1
|
Runa F, Ortiz-Soto G, de Barros NR, Kelber JA. Targeting SMAD-Dependent Signaling: Considerations in Epithelial and Mesenchymal Solid Tumors. Pharmaceuticals (Basel) 2024; 17:326. [PMID: 38543112 PMCID: PMC10975212 DOI: 10.3390/ph17030326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 04/01/2024] Open
Abstract
SMADs are the canonical intracellular effector proteins of the TGF-β (transforming growth factor-β). SMADs translocate from plasma membrane receptors to the nucleus regulated by many SMAD-interacting proteins through phosphorylation and other post-translational modifications that govern their nucleocytoplasmic shuttling and subsequent transcriptional activity. The signaling pathway of TGF-β/SMAD exhibits both tumor-suppressing and tumor-promoting phenotypes in epithelial-derived solid tumors. Collectively, the pleiotropic nature of TGF-β/SMAD signaling presents significant challenges for the development of effective cancer therapies. Here, we review preclinical studies that evaluate the efficacy of inhibitors targeting major SMAD-regulating and/or -interacting proteins, particularly enzymes that may play important roles in epithelial or mesenchymal compartments within solid tumors.
Collapse
Affiliation(s)
- Farhana Runa
- Department of Biology, California State University Northridge, Northridge, CA 91330, USA
| | | | | | - Jonathan A Kelber
- Department of Biology, California State University Northridge, Northridge, CA 91330, USA
- Department of Biology, Baylor University, Waco, TX 76706, USA
| |
Collapse
|
2
|
Ge Q, Shi Z, Zou KA, Ying J, Chen J, Yuan W, Wang W, Xiao L, Lin X, Chen D, Feng XH, Wang PE, Tong P, Jin H. Protein phosphatase PPM1A inhibition attenuates osteoarthritis via regulating TGF-β/Smad2 signaling in chondrocytes. JCI Insight 2023; 8:166688. [PMID: 36752205 PMCID: PMC9926971 DOI: 10.1172/jci.insight.166688] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/21/2022] [Indexed: 02/09/2023] Open
Abstract
TGF-β signaling is crucial for modulating osteoarthritis (OA), and protein phosphatase magnesium-dependent 1A (PPM1A) has been reported as a phosphatase of SMAD2 and regulates TGF-β signaling, while the role of PPM1A in cartilage homeostasis and OA development remains largely unexplored. In this study, we found increased PPM1A expression in OA chondrocytes and confirmed the interaction between PPM1A and phospho-SMAD2 (p-SMAD2). Importantly, our data show that PPM1A KO substantially protected mice treated with destabilization of medial meniscus (DMM) surgery against cartilage degeneration and subchondral sclerosis. Additionally, PPM1A ablation reduced the cartilage catabolism and cell apoptosis after the DMM operation. Moreover, p-SMAD2 expression in chondrocytes from KO mice was higher than that in WT controls with DMM induction. However, intraarticular injection with SD-208, repressing TGF-β/SMAD2 signaling, dramatically abolished protective phenotypes in PPM1A-KO mice. Finally, a specific pharmacologic PPM1A inhibitor, Sanguinarine chloride (SC) or BC-21, was able to ameliorate OA severity in C57BL/6J mice. In summary, our study identified PPM1A as a pivotal regulator of cartilage homeostasis and demonstrated that PPM1A inhibition attenuates OA progression via regulating TGF-β/SMAD2 signaling in chondrocytes and provided PPM1A as a potential target for OA treatment.
Collapse
Affiliation(s)
- Qinwen Ge
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhenyu Shi
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Kai-ao Zou
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jun Ying
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiali Chen
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenhua Yuan
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Weidong Wang
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Department of Orthopedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Luwei Xiao
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xia Lin
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xin-Hua Feng
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute and
| | - Ping-er Wang
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Peijian Tong
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hongting Jin
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Life Sciences Institute, Zhejiang University, Hangzhou, China
| |
Collapse
|
3
|
Zhang L, Gao J, Gong A, Dong Y, Hao X, Wang X, Zheng J, Ma W, Song Y, Zhang J, Xu W. The Long Noncoding RNA LINC00963 Inhibits Corneal Fibrosis Scar Formation by Targeting miR-143-3p. DNA Cell Biol 2022; 41:400-409. [PMID: 35262384 PMCID: PMC9063159 DOI: 10.1089/dna.2021.1034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Corneal fibrosis is a complication of severe corneal injury, one of the major causes of vision loss. The formation of myofibroblasts has emerged as a key stimulative factor of corneal fibrosis. In the current study, we focused on the role of LINC00963 in regulating corneal fibrosis. Transforming growth factor β1 (TGF-β1) was used to induce human corneal stromal cells differentiating into corneal myofibroblasts, and the significant increase of α-smooth muscle actin (α-SMA) was verified by quantitative real-time PCR (qRT-PCR), western blot, and immunofluorescence, respectively. LINC00963 was identified to be one-half decreased compared with nonstimulated human corneal stromal cells, indicating that it might play a role in corneal fibrosis. Interestingly, overexpression of LINC00963 resulted in decreased formation of myofibroblasts indicating that it might exhibit an inhibiting effect. Moreover, bioinformatics tool was applied to predict the downstream target of LINC00963. We investigated that LINC00963 suppressed α-SMA induced by TGF-β1 in corneal fibroblasts, at least in part, by downregulating the expression of miR-143-3p. In addition, either LINC00963 promotion or miR-143-3p inhibition could significantly decrease myofibroblast contractility and collagen I and III secretion, which are the key to contribute to corneal fibrosis. Taken together, our study identified LINC00963 as a promising therapeutic target.
Collapse
Affiliation(s)
- Lixia Zhang
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China.,Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jinning Gao
- Center for Molecular Genetics, Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Anjing Gong
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanhan Dong
- Center for Molecular Genetics, Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Xiaodan Hao
- Center for Molecular Genetics, Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Xuekang Wang
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China.,Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jian Zheng
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China
| | - Wenmeng Ma
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China
| | - Yiying Song
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China
| | - Jie Zhang
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China
| | - Wenhua Xu
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China
| |
Collapse
|
4
|
Metal dependent protein phosphatase PPM family in cardiac health and diseases. Cell Signal 2021; 85:110061. [PMID: 34091011 PMCID: PMC9107372 DOI: 10.1016/j.cellsig.2021.110061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/20/2022]
Abstract
Protein phosphorylation and dephosphorylation is central to signal transduction in nearly every aspect of cellular function, including cardiovascular regulation and diseases. While protein kinases are often regarded as the molecular drivers in cellular signaling with high specificity and tight regulation, dephosphorylation mediated by protein phosphatases is also gaining increasing appreciation as an important part of the signal transduction network essential for the robustness, specificity and homeostasis of cell signaling. Metal dependent protein phosphatases (PPM, also known as protein phosphatases type 2C, PP2C) belong to a highly conserved family of protein phosphatases with unique biochemical and molecular features. Accumulating evidence also indicates important and specific functions of individual PPM isoform in signaling and cellular processes, including proliferation, senescence, apoptosis and metabolism. At the physiological level, abnormal PPM expression and activity have been implicated in major human diseases, including cancer, neurological and cardiovascular disorders. Finally, inhibitors for some of the PPM members have been developed as a potential therapeutic strategy for human diseases. In this review, we will focus on the background information about the biochemical and molecular features of major PPM family members, with emphasis on their demonstrated or potential roles in cardiac pathophysiology. The current challenge and potential directions for future investigations will also be highlighted.
Collapse
|
5
|
Li X, Liu C, Zhu Y, Rao H, Liu M, Gui L, Feng W, Tang H, Xu J, Gao WQ, Li L. SETD2 epidermal deficiency promotes cutaneous wound healing via activation of AKT/mTOR Signalling. Cell Prolif 2021; 54:e13045. [PMID: 33949020 PMCID: PMC8168411 DOI: 10.1111/cpr.13045] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/26/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Objectives Cutaneous wound healing is one of the major medical problems worldwide. Epigenetic modifiers have been identified as important players in skin development, homeostasis and wound repair. SET domain–containing 2 (SETD2) is the only known histone H3K36 tri‐methylase; however, its role in skin wound healing remains unclear. Materials and Methods To elucidate the biological role of SETD2 in wound healing, conditional gene targeting was used to generate epidermis‐specific Setd2‐deficient mice. Wound‐healing experiments were performed on the backs of mice, and injured skin tissues were collected and analysed by haematoxylin and eosin (H&E) and immunohistochemical staining. In vitro, CCK8 and scratch wound‐healing assays were performed on Setd2‐knockdown and Setd2‐overexpression human immortalized keratinocyte cell line (HaCaT). In addition, RNA‐seq and H3K36me3 ChIP‐seq analyses were performed to identify the dysregulated genes modulated by SETD2. Finally, the results were validated in functional rescue experiments using AKT and mTOR inhibitors (MK2206 and rapamycin). Results Epidermis‐specific Setd2‐deficient mice were successfully established, and SETD2 deficiency resulted in accelerated re‐epithelialization during cutaneous wound healing by promoting keratinocyte proliferation and migration. Furthermore, the loss of SETD2 enhanced the scratch closure and proliferation of keratinocytes in vitro. Mechanistically, the deletion of Setd2 resulted in the activation of AKT/mTOR signalling pathway, while the pharmacological inhibition of AKT and mTOR with MK2206 and rapamycin, respectively, delayed wound closure. Conclusions Our results showed that SETD2 loss promoted cutaneous wound healing via the activation of AKT/mTOR signalling.
Collapse
Affiliation(s)
- Xiaoxue Li
- State Key Laboratory of Oncogenes and Related Genes, School of Medicine and School of Biomedical Engineering, Renji Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China.,School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Changwei Liu
- State Key Laboratory of Oncogenes and Related Genes, School of Medicine and School of Biomedical Engineering, Renji Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China.,School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yiwen Zhu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hanyu Rao
- State Key Laboratory of Oncogenes and Related Genes, School of Medicine and School of Biomedical Engineering, Renji Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China.,School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Min Liu
- State Key Laboratory of Oncogenes and Related Genes, School of Medicine and School of Biomedical Engineering, Renji Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China.,School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Liming Gui
- State Key Laboratory of Oncogenes and Related Genes, School of Medicine and School of Biomedical Engineering, Renji Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China.,School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Wenxin Feng
- State Key Laboratory of Oncogenes and Related Genes, School of Medicine and School of Biomedical Engineering, Renji Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China.,School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Huayuan Tang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Jin Xu
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, School of Medicine and School of Biomedical Engineering, Renji Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China.,School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Li Li
- State Key Laboratory of Oncogenes and Related Genes, School of Medicine and School of Biomedical Engineering, Renji Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China.,School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
6
|
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
|
7
|
Metal-dependent Ser/Thr protein phosphatase PPM family: Evolution, structures, diseases and inhibitors. Pharmacol Ther 2020; 215:107622. [PMID: 32650009 DOI: 10.1016/j.pharmthera.2020.107622] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
Protein phosphatases and kinases control multiple cellular events including proliferation, differentiation, and stress responses through regulating reversible protein phosphorylation, the most important post-translational modification. Members of metal-dependent protein phosphatase (PPM) family, also known as PP2C phosphatases, are Ser/Thr phosphatases that bind manganese/magnesium ions (Mn2+/Mg2+) in their active center and function as single subunit enzymes. In mammals, there are 20 isoforms of PPM phosphatases: PPM1A, PPM1B, PPM1D, PPM1E, PPM1F, PPM1G, PPM1H, PPM1J, PPM1K, PPM1L, PPM1M, PPM1N, ILKAP, PDP1, PDP2, PHLPP1, PHLPP2, PP2D1, PPTC7, and TAB1, whereas there are only 8 in yeast. Phylogenetic analysis of the DNA sequences of vertebrate PPM isoforms revealed that they can be divided into 12 different classes: PPM1A/PPM1B/PPM1N, PPM1D, PPM1E/PPM1F, PPM1G, PPM1H/PPM1J/PPM1M, PPM1K, PPM1L, ILKAP, PDP1/PDP2, PP2D1/PHLPP1/PHLPP2, TAB1, and PPTC7. PPM-family members have a conserved catalytic core region, which contains the metal-chelating residues. The different isoforms also have isoform specific regions within their catalytic core domain and terminal domains, and these regions may be involved in substrate recognition and/or functional regulation of the phosphatases. The twenty mammalian PPM phosphatases are involved in regulating diverse cellular functions, such as cell cycle control, cell differentiation, immune responses, and cell metabolism. Mutation, overexpression, or deletion of the PPM phosphatase gene results in abnormal cellular responses, which lead to various human diseases. This review focuses on the structures and biological functions of the PPM-phosphatase family and their associated diseases. The development of specific inhibitors against the PPM phosphatase family as a therapeutic strategy will also be discussed.
Collapse
|
8
|
Maarof M, Lokanathan Y, Ruszymah HI, Saim A, Chowdhury SR. Proteomic Analysis of Human Dermal Fibroblast Conditioned Medium (DFCM). Protein J 2019; 37:589-607. [PMID: 30343346 DOI: 10.1007/s10930-018-9800-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Growth factors and extracellular matrix (ECM) proteins are involved in wound healing. Human dermal fibroblasts secrete wound-healing mediators in culture medium known as dermal fibroblast conditioned medium (DFCM). However, the composition and concentration of the secreted proteins differ with culture conditions and environmental factors. We cultured human skin fibroblasts in vitro using serum-free keratinocyte-specific media (EpiLife™ Medium [KM1] and defined keratinocyte serum-free medium [KM2]) and serum-free fibroblast-specific medium (FM) to obtain DFCM-KM1, DFCM-KM2 and DFCM-FM, respectively. We identified and compared their proteomic profiles using bicinchoninic acid assay (BCA), 1-dimensional sodium dodecyl sulphate-polyacrylamide gel electrophoresis (1D SDS-PAGE), enzyme-linked immunosorbent assay (ELISA), matrix-assisted laser desorption ionisation-time-of-flight mass spectrometry (MALDI-TOF/TOF MS/MS) and liquid chromatography MS (LC-MS/MS). DFCM-KM1 and DFCM-KM2 had higher protein concentrations than DFCM-FM but not statistically significant. MALDI-TOF/TOF MS identified the presence of fibronectin, serotransferrin, serpin and serum albumin. LC-MS/MS and bioinformatics analysis identified 59, 46 and 58 secreted proteins in DFCM-KM1, DFCM-KM2 and DFCM-FM, respectively. The most significant biological processes identified in gene ontology were cellular process, metabolic process, growth and biological regulation. STRING® analysis showed that most secretory proteins in the DFCMs were associated with biological processes (e.g. wound healing and ECM organisation), molecular function (e.g. ECM binding) and cellular component (e.g. extracellular space). ELISA confirmed the presence of fibronectin and collagen in the DFCMs. In conclusion, DFCM secretory proteins are involved in cell adhesion, attachment, proliferation and migration, which were demonstrated to have potential wound-healing effects by in vitro and in vivo studies.
Collapse
Affiliation(s)
- Manira Maarof
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaccob Latiff, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Yogeswaran Lokanathan
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaccob Latiff, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Hj Idrus Ruszymah
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaccob Latiff, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Aminuddin Saim
- Ear Nose & Throat Consultant Clinic, Ampang Puteri Specialist Hospital, 1 Jalan Mamanda 9, 68000, Ampang, Selangor, Malaysia
| | - Shiplu Roy Chowdhury
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaccob Latiff, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia.
| |
Collapse
|
9
|
Wu Y, Zhong JL, Hou N, Sun Y, Ma B, Nisar MF, Teng Y, Tan Z, Chen K, Wang Y, Yang X. MicroRNA Let-7b inhibits keratinocyte migration in cutaneous wound healing by targeting IGF2BP2. Exp Dermatol 2017; 26:116-123. [DOI: 10.1111/exd.13164] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Yan Wu
- State Key Laboratory of Proteomics; Genetic Laboratory of Development and Diseases; Institute of Biotechnology; Beijing China
- 111 Project Laboratory of Biomechanics and Tissue Repair; College of Bioengineering & Key Laboratory of Biorheological Science and Technology; Ministry of Education Chongqing University; Chongqing China
- College of Life Sciences; Jiangsu University; Zhenjiang China
| | - Julia Li Zhong
- 111 Project Laboratory of Biomechanics and Tissue Repair; College of Bioengineering & Key Laboratory of Biorheological Science and Technology; Ministry of Education Chongqing University; Chongqing China
| | - Ning Hou
- State Key Laboratory of Proteomics; Genetic Laboratory of Development and Diseases; Institute of Biotechnology; Beijing China
| | - Yaolan Sun
- State Key Laboratory of Proteomics; Genetic Laboratory of Development and Diseases; Institute of Biotechnology; Beijing China
| | - Benting Ma
- State Key Laboratory of Proteomics; Genetic Laboratory of Development and Diseases; Institute of Biotechnology; Beijing China
| | - Muhammad Farrukh Nisar
- 111 Project Laboratory of Biomechanics and Tissue Repair; College of Bioengineering & Key Laboratory of Biorheological Science and Technology; Ministry of Education Chongqing University; Chongqing China
| | - Yan Teng
- State Key Laboratory of Proteomics; Genetic Laboratory of Development and Diseases; Institute of Biotechnology; Beijing China
| | - Zhaoli Tan
- State Key Laboratory of Proteomics; Genetic Laboratory of Development and Diseases; Institute of Biotechnology; Beijing China
| | - Keping Chen
- College of Life Sciences; Jiangsu University; Zhenjiang China
| | - Youliang Wang
- State Key Laboratory of Proteomics; Genetic Laboratory of Development and Diseases; Institute of Biotechnology; Beijing China
| | - Xiao Yang
- State Key Laboratory of Proteomics; Genetic Laboratory of Development and Diseases; Institute of Biotechnology; Beijing China
| |
Collapse
|
10
|
Xiang W, Zhang Q, Lin X, Wu S, Zhou Y, Meng F, Fan Y, Shen T, Xiao M, Xia Z, Zou J, Feng XH, Xu P. PPM1A silences cytosolic RNA sensing and antiviral defense through direct dephosphorylation of MAVS and TBK1. SCIENCE ADVANCES 2016; 2:e1501889. [PMID: 27419230 PMCID: PMC4942338 DOI: 10.1126/sciadv.1501889] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 05/31/2016] [Indexed: 05/20/2023]
Abstract
Cytosolic RNA sensing is a prerequisite for initiation of innate immune response against RNA viral pathogens. Signaling through RIG-I (retinoic acid-inducible gene I)-like receptors (RLRs) to TBK1 (Tank-binding kinase 1)/IKKε (IκB kinase ε) kinases is transduced by mitochondria-associated MAVS (mitochondrial antiviral signaling protein). However, the precise mechanism of how MAVS-mediated TBK1/IKKε activation is strictly controlled still remains obscure. We reported that protein phosphatase magnesium-dependent 1A (PPM1A; also known as PP2Cα), depending on its catalytic ability, dampened the RLR-IRF3 (interferon regulatory factor 3) axis to silence cytosolic RNA sensing signaling. We demonstrated that PPM1A was an inherent partner of the TBK1/IKKε complex, targeted both MAVS and TBK1/IKKε for dephosphorylation, and thus disrupted MAVS-driven formation of signaling complex. Conversely, a high level of MAVS can dissociate the TBK1/PPM1A complex to override PPM1A-mediated inhibition. Loss of PPM1A through gene ablation in human embryonic kidney 293 cells and mouse primary macrophages enabled robustly enhanced antiviral responses. Consequently, Ppm1a(-/-) mice resisted to RNA virus attack, and transgenic zebrafish expressing PPM1A displayed profoundly increased RNA virus vulnerability. These findings identify PPM1A as the first known phosphatase of MAVS and elucidate the physiological function of PPM1A in antiviral immunity on whole animals.
Collapse
Affiliation(s)
- Weiwen Xiang
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Qian Zhang
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Xia Lin
- Michael E. DeBakey Department of Surgery and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shiying Wu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Yao Zhou
- Eye Center of the Second Affiliated Hospital School of Medicine, Institute of Translational Medicine, Zhejiang University, Hangzhou 310058, China
| | - Fansen Meng
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Yunyun Fan
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Tao Shen
- Michael E. DeBakey Department of Surgery and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mu Xiao
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Zongping Xia
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Jian Zou
- Eye Center of the Second Affiliated Hospital School of Medicine, Institute of Translational Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xin-Hua Feng
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
- Michael E. DeBakey Department of Surgery and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Corresponding author. (X.-H.F.); (P.X.)
| | - Pinglong Xu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
- Corresponding author. (X.-H.F.); (P.X.)
| |
Collapse
|
11
|
Samarakoon R, Rehfuss A, Khakoo NS, Falke LL, Dobberfuhl AD, Helo S, Overstreet JM, Goldschmeding R, Higgins PJ. Loss of expression of protein phosphatase magnesium-dependent 1A during kidney injury promotes fibrotic maladaptive repair. FASEB J 2016; 30:3308-3320. [PMID: 27328942 DOI: 10.1096/fj.201500105r] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 06/14/2016] [Indexed: 11/11/2022]
Abstract
Protein phosphatase magnesium-dependent-1A (PPM1A) dephosphorylates SMAD2/3, which suppresses TGF-β signaling in keratinocytes and during Xenopus development; however, potential involvement of PPM1A in chronic kidney disease is unknown. PPM1A expression was dramatically decreased in the tubulointerstitium in obstructive and aristolochic acid nephropathy, which correlates with progression of fibrotic disease. Stable silencing of PPM1A in human kidney-2 human renal epithelial cells increased SMAD3 phosphorylation, stimulated expression of fibrotic genes, induced dedifferentiation, and orchestrated epithelial cell-cycle arrest via SMAD3-mediated connective tissue growth factor and plasminogen activator inhibitor-1 up-regulation. PPM1A stable suppression in normal rat kidney-49 renal fibroblasts, in contrast, promoted a SMAD3-dependent connective tissue growth factor and plasminogen activator inhibitor-1-induced proliferative response. Paracrine factors secreted by PPM1A-depleted epithelial cells augmented fibroblast proliferation (>50%) compared with controls. PPM1A suppression in renal cells further enhanced TGF-β1-induced SMAD3 phosphorylation and fibrotic gene expression, whereas PPM1A overexpression inhibited both responses. Moreover, phosphate tensin homolog on chromosome 10 depletion in human kidney-2 cells resulted in loss of expression and decreased nuclear levels of PPM1A, which enhanced SMAD3-mediated fibrotic gene induction and growth arrest that were reversed by ectopic PPM1A expression. Thus, phosphate tensin homolog on chromosome 10 is an upstream regulator of renal PPM1A deregulation. These findings establish PPM1A as a novel repressor of the SMAD3 pathway in renal fibrosis and as a new therapeutic target in patients with chronic kidney disease.-Samarakoon, R., Rehfuss, A., Khakoo, N. S., Falke, L. L., Dobberfuhl, A. D., Helo, S., Overstreet, J. M., Goldschmeding, R., Higgins, P. J. Loss of expression of protein phosphatase magnesium-dependent 1A during kidney injury promotes fibrotic maladaptive repair.
Collapse
Affiliation(s)
- Rohan Samarakoon
- Center for Cell Biology and Cancer Research, Albany Medical Center, Albany, New York, USA;
| | | | - Nidah S Khakoo
- Center for Cell Biology and Cancer Research, Albany Medical Center, Albany, New York, USA
| | - Lucas L Falke
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Amy D Dobberfuhl
- Department of Urology, Stanford University School of Medicine, Stanford, California, USA; and
| | - Sevann Helo
- Division of Urology, Albany Medical Center, Albany, New York, USA
| | - Jessica M Overstreet
- Division of Nephrology and Hypertension, Vanderbilt Medical School, Nashville, Tennessee, USA
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Paul J Higgins
- Center for Cell Biology and Cancer Research, Albany Medical Center, Albany, New York, USA;
| |
Collapse
|
12
|
Genes related with apoptosis by inflammation in diabetic keratocytes. Genes Genomics 2015. [DOI: 10.1007/s13258-015-0290-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
13
|
Li Z, Liu G, Sun L, Teng Y, Guo X, Jia J, Sha J, Yang X, Chen D, Sun Q. PPM1A regulates antiviral signaling by antagonizing TBK1-mediated STING phosphorylation and aggregation. PLoS Pathog 2015; 11:e1004783. [PMID: 25815785 PMCID: PMC4376777 DOI: 10.1371/journal.ppat.1004783] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 03/03/2015] [Indexed: 12/12/2022] Open
Abstract
Stimulator of interferon genes (STING, also known as MITA and ERIS) is critical in protecting the host against DNA pathogen invasion. However, the molecular mechanism underlying the regulation of STING remains unclear. Here, we show that PPM1A negatively regulates antiviral signaling by targeting STING in its phosphatase activity-dependent manner, and in a line with this, PPM1A catalytically dephosphorylates STING and TBK1 in vitro. Importantly, we provide evidence that whereas TBK1 promotes STING aggregation in a phosphorylation-dependent manner, PPM1A antagonizes STING aggregation by dephosphorylating both STING and TBK1, emphasizing that phosphorylation is crucial for the efficient activation of STING. Our findings demonstrate a novel regulatory circuit in which STING and TBK1 reciprocally regulate each other to enable efficient antiviral signaling activation, and PPM1A dephosphorylates STING and TBK1, thereby balancing this antiviral signal transduction. Innate antiviral immunity is essential for the host defense system that rapidly detects and eliminates invading viruses. STING, an endoplasmic reticulum (ER)-associated protein, plays important roles in the activation of type I IFN in response to DNA virus infection. Whereas excessive activation of STING can potentially cause lethal inflammatory diseases, STING activity must thus be precisely controlled to ensure the proper antiviral signaling transduction. However, the mechanisms of how STING activation is regulated are not fully understood. In this study, we find that PPM1A physically interacts with STING and negatively regulates STING-mediated antiviral signaling. Mechanistically, we find that PPM1A functions as a phosphatase that targets both STING and TBK1 for their dephosphorylation. Moreover, our study demonstrates that while TBK1 enhances STING aggregation in a kinase activity-dependent manner, PPM1A suppresses STING aggregation by dephosphorylating both STING and TBK1. Collectively, our study not only reveals that STING and TBK1 reciprocally regulate each other’s activity to elicit antiviral signaling, but also shows that PPM1A antagonizes STING aggregation by targeting both STING and TBK1, thereby maintaining proper antiviral responses.
Collapse
Affiliation(s)
- Zexing Li
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Chinese Academy of Sciences, Chaoyang District, Beijing, China
| | - Ge Liu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, China
| | - Liwei Sun
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Chinese Academy of Sciences, Chaoyang District, Beijing, China
| | - Yan Teng
- State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, China
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Jianhang Jia
- Markey Cancer Center, Department of Molecular and Cellular Biochemistry, The University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Jiahao Sha
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Xiao Yang
- State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, China
| | - Dahua Chen
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, China
| | - Qinmiao Sun
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Chinese Academy of Sciences, Chaoyang District, Beijing, China
- * E-mail:
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Organista-Juárez D, Carretero-Ortega J, Vicente-Fermín O, Vázquez-Victorio G, Sosa-Garrocho M, Vázquez-Prado J, Macías-Silva M, Reyes-Cruz G. Calcium-sensing receptor inhibits TGF-β-signaling by decreasing Smad2 phosphorylation. IUBMB Life 2013; 65:1035-42. [DOI: 10.1002/iub.1232] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 11/12/2013] [Indexed: 12/13/2022]
Affiliation(s)
- Diana Organista-Juárez
- Department of Cell Biology; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional; Apartado postal 14-740 México D.F. 07000 Mexico
| | - Jorge Carretero-Ortega
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México; México D.F. 04510 Mexico
| | - Onasis Vicente-Fermín
- Department of Cell Biology; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional; Apartado postal 14-740 México D.F. 07000 Mexico
| | - Genaro Vázquez-Victorio
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México; México D.F. 04510 Mexico
| | - Marcela Sosa-Garrocho
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México; México D.F. 04510 Mexico
| | - José Vázquez-Prado
- Department of Pharmacology; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional; Apartado postal 14-740 México D.F. 07000 Mexico
| | - Marina Macías-Silva
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México; México D.F. 04510 Mexico
| | - Guadalupe Reyes-Cruz
- Department of Cell Biology; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional; Apartado postal 14-740 México D.F. 07000 Mexico
| |
Collapse
|
17
|
Finnson KW, Arany PR, Philip A. Transforming Growth Factor Beta Signaling in Cutaneous Wound Healing: Lessons Learned from Animal Studies. Adv Wound Care (New Rochelle) 2013; 2:225-237. [PMID: 24761336 DOI: 10.1089/wound.2012.0419] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Indexed: 12/11/2022] Open
Abstract
SIGNIFICANCE Wound healing is a complex physiological process involving a multitude of growth factors, among which transforming growth factor beta (TGF-β) has the broadest spectrum of effects. Animal studies have provided key information on the mechanisms of TGF-β action in wound healing and have guided the development of therapeutic strategies targeting the TGF-β pathway to improve wound healing and scarring outcome. RECENT ADVANCES Development of tissue-specific expression systems for overexpression or knockout of TGF-β signaling pathway components has led to novel insight into the role of TGF-β signaling in wound healing. This work has also identified molecules that might serve as molecular targets for the treatment of pathological skin conditions such as chronic wounds and excessive scarring (fibrosis). CRITICAL ISSUES Many of the mouse models with genetic alterations in the TGF-β signaling pathway develop an underlying skin abnormality, which may pose some limitations on the interpretation of wound-healing results obtained in these animals. Also, TGF-β's pleiotropic effects on many cell types throughout all phases of wound healing present a challenge in designing specific strategies for targeting the TGF-β signaling pathway to promote wound healing or reduce scarring. FUTURE DIRECTIONS Further characterization of TGF-β signaling pathway components using inducible tissue-specific overexpression or knockout technology will be needed to corroborate results obtained in mouse models that display a skin phenotype, and to better understand the role of TGF-β signaling during distinct phases of the wound-healing process. Such studies will also provide a better understanding of how TGF-β mediates its autocrine, paracrine, and double paracrine effects on cellular responses in vivo during wound healing.
Collapse
Affiliation(s)
- Kenneth W. Finnson
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal General Hospital, Montreal, Canada
| | - Praveen R. Arany
- Cell Regulation and Control Unit, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Anie Philip
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal General Hospital, Montreal, Canada
| |
Collapse
|
18
|
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
|
19
|
Kim SA, Ryu HW, Lee KS, Cho JW. Application of platelet-rich plasma accelerates the wound healing process in acute and chronic ulcers through rapid migration and upregulation of cyclin A and CDK4 in HaCaT cells. Mol Med Rep 2012; 7:476-80. [PMID: 23242428 DOI: 10.3892/mmr.2012.1230] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 11/29/2012] [Indexed: 11/06/2022] Open
Abstract
Application of autologous platelet-rich plasma (PRP) has been used for chronic wound healing. The aim of this study was to evaluate the effect of PRP on the wound healing processes of both acute and chronic ulcers and the underlying molecular mechanisms involved. We treated 16 patients affected by various acute and chronic ulcers with PRP. We performed molecular studies of cell proliferation, migration assays, immunoblotting and chloramphenicol acetyltransferase (CAT) assays in PRP-treated HaCaT keratinocyte cells. PRP treatment induced increased rates of cell proliferation and cell migration of HaCaT cells. In addition, the expression of cyclin A and cyclin dependent kinase (CDK) 4 proteins was markedly increased with a low concentration (0.5%) of PRP treatment in HaCaT cells. In 11 patients with chronic ulcers, including stasis ulcers, diabetic ulcers, venous leg ulcers, livedoid vasculitis, claw foot and traumatic ulcers, 9 patients showed 90-100% epithelization after 15.18 days. In 5 patients with acute ulcers, such as dehiscence, open wound and burn wound, 80-100% epithelization was achieved between 4 to 20 days. Topical application of PRP to acute and chronic skin ulcers significantly accelerated the epithelization process, likely through upregulation of the cell cycle regulatory proteins cyclin A and CDK4.
Collapse
Affiliation(s)
- Sung-Ae Kim
- Department of Dermatology, Keimyung University School of Medicine, Daegu 700-712, Republic of Korea
| | | | | | | |
Collapse
|
20
|
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
|
21
|
Phosphatase PPM1A negatively regulates P-TEFb function in resting CD4(+) T cells and inhibits HIV-1 gene expression. Retrovirology 2012; 9:52. [PMID: 22727189 PMCID: PMC3406988 DOI: 10.1186/1742-4690-9-52] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2012] [Accepted: 06/22/2012] [Indexed: 11/16/2022] Open
Abstract
Background Processive elongation of the integrated HIV-1 provirus is dependent on recruitment of P-TEFb by the viral Tat protein to the viral TAR RNA element. P-TEFb kinase activity requires phosphorylation of Thr186 in the T-loop of the CDK9 subunit. In resting CD4+T cells, low levels of T-loop phosphorylated CDK9 are found, which increase significantly upon activation. This suggests that the phosphorylation status of the T-loop is actively regulated through the concerted actions of cellular proteins such as Ser/Thr phosphatases. We investigated the role of phosphatase PPM1A in regulating CDK9 T-loop phosphorylation and its effect on HIV-1 proviral transcription. Results We found that overexpression of PPM1A inhibits HIV-1 gene expression during viral infection and this required PPM1A catalytic function. Using an artificial CDK tethering system, we further found that PPM1A inhibits CDK9, but not CDK8 mediated activation of the HIV-1 LTR. SiRNA depletion of PPM1A in resting CD4+T cells increased the level of CDK9 T-loop phosphorylation and enhanced HIV-1 gene expression. We also observed that PPM1A protein levels are relatively high in resting CD4+T cells and are not up-regulated upon T cell activation. Conclusions Our results establish a functional link between HIV-1 replication and modulation of CDK9 T-loop phosphorylation by PPM1A. PPM1A represses HIV-1 gene expression by inhibiting CDK9 T-loop phosphorylation, thus reducing the amount of active P-TEFb available for recruitment to the viral LTR. We also infer that PPM1A enzymatic activity in resting and activated CD4+ T cells are likely regulated by as yet undefined factors.
Collapse
|
22
|
Bruce DL, Sapkota GP. Phosphatases in SMAD regulation. FEBS Lett 2012; 586:1897-905. [PMID: 22576046 DOI: 10.1016/j.febslet.2012.02.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 02/02/2012] [Accepted: 02/02/2012] [Indexed: 11/28/2022]
Abstract
SMAD transcription factors are key mediators of the transforming growth factor-beta (TGFß) family of cytokines. Reversible phosphorylation of SMAD proteins plays a key role in regulating their function. Several phosphatases have been proposed to act on SMAD proteins to influence TGFß/BMP signalling. Here we provide an overview of the SMAD regulation by different protein phosphatases and review the evidence supporting each phosphatase as a candidate SMAD-phosphatase.
Collapse
Affiliation(s)
- David L Bruce
- MRC Protein Phosphorylation Unit, College of Life Sciences, University of Dundee, Dow Street, DD1 5EH Dundee, Scotland, UK
| | | |
Collapse
|