [Indexed: 10/19/2022]
Abstract
Inactivation of genes in the transforming growth factor (TGF)-β/SMAD signaling pathway is a well-known step for the progression of colorectal cancers (CRCs). Genetic mutations can occur in the precursors, and the combined prevalence of SMAD4, SMAD2, and SMAD3 mutations was seen in up to 50% of CRCs. High levels of serum TGF-β1 were reported in patients with CRC and were associated with poor clinical outcome. PPM1A is an important inhibitory regulator in the TGF-β signaling pathway and contributes to terminating the TGF-β/SMAD signaling activity. We recently showed that PPM1A expression was lost in approximately 45% of pancreatic ductal adenocarcinomas and loss of PPM1A was associated with worse overall survival. Genome-wide analyses from The Cancer Genome Atlas revealed that abnormal TGF-β signaling pathway is among the most common molecular changes in CRC. The complexity of the TGF-β signaling pathway is its dual function as a tumor suppressor and tumor-promoting factor, depending on the cellular and molecular context. In this study, we simultaneously investigated the protein expression pattern of 3 regulators in the TGF-β/SMAD signaling pathway, including SMAD4, PPM1A, and TGF-β1, and their clinicopathological correlations in CRCs by immunohistochemistry. We observed that loss of SMAD4 and PPM1A was seen in 37.8% and 7.3% of CRCs, respectively. Loss of SMAD4, lymphovascular invasion, and distant metastasis were independently associated with worse overall survival in multivariate analysis. However, loss of PPM1A was associated with worse overall survival with less statistical strength. Our findings would provide new insights into the pathophysiological function of different components in the TGF-β signaling pathway in CRC.
Collapse
Affiliation(s)
- Ryan F Coates
- Department of Pathology and Laboratory Medicine, University of Vermont Medical Center, Burlington, VT 05401, United States
| | - Juli-Anne Gardner
- Department of Pathology and Laboratory Medicine, University of Vermont Medical Center, Burlington, VT 05401, United States
| | - Yuan Gao
- Department of Gastrointestinal Surgery, Changzhou 2nd People's Hospital, Changzhou, Jiangsu Province, 213000, China
| | - Valerie M Cortright
- Department of Pathology and Laboratory Medicine, University of Vermont Medical Center, Burlington, VT 05401, United States
| | - Jeannette M Mitchell
- Department of Pathology and Laboratory Medicine, University of Vermont Medical Center, Burlington, VT 05401, United States
| | - Takamaru Ashikaga
- University of Vermont Medical Biostatistics Department, Burlington, VT, 05401, United States
| | - Joan Skelly
- University of Vermont Medical Biostatistics Department, Burlington, VT, 05401, United States
| | - Michelle X Yang
- Department of Pathology and Laboratory Medicine, University of Vermont Medical Center, Burlington, VT 05401, United States.
| |
Collapse
|
13
|
Abstract
Cells respond to changes in their environment, to developmental cues, and to pathogen aggression through the action of a complex network of proteins. These networks can be decomposed into a multitude of signaling pathways that relay signals from the microenvironment to the cellular components involved in eliciting a specific response. Perturbations in these signaling processes are at the root of multiple pathologies, the most notable of these being cancer. The study of receptor tyrosine kinase (RTK) signaling led to the first description of a mechanism whereby an extracellular signal is transmitted to the nucleus to induce a transcriptional response. Genetic studies conducted in drosophila and nematodes have provided key elements to this puzzle. Here, we briefly discuss the somewhat lesser known contribution of these multicellular organisms to our understanding of what has come to be known as the prototype of signaling pathways. We also discuss the ostensibly much larger network of regulators that has emerged from recent functional genomic investigations of RTK/RAS/ERK signaling.
Collapse
Affiliation(s)
- Dariel Ashton-Beaucage
- Institute for Research in Immunology and Cancer, Laboratory of Intracellular Signaling, Université de Montréal, C.P. 6128, Succursale Centre-Ville, Montreal, QC, Canada, H3C 3J7
| | - Marc Therrien
- Institute for Research in Immunology and Cancer, Laboratory of Intracellular Signaling, Université de Montréal, C.P. 6128, Succursale Centre-Ville, Montreal, QC, Canada, H3C 3J7.
- Département de Pathologie et de Biologie Cellulaire, Université de Montréal, C.P. 6128, Succursale Centre-Ville, Montreal, QC, Canada, H3C 3J7.
| |
Collapse
|
14
|
Fan J, Yang MX, Ouyang Q, Fu D, Xu Z, Liu X, Mino-Kenudson M, Geng J, Tang F. Phosphatase PPM1A is a novel prognostic marker in pancreatic ductal adenocarcinoma. Hum Pathol 2016; 55:151-158. [PMID: 27195906 DOI: 10.1016/j.humpath.2016.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 04/24/2016] [Accepted: 05/05/2016] [Indexed: 01/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) harbors complex molecular alterations and remains a lethal disease. Aberrant TGF-β/Smads signaling is a well-known mechanism involved in the progression of PDACs. However, loss of Smad4 expression is reported in only ~50% of PDACs and is generally associated with worse prognosis. Investigating additional prognostic markers is warranted. PPM1A is a phosphatase that dephosphorylates TGF-β-activated Smad2/3 and inactivates the TGF-β signaling. Little is known about the clinical significance of PPM1A in PDACs and its functional relationship to Smad4. In this study, PPM1A and Smad4 immunohistochemistry was assessed in 180 R0 resected human PDACs. PPM1A was lost in 41.7% cases, whereas Smad4 was lost in 45.7% cases. The median survival rate with negative and positive PPM1A was 10.9 and 16.8 months, respectively. Loss of PPM1A was significantly associated with larger tumor size and higher stage and was an independent predictor of unfavorable outcomes. Intriguingly, the overall survival of this cohort was divided into 3 groups based on the expression pattern of PPM1A and Smad4, with the Smad4+/PPM1A+ pattern associated with favorable survival, the Smad4+/PPM1A- or Smad4-/PPM1A- pattern associated with unfavorable, and the PPM1A+/Smad4- pattern fell between these 2 groups. In 82 cases with negative Smad4, PPM1A or P-Smad2/3 expression was retained. Using a SMAD4-deficient human PDAC cell line, BxPC3, we further demonstrated that TGF-β1 treatment induced PPM1A and P-Smad2/3 expression in this cell line. PPM1A and Smad4 immunohistochemistry in surgical specimens may provide more accurate prognostic stratification for patients with PDAC.
Collapse
Affiliation(s)
- Jie Fan
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Michelle X Yang
- Department of Pathology, University of Vermont Medical Center, Burlington, VT 05401
| | - Qi Ouyang
- Department of Pathology, Huadong Hospital; Fudan University, Shanghai 200040, China
| | - Deliang Fu
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zude Xu
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiuping Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114
| | - Jiang Geng
- Department of Urology, Shanghai 10th People's Hospital, Tongji University, Shanghai 200072, China.
| | - Feng Tang
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
15
|
Mummidi S, Das NA, Carpenter AJ, Kandikattu H, Krenz M, Siebenlist U, Valente AJ, Chandrasekar B. Metformin inhibits aldosterone-induced cardiac fibroblast activation, migration and proliferation in vitro, and reverses aldosterone+salt-induced cardiac fibrosis in vivo. J Mol Cell Cardiol 2016; 98:95-102. [PMID: 27423273 DOI: 10.1016/j.yjmcc.2016.07.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 07/08/2016] [Accepted: 07/13/2016] [Indexed: 11/29/2022]
Abstract
The overall goals of this study were to investigate whether metformin exerts anti-fibrotic effects in aldosterone (Aldo)+salt-treated wild type mouse hearts, and determine the underlying molecular mechanisms in isolated adult cardiac fibroblasts (CF). In vitro, Aldo induced CF activation, migration, and proliferation, and these effects were inhibited by metformin. Further, Aldo induced PPM1A (Protein Phosphatase Magnesium Dependent 1A) activation and inhibited AMPK phosphorylation. At a pharmacologically relevant concentration, metformin restored AMPK activation, and inhibited Aldo-induced Nox4/H2O2-dependent TRAF3IP2 induction, pro-inflammatory cytokine expression, and CF migration and proliferation. Further, metformin potentiated the inhibitory effects of spironolactone, a mineralocorticoid receptor antagonist, on Aldo-induced collagen expression, and CF migration and proliferation. These results were recapitulated in vivo, where metformin reversed Aldo+salt-induced oxidative stress, suppression of AMPK activation, TRAF3IP2 induction, pro-inflammatory cytokine expression, and cardiac fibrosis, without significantly modulating systolic blood pressure. These in vitro and in vivo data indicate that metformin has the potential to reduce adverse cardiac remodeling in hypertensive heart disease.
Collapse
Affiliation(s)
- Srinivas Mummidi
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Nitin A Das
- Department of Cardiothoracic Surgery, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Andrea J Carpenter
- Department of Cardiothoracic Surgery, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | | | - Maike Krenz
- Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65211, USA
| | | | - Anthony J Valente
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Bysani Chandrasekar
- Medicine/Cardiology, University of Missouri School of Medicine, Columbia, MO 65211, USA; Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO 65201, USA.
| |
Collapse
|
16
|
Pondugula SR, Flannery PC, Apte U, Babu JR, Geetha T, Rege SD, Chen T, Abbott KL. Mg2+/Mn2+-dependent phosphatase 1A is involved in regulating pregnane X receptor-mediated cytochrome p450 3A4 gene expression. Drug Metab Dispos 2015; 43:385-91. [PMID: 25561723 PMCID: PMC11024896 DOI: 10.1124/dmd.114.062083] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 01/05/2015] [Indexed: 04/20/2024] Open
Abstract
Variations in the expression of human pregnane X receptor (hPXR)-mediated cytochrome p450 3A4 (CYP3A4) in liver can alter therapeutic response to a variety of drugs and may lead to potential adverse drug interactions. We sought to determine whether Mg(2+)/Mn(2+)-dependent phosphatase 1A (PPM1A) regulates hPXR-mediated CYP3A4 expression. PPM1A was found to be coimmunoprecipitated with hPXR. Genetic or pharmacologic activation of PPM1A led to a significant increase in hPXR transactivation of CYP3A4 promoter activity. In contrast, knockdown of endogenous PPM1A not only attenuated hPXR transactivation, but also increased proliferation of HepG2 human liver carcinoma cells, suggesting that PPM1A expression levels regulate hPXR, and that PPM1A expression is regulated in a proliferation-dependent manner. Indeed, PPM1A expression and hPXR transactivation were found to be significantly reduced in subconfluent HepG2 cells compared with confluent HepG2 cells, suggesting that both PPM1A expression and hPXR-mediated CYP3A4 expression may be downregulated in proliferating livers. Elevated PPM1A levels led to attenuation of hPXR inhibition by tumor necrosis factor-α and cyclin-dependent kinase-2, which are known to be upregulated and essential during liver regeneration. In mouse regenerating livers, similar to subconfluent HepG2 cells, expression of both PPM1A and the mouse PXR target gene cyp3a11 was found to be downregulated. Our results show that PPM1A can positively regulate PXR activity by counteracting PXR inhibitory signaling pathways that play a major role in liver regeneration. These results implicate a novel role for PPM1A in regulating hPXR-mediated CYP3A4 expression in hepatocytes and may explain a mechanism for CYP3A repression in regenerating livers.
Collapse
Affiliation(s)
- Satyanarayana R Pondugula
- Department of Anatomy, Physiology, and Pharmacology (S.R.P., P.C.F., K.L.A.) and Department of Nutrition, Dietetics, and Hospitality Management (J.R.B., S.D.R.), Auburn University, Auburn, Alabama; Department of Chemistry (T.G.), Auburn University at Montgomery, Montgomery, Alabama; Department of Chemical Biology and Therapeutics (T.C.), St. Jude Children's Research Hospital, Memphis, Tennessee; and Department of Pharmacology, Toxicology, and Therapeutics (U.A.), University of Kansas, Kansas City, Kansas
| | - Patrick C Flannery
- Department of Anatomy, Physiology, and Pharmacology (S.R.P., P.C.F., K.L.A.) and Department of Nutrition, Dietetics, and Hospitality Management (J.R.B., S.D.R.), Auburn University, Auburn, Alabama; Department of Chemistry (T.G.), Auburn University at Montgomery, Montgomery, Alabama; Department of Chemical Biology and Therapeutics (T.C.), St. Jude Children's Research Hospital, Memphis, Tennessee; and Department of Pharmacology, Toxicology, and Therapeutics (U.A.), University of Kansas, Kansas City, Kansas
| | - Udayan Apte
- Department of Anatomy, Physiology, and Pharmacology (S.R.P., P.C.F., K.L.A.) and Department of Nutrition, Dietetics, and Hospitality Management (J.R.B., S.D.R.), Auburn University, Auburn, Alabama; Department of Chemistry (T.G.), Auburn University at Montgomery, Montgomery, Alabama; Department of Chemical Biology and Therapeutics (T.C.), St. Jude Children's Research Hospital, Memphis, Tennessee; and Department of Pharmacology, Toxicology, and Therapeutics (U.A.), University of Kansas, Kansas City, Kansas
| | - Jeganathan Ramesh Babu
- Department of Anatomy, Physiology, and Pharmacology (S.R.P., P.C.F., K.L.A.) and Department of Nutrition, Dietetics, and Hospitality Management (J.R.B., S.D.R.), Auburn University, Auburn, Alabama; Department of Chemistry (T.G.), Auburn University at Montgomery, Montgomery, Alabama; Department of Chemical Biology and Therapeutics (T.C.), St. Jude Children's Research Hospital, Memphis, Tennessee; and Department of Pharmacology, Toxicology, and Therapeutics (U.A.), University of Kansas, Kansas City, Kansas
| | - Thangiah Geetha
- Department of Anatomy, Physiology, and Pharmacology (S.R.P., P.C.F., K.L.A.) and Department of Nutrition, Dietetics, and Hospitality Management (J.R.B., S.D.R.), Auburn University, Auburn, Alabama; Department of Chemistry (T.G.), Auburn University at Montgomery, Montgomery, Alabama; Department of Chemical Biology and Therapeutics (T.C.), St. Jude Children's Research Hospital, Memphis, Tennessee; and Department of Pharmacology, Toxicology, and Therapeutics (U.A.), University of Kansas, Kansas City, Kansas
| | - Shraddha D Rege
- Department of Anatomy, Physiology, and Pharmacology (S.R.P., P.C.F., K.L.A.) and Department of Nutrition, Dietetics, and Hospitality Management (J.R.B., S.D.R.), Auburn University, Auburn, Alabama; Department of Chemistry (T.G.), Auburn University at Montgomery, Montgomery, Alabama; Department of Chemical Biology and Therapeutics (T.C.), St. Jude Children's Research Hospital, Memphis, Tennessee; and Department of Pharmacology, Toxicology, and Therapeutics (U.A.), University of Kansas, Kansas City, Kansas
| | - Taosheng Chen
- Department of Anatomy, Physiology, and Pharmacology (S.R.P., P.C.F., K.L.A.) and Department of Nutrition, Dietetics, and Hospitality Management (J.R.B., S.D.R.), Auburn University, Auburn, Alabama; Department of Chemistry (T.G.), Auburn University at Montgomery, Montgomery, Alabama; Department of Chemical Biology and Therapeutics (T.C.), St. Jude Children's Research Hospital, Memphis, Tennessee; and Department of Pharmacology, Toxicology, and Therapeutics (U.A.), University of Kansas, Kansas City, Kansas
| | - Kodye L Abbott
- Department of Anatomy, Physiology, and Pharmacology (S.R.P., P.C.F., K.L.A.) and Department of Nutrition, Dietetics, and Hospitality Management (J.R.B., S.D.R.), Auburn University, Auburn, Alabama; Department of Chemistry (T.G.), Auburn University at Montgomery, Montgomery, Alabama; Department of Chemical Biology and Therapeutics (T.C.), St. Jude Children's Research Hospital, Memphis, Tennessee; and Department of Pharmacology, Toxicology, and Therapeutics (U.A.), University of Kansas, Kansas City, Kansas
| |
Collapse
|
17
|
Pan C, Tang JY, Xu YF, Xiao P, Liu HD, Wang HA, Wang WB, Meng FG, Yu X, Sun JP. The catalytic role of the M2 metal ion in PP2Cα. Sci Rep 2015; 5:8560. [PMID: 25708299 PMCID: PMC5390078 DOI: 10.1038/srep08560] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 01/26/2015] [Indexed: 01/16/2023] Open
Abstract
PP2C family phosphatases (the type 2C family of protein phosphatases; or metal-dependent phosphatase, PPM) constitute an important class of signaling enzymes that regulate many fundamental life activities. All PP2C family members have a conserved binuclear metal ion active center that is essential for their catalysis. However, the catalytic role of each metal ion during catalysis remains elusive. In this study, we discovered that mutations in the structurally buried D38 residue of PP2Cα (PPM1A) redefined the water-mediated hydrogen network in the active site and selectively disrupted M2 metal ion binding. Using the D38A and D38K mutations of PP2Cα as specific tools in combination with enzymology analysis, our results demonstrated that the M2 metal ion determines the rate-limiting step of substrate hydrolysis, participates in dianion substrate binding and stabilizes the leaving group after P-O bond cleavage. The newly characterized catalytic role of the M2 metal ion in this family not only provides insight into how the binuclear metal centers of the PP2C phosphatases are organized for efficient catalysis but also helps increase our understanding of the function and substrate specificity of PP2C family members.
Collapse
Affiliation(s)
- Chang Pan
- 1] Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University, School of Medicine, Jinan, Shandong, China [2] Qilu Hospital of Shandong University, Jinan, China
| | - Jun-yi Tang
- 1] Shandong Provincial School Key laboratory for Protein Science of Chronic Degenerative Diseases, Jinan, Shandong, China [2] Department of Physiology, Shandong University, School of Medicine, Jinan, Shandong, China
| | - Yun-fei Xu
- 1] Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University, School of Medicine, Jinan, Shandong, China [2] Qilu Hospital of Shandong University, Jinan, China
| | - Peng Xiao
- 1] Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University, School of Medicine, Jinan, Shandong, China [2] Shandong Provincial School Key laboratory for Protein Science of Chronic Degenerative Diseases, Jinan, Shandong, China
| | - Hong-da Liu
- 1] Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University, School of Medicine, Jinan, Shandong, China [2] Qilu Hospital of Shandong University, Jinan, China [3] Shandong Provincial School Key laboratory for Protein Science of Chronic Degenerative Diseases, Jinan, Shandong, China
| | - Hao-an Wang
- Department of Human Biology, University of Toronto, Toronto, Ontario, Canada
| | - Wen-bo Wang
- 1] Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University, School of Medicine, Jinan, Shandong, China [2] Qilu Hospital of Shandong University, Jinan, China [3] Shandong Provincial School Key laboratory for Protein Science of Chronic Degenerative Diseases, Jinan, Shandong, China
| | - Fan-guo Meng
- Yangtze Delta Region Institute of Tsinghua University, Zhejiang, China
| | - Xiao Yu
- 1] Shandong Provincial School Key laboratory for Protein Science of Chronic Degenerative Diseases, Jinan, Shandong, China [2] Department of Physiology, Shandong University, School of Medicine, Jinan, Shandong, China
| | - Jin-peng Sun
- 1] Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University, School of Medicine, Jinan, Shandong, China [2] Shandong Provincial School Key laboratory for Protein Science of Chronic Degenerative Diseases, Jinan, Shandong, China [3] Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| |
Collapse
|
18
|
Humphrey S, Kirby R, Rudloff E. Magnesium physiology and clinical therapy in veterinary critical care. J Vet Emerg Crit Care (San Antonio) 2014; 25:210-25. [PMID: 25427407 DOI: 10.1111/vec.12253] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 09/30/2014] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To review magnesium physiology including absorption, excretion, and function within the body, causes of magnesium abnormalities, and the current applications of magnesium monitoring and therapy in people and animals. ETIOLOGY Magnesium plays a pivotal role in energy production and specific functions in every cell in the body. Disorders of magnesium can be correlated with severity of disease, length of hospital stay, and recovery of the septic patient. Hypermagnesemia is seen infrequently in people and animals with significant consequences reported. Hypomagnesemia is more common in critically ill people and animals, and can be associated with platelet, immune system, neurological, and cardiovascular dysfunction as well as alterations in insulin responsiveness and electrolyte imbalance. DIAGNOSIS Measurement of serum ionized magnesium in critically or chronically ill veterinary patients is practical and provides information necessary for stabilization and treatment. Tissue magnesium concentrations may be assessed using nuclear magnetic resonance spectroscopy as well as through the application of fluorescent dye techniques. THERAPY Magnesium infusions may play a therapeutic role in reperfusion injury, myocardial ischemia, cerebral infarcts, systemic inflammatory response syndromes, tetanus, digitalis toxicity, bronchospasms, hypercoagulable states, and as an adjunct to specific anesthetic or analgesic protocols. Further veterinary studies are needed to establish the frequency and importance of magnesium disorders in animals and the potential benefit of magnesium infusions as a therapeutic adjunct to specific diseases. PROGNOSIS The prognosis for most patients with magnesium disorders is variable and largely dependent on the underlying cause of the disorder.
Collapse
Affiliation(s)
- Sarah Humphrey
- From the Animal Emergency Center and Specialty Services, Glendale, WI 52309
| | | | | |
Collapse
|
19
|
Lu X, Yarbrough WG. Negative regulation of RelA phosphorylation: emerging players and their roles in cancer. Cytokine Growth Factor Rev 2014; 26:7-13. [PMID: 25438737 DOI: 10.1016/j.cytogfr.2014.09.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 09/03/2014] [Indexed: 01/25/2023]
Abstract
NF-κB signaling contributes to human disease processes, notably inflammatory diseases and cancer. Many advances have been made in understanding mechanisms responsible for abnormal NF-κB activation with RelA post-translational modification, particularly phosphorylation, proven to be critical for RelA function. While the majority of studies have focused on identifying kinases responsible for NF-κB phosphorylation and pathway activation, recently progress has also been made in understanding the negative regulators important for restraining RelA activity. Here we summarize negative regulators of RelA phosphorylation, their targeting sites in RelA and biological functions through negative regulation of RelA activation. Finally, we emphasize the tumor suppressor-like roles that these negative regulators can assume in human cancers.
Collapse
Affiliation(s)
- Xinyuan Lu
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA; Department of Medicine, University of California at San Francisco, San Francisco, CA, USA
| | - Wendell G Yarbrough
- Division of Otolaryngology, Department of Surgery, Yale University, New Haven, CT, USA; Department of Pathology, Yale University, New Haven, CT, USA; Yale Cancer Center, New Haven, CT, USA.
| |
Collapse
|
20
|
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
|
21
|
Pan C, Liu HD, Gong Z, Yu X, Hou XB, Xie DD, Zhu XB, Li HW, Tang JY, Xu YF, Yu JQ, Zhang LY, Fang H, Xiao KH, Chen YG, Wang JY, Pang Q, Chen W, Sun JP. Cadmium is a potent inhibitor of PPM phosphatases and targets the M1 binding site. Sci Rep 2014; 3:2333. [PMID: 23903585 PMCID: PMC3730172 DOI: 10.1038/srep02333] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 06/24/2013] [Indexed: 11/16/2022] Open
Abstract
The heavy metal cadmium is a non-degradable pollutant. By screening the effects of a panel of metal ions on the phosphatase activity, we unexpectedly identified cadmium as a potent inhibitor of PPM1A and PPM1G. In contrast, low micromolar concentrations of cadmium did not inhibit PP1 or tyrosine phosphatases. Kinetic studies revealed that cadmium inhibits PPM phosphatases through the M1 metal ion binding site. In particular, the negative charged D441 in PPM1G specific recognized cadmium. Our results suggest that cadmium is likely a potent inhibitor of most PPM family members except for PHLPPs. Furthermore, we demonstrated that cadmium inhibits PPM1A-regulated MAPK signaling and PPM1G-regulated AKT signaling potently in vivo. Cadmium reversed PPM1A-induced cell cycle arrest and cadmium insensitive PPM1A mutant rescued cadmium induced cell death. Taken together, these findings provide a better understanding of the effects of the toxicity of cadmium in the contexts of human physiology and pathology.
Collapse
Affiliation(s)
- Chang Pan
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University, School of Medicine, Jinan, Shandong 250012, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Lu X, An H, Jin R, Zou M, Guo Y, Su PF, Liu D, Shyr Y, Yarbrough WG. PPM1A is a RelA phosphatase with tumor suppressor-like activity. Oncogene 2013; 33:2918-27. [PMID: 23812431 DOI: 10.1038/onc.2013.246] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 05/06/2013] [Accepted: 05/20/2013] [Indexed: 12/28/2022]
Abstract
Nuclear factor-κB (NF-κB) signaling contributes to human disease processes, notably inflammatory diseases and cancer. NF-κB has a role in tumorigenesis and tumor growth, as well as promotion of metastases. Mechanisms responsible for abnormal NF-κB activation are not fully elucidated; however, RelA phosphorylation, particularly at serine residues S536 and S276, is critical for RelA function. Kinases that phosphorylate RelA promote oncogenic behaviors, suggesting that phosphatases targeting RelA could have tumor-inhibiting activities; however, few RelA phosphatases have been identified. Here, we identified tumor inhibitory and RelA phosphatase activities of the protein phosphatase 2C (PP2C) phosphatase family member, PPM1A. We show that PPM1A directly dephosphorylated RelA at residues S536 and S276 and selectively inhibited NF-κB transcriptional activity, resulting in decreased expression of monocyte chemotactic protein-1/chemokine (C-C motif) ligand 2 and interleukin-6, cytokines implicated in cancer metastasis. PPM1A depletion enhanced NF-κB-dependent cell invasion, whereas PPM1A expression inhibited invasion. Analyses of human expression data revealed that metastatic prostate cancer deposits had lower PPM1A expression compared with primary tumors without distant metastases. A hematogenous metastasis mouse model revealed that PPM1A expression inhibited bony metastases of prostate cancer cells after vascular injection. In summary, our findings suggest that PPM1A is a RelA phosphatase that regulates NF-κB activity and that PPM1A has tumor suppressor-like activity. Our analyses also suggest that PPM1A inhibits prostate cancer metastases and as neither gene deletions nor inactivating mutations of PPM1A have been described, increasing PPM1A activity in tumors represents a potential therapeutic strategy to inhibit NF-κB signaling or bony metastases in human cancer.
Collapse
Affiliation(s)
- X Lu
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - H An
- 1] Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA [2] Division of Surgical Sciences, Vanderbilt University, Nashville, TN, USA
| | - R Jin
- 1] Vanderbilt Prostate Cancer Center, Vanderbilt University, Nashville, TN, USA [2] Department of Urology, Vanderbilt University, Nashville, TN, USA
| | - M Zou
- Division of Otolaryngology, Department of Surgery, Yale University, New Haven, CT, USA
| | - Y Guo
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - P-F Su
- Department of Biostatistics, Vanderbilt University, Nashville, TN, USA
| | - D Liu
- Division of Otolaryngology, Department of Surgery, Yale University, New Haven, CT, USA
| | - Y Shyr
- Department of Biostatistics, Vanderbilt University, Nashville, TN, USA
| | - W G Yarbrough
- 1] Division of Otolaryngology, Department of Surgery, Yale University, New Haven, CT, USA [2] Department of Pathology, Yale University, New Haven, CT, USA [3] Yale Cancer Center, New Haven, CT, USA
| |
Collapse
|