1
|
Li X, Si Y, Liang J, Li M, Wang Z, Qin Y, Sun L. Enhancing bone regeneration and immunomodulation via gelatin methacryloyl hydrogel-encapsulated exosomes from osteogenic pre-differentiated mesenchymal stem cells. J Colloid Interface Sci 2024; 672:179-199. [PMID: 38838627 DOI: 10.1016/j.jcis.2024.05.209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/13/2024] [Accepted: 05/27/2024] [Indexed: 06/07/2024]
Abstract
Mesenchymal stem cell-derived exosomes (MSC-Exos) have emerged as promising candidates for cell-free therapy in tissue regeneration. However, the native osteogenic and angiogenic capacities of MSC-Exos are often insufficient to repair critical-sized bone defects, and the underlying immune mechanisms remain elusive. Furthermore, achieving sustained delivery and stable activity of MSC-Exos at the defect site is essential for optimal therapeutic outcomes. Here, we extracted exosomes from osteogenically pre-differentiated human bone marrow mesenchymal stem cells (hBMSCs) by ultracentrifugation and encapsulated them in gelatin methacryloyl (GelMA) hydrogel to construct a composite scaffold. The resulting exosome-encapsulated hydrogel exhibited excellent mechanical properties and biocompatibility, facilitating sustained delivery of MSC-Exos. Osteogenic pre-differentiation significantly enhanced the osteogenic and angiogenic properties of MSC-Exos, promoting osteogenic differentiation of hBMSCs and angiogenesis of human umbilical vein endothelial cells (HUVECs). Furthermore, MSC-Exos induced polarization of Raw264.7 cells from a pro-inflammatory phenotype to an anti-inflammatory phenotype under simulated inflammatory conditions, thereby creating an immune microenvironment conducive to osteogenesis. RNA sequencing and bioinformatics analysis revealed that MSC-Exos activate the p53 pathway through targeted delivery of internal microRNAs and regulate macrophage polarization by reducing DNA oxidative damage. Our study highlights the potential of osteogenic exosome-encapsulated composite hydrogels for the development of cell-free scaffolds in bone tissue engineering.
Collapse
Affiliation(s)
- Xiaorong Li
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yunhui Si
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China.
| | - Jingxian Liang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Mengsha Li
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Zhiwei Wang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Yinying Qin
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Litao Sun
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
2
|
Brewer A, Zhao JF, Fasimoye R, Shpiro N, Macartney TJ, Wood NT, Wightman M, Alessi DR, Sapkota GP. Targeted dephosphorylation of SMAD3 as an approach to impede TGF-β signaling. iScience 2024; 27:110423. [PMID: 39104417 PMCID: PMC11298613 DOI: 10.1016/j.isci.2024.110423] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/09/2024] [Accepted: 06/27/2024] [Indexed: 08/07/2024] Open
Abstract
TGF-β (transforming growth factor-β) signaling is involved in a myriad of cellular processes and its dysregulation has been implicated in many human diseases, including fibrosis and cancer. TGF-β transcriptional responses are controlled by tail phosphorylation of transcription factors SMAD2 and SMAD3 (mothers against decapentaplegic homolog 2/3). Therefore, targeted dephosphorylation of phospho-SMAD3 could provide an innovative mechanism to block some TGF-β-induced transcriptional responses, such as the transcription of SERPINE-1, which encodes plasminogen activator inhibitor 1 (PAI-1). Here, by developing and employing a bifunctional molecule, BDPIC (bromoTAG-dTAG proximity-inducing chimera), we redirected multiple phosphatases, tagged with bromoTAG, to dephosphorylate phospho-SMAD3, tagged with dTAG. Using CRISPR-Cas9 technology, we generated homozygous double knock-in A549 bromoTAG/bromoTAG PPM1H/ dTAG/dTAG SMAD3 cells, in which the BDPIC-induced proximity between bromoTAG-PPM1H and dTAG-SMAD3 led to a robust dephosphorylation of dTAG-SMAD3 and a significant decrease in SERPINE-1 transcription. Our work demonstrates targeted dephosphorylation of phospho-proteins as an exciting modality for rewiring cell signaling.
Collapse
Affiliation(s)
- Abigail Brewer
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Jin-Feng Zhao
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Rotimi Fasimoye
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Natalia Shpiro
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Thomas J. Macartney
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Nicola T. Wood
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Melanie Wightman
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Dario R. Alessi
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Gopal P. Sapkota
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| |
Collapse
|
3
|
Liu Y, Lu S, Yang J, Yang Y, Jiao L, Hu J, Li Y, Yang F, Pang Y, Zhao Y, Gao Y, Liu W, Shu P, Ge W, He Z, Peng X. Analysis of the aging-related biomarker in a nonhuman primate model using multilayer omics. BMC Genomics 2024; 25:639. [PMID: 38926642 PMCID: PMC11209966 DOI: 10.1186/s12864-024-10556-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 06/24/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Aging is a prominent risk factor for diverse diseases; therefore, an in-depth understanding of its physiological mechanisms is required. Nonhuman primates, which share the closest genetic relationship with humans, serve as an ideal model for exploring the complex aging process. However, the potential of the nonhuman primate animal model in the screening of human aging markers is still not fully exploited. Multiomics analysis of nonhuman primate peripheral blood offers a promising approach to evaluate new therapies and biomarkers. This study explores aging-related biomarker through multilayer omics, including transcriptomics (mRNA, lncRNA, and circRNA) and proteomics (serum and serum-derived exosomes) in rhesus monkeys (Macaca mulatta). RESULTS Our findings reveal that, unlike mRNAs and circRNAs, highly expressed lncRNAs are abundant during the key aging period and are associated with cancer pathways. Comparative analysis highlighted exosomal proteins contain more types of proteins than serum proteins, indicating that serum-derived exosomes primarily regulate aging through metabolic pathways. Finally, eight candidate aging biomarkers were identified, which may serve as blood-based indicators for detecting age-related brain changes. CONCLUSIONS Our results provide a comprehensive understanding of nonhuman primate blood transcriptomes and proteomes, offering novel insights into the aging mechanisms for preventing or treating age-related diseases.
Collapse
Affiliation(s)
- Yunpeng Liu
- State Key Laboratory of Respiratory Health and Multimorbidity, National Center of Technology Innovation for Animal Model, National Human Diseases Animal Model Resource Center, NHC Key Laboratory of Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Sciences, CAMS & PUMC, Beijing, 100021, China
| | - Shuaiyao Lu
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China
| | - Jing Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China
| | - Yun Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China
| | - Li Jiao
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China
| | - Jingwen Hu
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China
| | - Yanyan Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China
| | - Fengmei Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China
| | - Yunli Pang
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China
| | - Yuan Zhao
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China
| | - Yanpan Gao
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, CAMS & PUMC, Beijing, 100005, China
| | - Wei Liu
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, CAMS & PUMC, Beijing, 100005, China
| | - Pengcheng Shu
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, CAMS & PUMC, Beijing, 100005, China
| | - Wei Ge
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, CAMS & PUMC, Beijing, 100005, China
| | - Zhanlong He
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China.
| | - Xiaozhong Peng
- State Key Laboratory of Respiratory Health and Multimorbidity, National Center of Technology Innovation for Animal Model, National Human Diseases Animal Model Resource Center, NHC Key Laboratory of Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Sciences, CAMS & PUMC, Beijing, 100021, China.
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China.
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, CAMS & PUMC, Beijing, 100005, China.
| |
Collapse
|
4
|
Hu Y, Lu Y, Fang Y, Zhang Q, Zheng Z, Zheng X, Ye X, Chen Y, Ding J, Yang J. Role of long non-coding RNA in inflammatory bowel disease. Front Immunol 2024; 15:1406538. [PMID: 38895124 PMCID: PMC11183289 DOI: 10.3389/fimmu.2024.1406538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/13/2024] [Indexed: 06/21/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a group of recurrent chronic inflammatory diseases, including Crohn's disease (CD) and ulcerative colitis (UC). Although IBD has been extensively studied for decades, its cause and pathogenesis remain unclear. Existing research suggests that IBD may be the result of an interaction between genetic factors, environmental factors and the gut microbiome. IBD is closely related to non-coding RNAs (ncRNAs). NcRNAs are composed of microRNA(miRNA), long non-coding RNA(lnc RNA) and circular RNA(circ RNA). Compared with miRNA, the role of lnc RNA in IBD has been little studied. Lnc RNA is an RNA molecule that regulates gene expression and regulates a variety of molecular pathways involved in the pathbiology of IBD. Targeting IBD-associated lnc RNAs may promote personalized treatment of IBD and have therapeutic value for IBD patients. Therefore, this review summarized the effects of lnc RNA on the intestinal epithelial barrier, inflammatory response and immune homeostasis in IBD, and summarized the potential of lnc RNA as a biomarker of IBD and as a predictor of therapeutic response to IBD in the future.
Collapse
Affiliation(s)
- Yufei Hu
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Yifan Lu
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Yi Fang
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Qizhe Zhang
- Department of Geriatrics, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Zhuoqun Zheng
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Xiaojuan Zheng
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Xiaohua Ye
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Yanping Chen
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Jin Ding
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Jianfeng Yang
- Department of Gastroenterology, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Miltefosine as a PPM1A activator improves AD-like pathology in mice by alleviating tauopathy via microglia/neurons crosstalk. Brain Behav Immun Health 2022; 26:100546. [DOI: 10.1016/j.bbih.2022.100546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/10/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022] Open
|
6
|
Rozenberg JM, Kamynina M, Sorokin M, Zolotovskaia M, Koroleva E, Kremenchutckaya K, Gudkov A, Buzdin A, Borisov N. The Role of the Metabolism of Zinc and Manganese Ions in Human Cancerogenesis. Biomedicines 2022; 10:biomedicines10051072. [PMID: 35625809 PMCID: PMC9139143 DOI: 10.3390/biomedicines10051072] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 04/30/2022] [Accepted: 05/03/2022] [Indexed: 12/14/2022] Open
Abstract
Metal ion homeostasis is fundamental for life. Specifically, transition metals iron, manganese and zinc play a pivotal role in mitochondrial metabolism and energy generation, anti-oxidation defense, transcriptional regulation and the immune response. The misregulation of expression or mutations in ion carriers and the corresponding changes in Mn2+ and Zn2+ levels suggest that these ions play a pivotal role in cancer progression. Moreover, coordinated changes in Mn2+ and Zn2+ ion carriers have been detected, suggesting that particular mechanisms influenced by both ions might be required for the growth of cancer cells, metastasis and immune evasion. Here, we present a review of zinc and manganese pathophysiology suggesting that these ions might cooperatively regulate cancerogenesis. Zn and Mn effects converge on mitochondria-induced apoptosis, transcriptional regulation and the cGAS-STING signaling pathway, mediating the immune response. Both Zn and Mn influence cancer progression and impact treatment efficacy in animal models and clinical trials. We predict that novel strategies targeting the regulation of both Zn and Mn in cancer will complement current therapeutic strategies.
Collapse
Affiliation(s)
- Julian Markovich Rozenberg
- Moscow Institute of Physics and Technology, National Research University, 141700 Moscow, Russia; (M.S.); (M.Z.); (E.K.); (K.K.); (A.B.); (N.B.)
- Correspondence:
| | - Margarita Kamynina
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.K.); (A.G.)
| | - Maksim Sorokin
- Moscow Institute of Physics and Technology, National Research University, 141700 Moscow, Russia; (M.S.); (M.Z.); (E.K.); (K.K.); (A.B.); (N.B.)
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.K.); (A.G.)
| | - Marianna Zolotovskaia
- Moscow Institute of Physics and Technology, National Research University, 141700 Moscow, Russia; (M.S.); (M.Z.); (E.K.); (K.K.); (A.B.); (N.B.)
- OmicsWay Corporation, Walnut, CA 91789, USA
| | - Elena Koroleva
- Moscow Institute of Physics and Technology, National Research University, 141700 Moscow, Russia; (M.S.); (M.Z.); (E.K.); (K.K.); (A.B.); (N.B.)
| | - Kristina Kremenchutckaya
- Moscow Institute of Physics and Technology, National Research University, 141700 Moscow, Russia; (M.S.); (M.Z.); (E.K.); (K.K.); (A.B.); (N.B.)
| | - Alexander Gudkov
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.K.); (A.G.)
| | - Anton Buzdin
- Moscow Institute of Physics and Technology, National Research University, 141700 Moscow, Russia; (M.S.); (M.Z.); (E.K.); (K.K.); (A.B.); (N.B.)
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.K.); (A.G.)
- OmicsWay Corporation, Walnut, CA 91789, USA
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Oncobox Ltd., 121205 Moscow, Russia
| | - Nicolas Borisov
- Moscow Institute of Physics and Technology, National Research University, 141700 Moscow, Russia; (M.S.); (M.Z.); (E.K.); (K.K.); (A.B.); (N.B.)
- OmicsWay Corporation, Walnut, CA 91789, USA
| |
Collapse
|
7
|
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
|
8
|
Banerjee S, Maity S, Guchhait R, Chatterjee A, Biswas C, Adhikari M, Pramanick K. Toxic effects of cyanotoxins in teleost fish: A comprehensive review. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 240:105971. [PMID: 34560410 DOI: 10.1016/j.aquatox.2021.105971] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 09/03/2021] [Accepted: 09/13/2021] [Indexed: 06/13/2023]
Abstract
The phenomenon of eutrophication leads to the global occurrence of algal blooms. Cyanotoxins as produced by many cyanobacterial species can lead to detrimental effects to the biome due to their stability and potential biomagnification along food webs. Therefore, understanding of the potential risks these toxins pose to the most susceptible organisms is an important prerequisite for ecological risks assessment of cyanobacteria blooms. Fishes are an important component of aquatic ecosystems that are prone to direct exposure to cyanotoxins. However, relatively few investigations have focused on measuring the toxic potentials of cyanotoxins in teleost fishes. This review comprehensively describes the major toxicological impacts (such as hepatotoxicity, neurotoxicity, immune toxicity, reproductive toxicity and cytogenotoxicity) of commonly occurring cyanotoxins in teleost fishes. The present work encompasses recent research progresses with special emphasis on the basic molecular mechanisms by which different cyanotoxins impose their toxicities in teleost fishes. The major research areas, which need to be focused on in future scientific investigations, have also been highlighted. Protein kinase inhibition, transcriptional dysregulation, disruption of redox homeostasis and the induction of apoptotic pathways appear to be the key drivers of the toxicological effects of cyanotoxins in fish. Analyses also showed that the impacts of cyanotoxins on specific reproductive processes are relatively less described in teleosts in comparison to mammalian systems. In fact, as compared to other toxicological effects of cyanotoxins, their reproductive toxicity (such as impacts on oocyte development, maturation and their hormonal regulation) is poorly understood in fish, and thus requires further studies. Furthermore, additonal studies characterizing the molecular mechanisms responsible for the cellular uptake of cyanotoxins need to be investigated.
Collapse
Affiliation(s)
- Sambuddha Banerjee
- Integrative Biology Research Unit, Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata 700073, India
| | - Sukhendu Maity
- Integrative Biology Research Unit, Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata 700073, India
| | - Rajkumar Guchhait
- P.G. Department of Zoology, Mahishadal Raj College, Garkamalpur, Purba Medinipur, India
| | - Ankit Chatterjee
- Integrative Biology Research Unit, Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata 700073, India
| | - Chayan Biswas
- Integrative Biology Research Unit, Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata 700073, India
| | - Madhuchhanda Adhikari
- Integrative Biology Research Unit, Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata 700073, India
| | - Kousik Pramanick
- Integrative Biology Research Unit, Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata 700073, India.
| |
Collapse
|
9
|
Neumann J, Boknik P, Kirchhefer U, Gergs U. The role of PP5 and PP2C in cardiac health and disease. Cell Signal 2021; 85:110035. [PMID: 33964402 DOI: 10.1016/j.cellsig.2021.110035] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/16/2021] [Accepted: 05/03/2021] [Indexed: 02/08/2023]
Abstract
Protein phosphatases are important, for example, as functional antagonists of β-adrenergic stimulation of the mammalian heart. While β-adrenergic stimulations increase the phosphorylation state of regulatory proteins and therefore force of contraction in the heart, these phosphorylations are reversed and thus force is reduced by the activity of protein phosphatases. In this context the role of PP5 and PP2C is starting to unravel. They do not belong to the same family of phosphatases with regard to sequence homology, many similarities with regard to location, activation by lipids and putative substrates have been worked out over the years. We also suggest which pathways for regulation of PP5 and/or PP2C described in other tissues and not yet in the heart might be useful to look for in cardiac tissue. Both phosphatases might play a role in signal transduction of sarcolemmal receptors in the heart. Expression of PP5 and PP2C can be increased by extracellular stimuli in the heart. Because PP5 is overexpressed in failing animal and human hearts, and because overexpression of PP5 or PP2C leads to cardiac hypertrophy and KO of PP5 leads to cardiac hypotrophy, one might argue for a role of PP5 and PP2C in heart failure. Because PP5 and PP2C can reduce, at least in vitro, the phosphorylation state of proteins thought to be relevant for cardiac arrhythmias, a role of these phosphatases for cardiac arrhythmias is also probable. Thus, PP5 and PP2C might be druggable targets to treat important cardiac diseases like heart failure, cardiac hypertrophy and cardiac arrhythmias.
Collapse
Affiliation(s)
- Joachim Neumann
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Magdeburger Str. 4, D-06097 Halle, Germany.
| | - Peter Boknik
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Westfälische Wilhelms-Universität, Domagkstraße 12, D-48149 Münster, Germany.
| | - Uwe Kirchhefer
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Westfälische Wilhelms-Universität, Domagkstraße 12, D-48149 Münster, Germany.
| | - Ulrich Gergs
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Magdeburger Str. 4, D-06097 Halle, Germany.
| |
Collapse
|
10
|
Seumen CHT, Grimm TM, Hauck CR. Protein phosphatases in TLR signaling. Cell Commun Signal 2021; 19:45. [PMID: 33882943 PMCID: PMC8058998 DOI: 10.1186/s12964-021-00722-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptors (TLRs) are critical sensors for the detection of potentially harmful microbes. They are instrumental in initiating innate and adaptive immune responses against pathogenic organisms. However, exaggerated activation of TLR receptor signaling can also be responsible for the onset of autoimmune and inflammatory diseases. While positive regulators of TLR signaling, such as protein serine/threonine kinases, have been studied intensively, only little is known about phosphatases, which counterbalance and limit TLR signaling. In this review, we summarize protein phosphorylation events and their roles in the TLR pathway and highlight the involvement of protein phosphatases as negative regulators at specific steps along the TLR-initiated signaling cascade. Then, we focus on individual phosphatase families, specify the function of individual enzymes in TLR signaling in more detail and give perspectives for future research. A better understanding of phosphatase-mediated regulation of TLR signaling could provide novel access points to mitigate excessive immune activation and to modulate innate immune signaling.![]() Video Abstract
Collapse
Affiliation(s)
- Clovis H T Seumen
- Lehrstuhl Zellbiologie, Universität Konstanz, Universitätsstraße 10, Postablage 621, 78457, Konstanz, Germany
| | - Tanja M Grimm
- Lehrstuhl Zellbiologie, Universität Konstanz, Universitätsstraße 10, Postablage 621, 78457, Konstanz, Germany.,Konstanz Research School Chemical Biology, Universität Konstanz, 78457, Konstanz, Germany
| | - Christof R Hauck
- Lehrstuhl Zellbiologie, Universität Konstanz, Universitätsstraße 10, Postablage 621, 78457, Konstanz, Germany. .,Konstanz Research School Chemical Biology, Universität Konstanz, 78457, Konstanz, Germany.
| |
Collapse
|
11
|
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: 2.8] [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
|
12
|
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: 4.0] [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
|
13
|
Narla G, Sangodkar J, Ryder CB. The impact of phosphatases on proliferative and survival signaling in cancer. Cell Mol Life Sci 2018; 75:2695-2718. [PMID: 29725697 PMCID: PMC6023766 DOI: 10.1007/s00018-018-2826-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/24/2018] [Accepted: 04/23/2018] [Indexed: 02/06/2023]
Abstract
The dynamic and stringent coordination of kinase and phosphatase activity controls a myriad of physiologic processes. Aberrations that disrupt the balance of this interplay represent the basis of numerous diseases. For a variety of reasons, early work in this area portrayed kinases as the dominant actors in these signaling events with phosphatases playing a secondary role. In oncology, these efforts led to breakthroughs that have dramatically altered the course of certain diseases and directed vast resources toward the development of additional kinase-targeted therapies. Yet, more recent scientific efforts have demonstrated a prominent and sometimes driving role for phosphatases across numerous malignancies. This maturation of the phosphatase field has brought with it the promise of further therapeutic advances in the field of oncology. In this review, we discuss the role of phosphatases in the regulation of cellular proliferation and survival signaling using the examples of the MAPK and PI3K/AKT pathways, c-Myc and the apoptosis machinery. Emphasis is placed on instances where these signaling networks are perturbed by dysregulation of specific phosphatases to favor growth and persistence of human cancer.
Collapse
Affiliation(s)
| | - Jaya Sangodkar
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | |
Collapse
|
14
|
Chen J, Wan J, Ye J, Xia L, Lu N. Emerging role of lncRNAs in the normal and diseased intestinal barrier. Inflamm Res 2018; 67:757-764. [PMID: 30008030 DOI: 10.1007/s00011-018-1170-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/08/2018] [Accepted: 07/10/2018] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE A significant effort has been made to understand the intestinal barrier, but the effective means to prevent, reduce, and restore intestinal mucosal damage remains unclear. Recently, a few of studies have explained the mechanism of the intestinal barrier in long noncoding RNAs (lncRNAs). This review aims to summarize recent views on the function of lncRNAs in the intestinal barrier and discuss the emerging role of lncRNAs in intestinal barrier diseases caused by inflammatory diseases. METHODS Observations led us to believe that lncRNAs participate in inflammatory responses, cell proliferation, and control microbial susceptibility. In view of these, lncRNAs have been proved to involve in the intestinal barrier. RESULTS lncRNAs directly or indirectly affect TJ mRNA translation and intestinal epithelial cells (IECs) paracellular permeability, as well as IECs proliferation and susceptibility to apoptosis, to modulate the function of the intestinal barrier. miRNAs play a pivotal role in this process. CONCLUSIONS lncRNAs have been shown to be fundamentally involved in intestinal mucosal regeneration, protection, and epithelial barrier function. It may emerge as new and potential factors to be evaluated in the intestinal barrier diseases caused by acute pancreatitis, inflammatory bowel diseases, and imbalance of intestinal flora.
Collapse
Affiliation(s)
- Jie Chen
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Jianhua Wan
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Jianfang Ye
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Liang Xia
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China.
| | - Nonghua Lu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| |
Collapse
|
15
|
Zhu Y, Ni T, Deng W, Lin J, Zheng L, Zhang C, Luo M. Effects of NLRP6 on the proliferation and activation of human hepatic stellate cells. Exp Cell Res 2018; 370:383-388. [PMID: 29966662 DOI: 10.1016/j.yexcr.2018.06.040] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/27/2018] [Accepted: 06/29/2018] [Indexed: 12/29/2022]
Abstract
Nod-like receptor pyrin domain-containing proteins (NLRPs) are known to take part in the pathogenesis of chronic liver diseases, including liver fibrosis. However, no known direct role of NLRP6, a member of NLRPs, has been reported in liver fibrosis. Here, we found that NLRP6 expression was decreased in fibrotic and cirrhotic livers. In a human hepatic stellate cell line, LX-2, overexpression of NLRP6 suppressed cell proliferation, hydroxyproline accumulation, as well as the expression of type I and type III collagens (Col-I and Col-III), α-smooth muscle actin (α-SMA) and matrix metalloproteinases (MMP2 and MMP9), whereas NLRP6 knockdown displayed reverse effects. Furthermore, NLRP6 significantly suppressed the phosphorylation of Smad2/3 (p-Smad2/3) and enhanced the expression of protein phosphatase magnesium dependent 1 A (PPM1A), the only phosphatase for Smad2/3. NLRP6 overexpression abrogated TGF-β1-stimulated hydroxyproline accumulation and p-Smad2/3. Co-immunoprecipitation assay demonstrated that NLRP6 was able to form a complex with PPM1A. NLRP6 overexpression did not change the level of p-Smad2/3 in LX-2 cells with PPM1A knockdown. These data indicated that PPM1A was required for the inhibitory effects of NLRP6 on TGF-β1/Smad2/3 signaling. In conclusion, our results suggest that NLRP6 exerts anti-fibrotic effects in LX-2 cells via regulating PPM1A/Smad2/3 and that NLRP6 may be an effective target in the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Yiming Zhu
- Department of General Surgery, Shanghai Ninth People' Hospital, School of Medicine, Shanghai Jiao Tong University, Huangpu, Shanghai, China
| | - Tao Ni
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People' Hospital, School of Medicine, Shanghai Jiao Tong University, Huangpu, Shanghai, China
| | - Wensheng Deng
- Department of General Surgery, Shanghai Ninth People' Hospital, School of Medicine, Shanghai Jiao Tong University, Huangpu, Shanghai, China
| | - Jiayun Lin
- Department of General Surgery, Shanghai Ninth People' Hospital, School of Medicine, Shanghai Jiao Tong University, Huangpu, Shanghai, China
| | - Lei Zheng
- Department of General Surgery, Shanghai Ninth People' Hospital, School of Medicine, Shanghai Jiao Tong University, Huangpu, Shanghai, China
| | - Chihao Zhang
- Department of General Surgery, Shanghai Ninth People' Hospital, School of Medicine, Shanghai Jiao Tong University, Huangpu, Shanghai, China
| | - Meng Luo
- Department of General Surgery, Shanghai Ninth People' Hospital, School of Medicine, Shanghai Jiao Tong University, Huangpu, Shanghai, China.
| |
Collapse
|
16
|
Zhang Y, Tao R, Wu SS, Xu CC, Wang JL, Chen J, Yu YS, Tang ZH, Chen XH, Zang GQ. TRIM52 up-regulation in hepatocellular carcinoma cells promotes proliferation, migration and invasion through the ubiquitination of PPM1A. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:116. [PMID: 29898761 PMCID: PMC6001170 DOI: 10.1186/s13046-018-0780-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/21/2018] [Indexed: 02/07/2023]
Abstract
Background Many tripartite motif (TRIM) family proteins have been reported to be of great importance in the initiation and progression in hepatocellular carcinoma (HCC). However, the biological role and regulatory mechanism of tripartite motif containing 52 (TRIM52) in HCC development and progression are poorly defined. Methods Immunohistochemistry (IHC), quantitative real-time PCR (qRT-PCR) or Western blot analysis was used to detect TRIM52, p21, matrix metalloproteinase 2 (MMP2), protein phosphatase, Mg2+/Mn2+ dependent 1A (PPM1A), p-Smad2/3 and Smad2/3 levels in HCC tissues and cell lines. HCC cell proliferation and cell cycle were measured by Cell Counting Kit-8 (CCK-8) and flow cytometry analysis, respectively. HCC cell migration and invasion were measured by Transwell assay. Tumor growth of HCC cells in vivo was measured using the nude mouse xenograft model. The correlation between TRIM52 and PPM1A was measured by co-immunoprecipitation (Co-IP) and ubiquitination analysis in vitro. Results TRIM52 was significantly up-regulated in the HCC tissues in comparison with the adjacent non-tumor hepatic tissues. TRIM52 was also up-regulated in HCC cell lines (MHCC-97H and MHCC-97L cells) compared with normal human liver cell line LO2. TRIM52 down-regulation by RNA interfering in MHCC-97H cells enhanced inhibition of cell proliferation, migration and invasion. TRIM52 down-regulation also induced MHCC-97H cells arrest in G0-G1 phase cell cycle and inhibited MHCC-97H cell growth in the nude mice. However, TRIM52 up-regulation in MHCC-97L cells promoted cell proliferation, migration and invasion. Furthermore, TRIM52 down-regulation significantly increased p21 and PPM1A expression, but inhibited MMP2 expression and induced Smad2/3 dephosphorylation in MHCC-97H cells, which were reversed by TRIM52 up-regulation in MHCC-97L cells. TRIM52 was found interacted with PPM1A and TRIM52 down-regulation inhibited the ubiquitination of PPM1A. Importantly, PPM1A up-regulation in MHCC-97L cells significantly suppressed TRIM52-mediated enhancement on cell proliferation, invasion and migration. Conclusions Our findings suggest that TRIM52 up-regulation promotes proliferation, migration and invasion of HCC cells through the ubiquitination of PPM1A.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China
| | - Ran Tao
- Department of Cardiology, Central Hospital of Minhang District, Shanghai, 201199, China
| | - Shan-Shan Wu
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China
| | - Cui-Cui Xu
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China
| | - Jie-Ling Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, China
| | - Jie Chen
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China
| | - Yong-Sheng Yu
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China
| | - Zheng-Hao Tang
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China
| | - Xiao-Hua Chen
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| | - Guo-Qing Zang
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
17
|
Debnath S, Kosek D, Tagad HD, Durell SR, Appella DH, Acevedo R, Grishaev A, Dyda F, Appella E, Mazur SJ. A trapped human PPM1A-phosphopeptide complex reveals structural features critical for regulation of PPM protein phosphatase activity. J Biol Chem 2018; 293:7993-8008. [PMID: 29602904 DOI: 10.1074/jbc.ra117.001213] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/29/2018] [Indexed: 01/09/2023] Open
Abstract
Metal-dependent protein phosphatases (PPM) are evolutionarily unrelated to other serine/threonine protein phosphatases and are characterized by their requirement for supplementation with millimolar concentrations of Mg2+ or Mn2+ ions for activity in vitro The crystal structure of human PPM1A (also known as PP2Cα), the first PPM structure determined, displays two tightly bound Mn2+ ions in the active site and a small subdomain, termed the Flap, located adjacent to the active site. Some recent crystal structures of bacterial or plant PPM phosphatases have disclosed two tightly bound metal ions and an additional third metal ion in the active site. Here, the crystal structure of the catalytic domain of human PPM1A, PPM1Acat, complexed with a cyclic phosphopeptide, c(MpSIpYVA), a cyclized variant of the activation loop of p38 MAPK (a physiological substrate of PPM1A), revealed three metal ions in the active site. The PPM1Acat D146E-c(MpSIpYVA) complex confirmed the presence of the anticipated third metal ion in the active site of metazoan PPM phosphatases. Biophysical and computational methods suggested that complex formation results in a slightly more compact solution conformation through reduced conformational flexibility of the Flap subdomain. We also observed that the position of the substrate in the active site allows solvent access to the labile third metal-binding site. Enzyme kinetics of PPM1Acat toward a phosphopeptide substrate supported a random-order, bi-substrate mechanism, with substantial interaction between the bound substrate and the labile metal ion. This work illuminates the structural and thermodynamic basis of an innate mechanism regulating the activity of PPM phosphatases.
Collapse
Affiliation(s)
- Subrata Debnath
- Laboratory of Cell Biology, Center for Cancer Research, NCI, Bethesda, Maryland 20892
| | - Dalibor Kosek
- Laboratories of Molecular Biology, Bethesda, Maryland 20892
| | - Harichandra D Tagad
- Laboratory of Cell Biology, Center for Cancer Research, NCI, Bethesda, Maryland 20892
| | - Stewart R Durell
- Laboratory of Cell Biology, Center for Cancer Research, NCI, Bethesda, Maryland 20892
| | - Daniel H Appella
- Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Roderico Acevedo
- Institute for Bioscience and Biotechnology Research, Rockville, Maryland 20850
| | - Alexander Grishaev
- Institute for Bioscience and Biotechnology Research, Rockville, Maryland 20850; National Institute of Standards and Technology, Gaithersburg, Maryland 20899
| | - Fred Dyda
- Laboratories of Molecular Biology, Bethesda, Maryland 20892
| | - Ettore Appella
- Laboratory of Cell Biology, Center for Cancer Research, NCI, Bethesda, Maryland 20892
| | - Sharlyn J Mazur
- Laboratory of Cell Biology, Center for Cancer Research, NCI, Bethesda, Maryland 20892.
| |
Collapse
|
18
|
Meeusen B, Janssens V. Tumor suppressive protein phosphatases in human cancer: Emerging targets for therapeutic intervention and tumor stratification. Int J Biochem Cell Biol 2017; 96:98-134. [PMID: 29031806 DOI: 10.1016/j.biocel.2017.10.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 10/04/2017] [Accepted: 10/05/2017] [Indexed: 02/06/2023]
Abstract
Aberrant protein phosphorylation is one of the hallmarks of cancer cells, and in many cases a prerequisite to sustain tumor development and progression. Like protein kinases, protein phosphatases are key regulators of cell signaling. However, their contribution to aberrant signaling in cancer cells is overall less well appreciated, and therefore, their clinical potential remains largely unexploited. In this review, we provide an overview of tumor suppressive protein phosphatases in human cancer. Along their mechanisms of inactivation in defined cancer contexts, we give an overview of their functional roles in diverse signaling pathways that contribute to their tumor suppressive abilities. Finally, we discuss their emerging roles as predictive or prognostic markers, their potential as synthetic lethality targets, and the current feasibility of their reactivation with pharmacologic compounds as promising new cancer therapies. We conclude that their inclusion in clinical practice has obvious potential to significantly improve therapeutic outcome in various ways, and should now definitely be pushed forward.
Collapse
Affiliation(s)
- Bob Meeusen
- Laboratory of Protein Phosphorylation & Proteomics, Dept. of Cellular & Molecular Medicine, Faculty of Medicine, KU Leuven & Leuven Cancer Institute (LKI), KU Leuven, Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation & Proteomics, Dept. of Cellular & Molecular Medicine, Faculty of Medicine, KU Leuven & Leuven Cancer Institute (LKI), KU Leuven, Belgium.
| |
Collapse
|
19
|
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: 1.1] [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
|
20
|
Zhou Y, Zhao Y, Gao Y, Hu W, Qu Y, Lou N, Zhu Y, Zhang X, Yang H. Hepatitis C virus NS3 protein enhances hepatocellular carcinoma cell invasion by promoting PPM1A ubiquitination and degradation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:42. [PMID: 28283039 PMCID: PMC5345236 DOI: 10.1186/s13046-017-0510-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/23/2017] [Indexed: 01/03/2023]
Abstract
Background Growing evidence suggests that hepatitis C virus (HCV) contributes to hepatocellular carcinoma (HCC) by directly modulating oncogenic signaling pathways. Protein phosphatase magnesium-dependent 1A (PPM1A) has recently emerged as an important tumor suppressor as it can block a range of tumor-centric signaling pathways through protein dephosphorylation. However, the role and regulatory mechanisms of PPM1A in HCV-infected cells have not been reported. Methods Total, cytoplasmic, and nuclear PPM1A protein after HCV infection or overexpression of HCV nonstructural protein 3 (NS3) were detected by western blotting. The expression of PPM1A in normal liver and HCV-related HCC tissues was quantified by immunohistochemistry. The effects of HCV infection and NS3 expression on the PPM1A protein level were systematically analyzed, and the ubiquitination level of PPM1A was determined by precipitation with anti-PPM1A and immunoblotting with either anti-ubiquitin or anti-PPM1A antibody. Finally, the roles of NS3 and PPM1A in hepatoma cell migration and invasion were assessed by wound healing and transwell assays, respectively. Results HCV infection and replication decreased PPM1A abundance, mediated by NS3, in hepatoma cells. Compared to normal liver tissues, the expression of PPM1A was significantly decreased in the HCC tumor tissues and adjacent non-tumor tissues. NS3 directly interacted with PPM1A to promote PPM1A ubiquitination and degradation, which was dependent on its protease domain. Blockade of PPM1A through small interfering RNA significantly promoted HCC cell migration, invasion, and epithelial mesenchymal transition (EMT), which were further intensified by TGF-β1 stimulation, in vitro. Furthermore, restoration of PPM1A abrogated the NS3-mediated promotion of HCC migration and invasion to a great extent, which was dependent on its protein phosphatase function. Conclusions Our findings demonstrate that the HCV protein NS3 can downregulate PPM1A by promoting its ubiquitination and proteasomal degradation, which might contribute to the migration and invasion of hepatoma cells and may represent a new strategy of HCV in carcinogenesis. Electronic supplementary material The online version of this article (doi:10.1186/s13046-017-0510-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yali Zhou
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yan Zhao
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yaoying Gao
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Wenjun Hu
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yan Qu
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Ning Lou
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei Province, China
| | - Ying Zhu
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei Province, China
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei Province, China.
| | - Hongmei Yang
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
21
|
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; 55:151-8. [DOI: 10.1016/j.humpath.2016.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [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]
|
22
|
Ulcerative Colitis-Associated Long Noncoding RNA, BC012900, Regulates Intestinal Epithelial Cell Apoptosis. Inflamm Bowel Dis 2016; 22:782-95. [PMID: 26937624 DOI: 10.1097/mib.0000000000000691] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) were recently found to be key regulators of biological functions and associated with human diseases. Thus far, the roles of lncRNAs in inflammatory bowel disease (IBD) remain unknown. We aimed to determine whether lncRNAs are associated with IBD and regulate epithelial cell physiology. METHODS lncRNAs microarray and quantitative RT-PCR were performed on 60 sigmoid colon biopsies from patients with active ulcerative colitis (UC) and relevant controls. Localization of lncRNAs was detected by in situ hybridization and on subcellular RNA. The boundaries of BC012900 were assessed by 5' and 3'-rapid amplification of cDNA ends. Apoptosis and proliferation assays were performed on BC012900-expressing construct or siRNA-transfected cells. RESULTS We identified 329 lncRNAs with increased and 126 lncRNAs with decreased expression in active UC tissues compared with normal control tissues, including the most significantly upregulated (BC012900, AK001903, and AK023330) and downregulated (BC029135, CDKN2B-AS1, and BC062296) transcripts. We found that most of the lncRNAs are localized to the nucleus. In particular, BC012900 expression was significantly increased in active UC and stimulated by cytokines and pathogenic molecules. Furthermore, BC012900 overexpression in epithelial cells results in a significant inhibition of cell proliferation and an increased susceptibility to apoptosis, which differ from its adjacent gene DUSP4. CONCLUSIONS Multiple lncRNAs are differentially expressed in IBD and play a role in regulating cellular physiology. Our results indicate that lncRNAs may be integral modulators of intestinal inflammation associated with IBD and represent novel targets for future therapeutics and diagnostic marker development.
Collapse
|
23
|
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: 2.2] [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
|
24
|
Global analysis of serine/threonine and tyrosine protein phosphatase catalytic subunit genes in Neurospora crassa reveals interplay between phosphatases and the p38 mitogen-activated protein kinase. G3-GENES GENOMES GENETICS 2014; 4:349-65. [PMID: 24347630 PMCID: PMC3931568 DOI: 10.1534/g3.113.008813] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Protein phosphatases are integral components of the cellular signaling machinery in eukaryotes, regulating diverse aspects of growth and development. The genome of the filamentous fungus and model organism Neurospora crassa encodes catalytic subunits for 30 protein phosphatase genes. In this study, we have characterized 24 viable N. crassa phosphatase catalytic subunit knockout mutants for phenotypes during growth, asexual development, and sexual development. We found that 91% of the mutants had defects in at least one of these traits, whereas 29% possessed phenotypes in all three. Chemical sensitivity screens were conducted to reveal additional phenotypes for the mutants. This resulted in the identification of at least one chemical sensitivity phenotype for 17 phosphatase knockout mutants, including novel chemical sensitivities for two phosphatase mutants lacking a growth or developmental phenotype. Hence, chemical sensitivity or growth/developmental phenotype was observed for all 24 viable mutants. We investigated p38 mitogen-activated protein kinase (MAPK) phosphorylation profiles in the phosphatase mutants and identified nine potential candidates for regulators of the p38 MAPK. We demonstrated that the PP2C class phosphatase pph-8 (NCU04600) is an important regulator of female sexual development in N. crassa. In addition, we showed that the Δcsp-6 (ΔNCU08380) mutant exhibits a phenotype similar to the previously identified conidial separation mutants, Δcsp-1 and Δcsp-2, that lack transcription factors important for regulation of conidiation and the circadian clock.
Collapse
|
25
|
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: 4.9] [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
|
26
|
Wang ZQ, Keita M, Bachvarova M, Gobeil S, Morin C, Plante M, Gregoire J, Renaud MC, Sebastianelli A, Trinh XB, Bachvarov D. Inhibition of RUNX2 transcriptional activity blocks the proliferation, migration and invasion of epithelial ovarian carcinoma cells. PLoS One 2013; 8:e74384. [PMID: 24124450 PMCID: PMC3790792 DOI: 10.1371/journal.pone.0074384] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 07/31/2013] [Indexed: 01/19/2023] Open
Abstract
Previously, we have identified the RUNX2 gene as hypomethylated and overexpressed in post-chemotherapy (CT) primary cultures derived from serous epithelial ovarian cancer (EOC) patients, when compared to primary cultures derived from matched primary (prior to CT) tumors. However, we found no differences in the RUNX2 methylation in primary EOC tumors and EOC omental metastases, suggesting that DNA methylation-based epigenetic mechanisms have no impact on RUNX2 expression in advanced (metastatic) stage of the disease. Moreover, RUNX2 displayed significantly higher expression not only in metastatic tissue, but also in high-grade primary tumors and even in low malignant potential tumors. Knockdown of the RUNX2 expression in EOC cells led to a sharp decrease of cell proliferation and significantly inhibited EOC cell migration and invasion. Gene expression profiling and consecutive network and pathway analyses confirmed these findings, as various genes and pathways known previously to be implicated in ovarian tumorigenesis, including EOC tumor invasion and metastasis, were found to be downregulated upon RUNX2 suppression, while a number of pro-apoptotic genes and some EOC tumor suppressor genes were induced. Taken together, our data are indicative for a strong oncogenic potential of the RUNX2 gene in serous EOC progression and suggest that RUNX2 might be a novel EOC therapeutic target. Further studies are needed to more completely elucidate the functional implications of RUNX2 and other members of the RUNX gene family in ovarian tumorigenesis.
Collapse
Affiliation(s)
- Zhi-Qiang Wang
- Department of Molecular Medicine, Laval University, Québec (Québec), Canada
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec (Québec), Canada
| | - Mamadou Keita
- Department of Molecular Medicine, Laval University, Québec (Québec), Canada
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec (Québec), Canada
| | - Magdalena Bachvarova
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec (Québec), Canada
| | - Stephane Gobeil
- Department of Molecular Medicine, Laval University, Québec (Québec), Canada
- Centre de recherche du CHU de Québec, CHUL, Québec (Québec), Canada
| | - Chantale Morin
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec (Québec), Canada
| | - Marie Plante
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec (Québec), Canada
- Department of Obstetrics and Gynecology, Laval University, Québec (Québec), Canada
| | - Jean Gregoire
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec (Québec), Canada
- Department of Obstetrics and Gynecology, Laval University, Québec (Québec), Canada
| | - Marie-Claude Renaud
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec (Québec), Canada
- Department of Obstetrics and Gynecology, Laval University, Québec (Québec), Canada
| | - Alexandra Sebastianelli
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec (Québec), Canada
- Department of Obstetrics and Gynecology, Laval University, Québec (Québec), Canada
| | - Xuan Bich Trinh
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec (Québec), Canada
- Department of Gynecological Oncology, Antwerp University Hospital, Antwerp, Belgium
| | - Dimcho Bachvarov
- Department of Molecular Medicine, Laval University, Québec (Québec), Canada
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec (Québec), Canada
- * E-mail:
| |
Collapse
|
27
|
Tanoue K, Miller Jenkins LM, Durell SR, Debnath S, Sakai H, Tagad HD, Ishida K, Appella E, Mazur SJ. Binding of a third metal ion by the human phosphatases PP2Cα and Wip1 is required for phosphatase activity. Biochemistry 2013; 52:5830-43. [PMID: 23906386 DOI: 10.1021/bi4005649] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The PPM phosphatases require millimolar concentrations of Mg²⁺ or Mn²⁺ to activate phosphatase activity in vitro. The human phosphatases PP2Cα (PPM1A) and Wip1 (PPM1D) differ in their physiological function, substrate specificity, and apparent metal affinity. A crystallographic structure of PP2Cα shows only two metal ions in the active site. However, recent structural studies of several bacterial PP2C phosphatases have indicated three metal ions in the active site. Two residues that coordinate the third metal ion are highly conserved, suggesting that human PP2C phosphatases may also bind a third ion. Here, isothermal titration calorimetry analysis of Mg²⁺ binding to PP2Cα distinguished binding of two ions to high affinity sites from the binding of a third ion with a millimolar affinity, similar to the apparent metal affinity required for catalytic activity. Mutational analysis indicated that Asp239 and either Asp146 or Asp243 was required for low-affinity binding of Mg²⁺, but that both Asp146 and Asp239 were required for catalysis. Phosphatase activity assays in the presence of MgCl₂, MnCl₂, or mixtures of the two, demonstrate high phosphatase activity toward a phosphopeptide substrate when Mg²⁺ was bound to the low-affinity site, whether Mg²⁺ or Mn²⁺ ions were bound to the high affinity sites. Mutation of the corresponding putative third metal ion-coordinating residues of Wip1 affected catalytic activity similarly both in vitro and in human cells. These results suggest that phosphatase activity toward phosphopeptide substrates by PP2Cα and Wip1 requires the binding of a Mg²⁺ ion to the low-affinity site.
Collapse
Affiliation(s)
- Kan Tanoue
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
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: 3.1] [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
|
29
|
Liu Y, Maxwell S, Feng T, Zhu X, Elston RC, Koyutürk M, Chance MR. Gene, pathway and network frameworks to identify epistatic interactions of single nucleotide polymorphisms derived from GWAS data. BMC SYSTEMS BIOLOGY 2012; 6 Suppl 3:S15. [PMID: 23281810 PMCID: PMC3524014 DOI: 10.1186/1752-0509-6-s3-s15] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background Interactions among genomic loci (also known as epistasis) have been suggested as one of the potential sources of missing heritability in single locus analysis of genome-wide association studies (GWAS). The computational burden of searching for interactions is compounded by the extremely low threshold for identifying significant p-values due to multiple hypothesis testing corrections. Utilizing prior biological knowledge to restrict the set of candidate SNP pairs to be tested can alleviate this problem, but systematic studies that investigate the relative merits of integrating different biological frameworks and GWAS data have not been conducted. Results We developed four biologically based frameworks to identify pairwise interactions among candidate SNP pairs as follows: (1) for each human protein-coding gene, a set of SNPs associated with that gene was constructed providing a gene-based interaction model, (2) for each known biological pathway, a set of SNPs associated with the genes in the pathway was constructed providing a pathway-based interaction model, (3) a set of SNPs associated with genes in a disease-related subnetwork provides a network-based interaction model, and (4) a framework is based on the function of SNPs. The last approach uses expression SNPs (eSNPs or eQTLs), which are SNPs or loci that have defined effects on the abundance of transcripts of other genes. We constructed pairs of eSNPs and SNPs located in the target genes whose expression is regulated by eSNPs. For all four frameworks the SNP sets were exhaustively tested for pairwise interactions within the sets using a traditional logistic regression model after excluding genes that were previously identified to associate with the trait. Using previously published GWAS data for type 2 diabetes (T2D) and the biologically based pair-wise interaction modeling, we identify twelve genes not seen in the previous single locus analysis. Conclusion We present four approaches to detect interactions associated with complex diseases. The results show our approaches outperform the traditional single locus approaches in detecting genes that previously did not reach significance; the results also provide novel drug targets and biomarkers relevant to the underlying mechanisms of disease.
Collapse
Affiliation(s)
- Yu Liu
- Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, OH, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Zhang WJ, Zhang WG, Zhang PY, Cao XM, He AL, Chen YX, Gu LF. The expression and functional characterization associated with cell apoptosis and proteomic analysis of the novel gene MLAA-34 in U937 cells. Oncol Rep 2012; 29:491-506. [PMID: 23135622 DOI: 10.3892/or.2012.2129] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Accepted: 09/25/2012] [Indexed: 11/06/2022] Open
Abstract
MLAA-34 is a novel acute monocytic leukemia (M5)-associated antigen (MLAA) that plays a role in the apoptosis of U937 cells. However, the expression and molecular mechanism of MLAA-34 in U937 cells remain largely unclear. Here, we utilized three strategies to gain insight into the expression and molecular functions of MLAA-34 and to identify its interacting proteins and pathways involved in the fine-tuning of the MLAA-34 response. Western blot analysis was performed to assess the expression of MLAA-34 in 41 cell lines and five mixed cell types, which revealed that MLAA-34 is most strongly expressed in U937 cells. Immunostaining indicated that MLAA-34 is localized in the cytoplasm and cell membrane. Furthermore, lentivirus-mediated overexpression of MLAA-34 in the U937 cell line led to significant suppression of apoptosis and increased the potential of tumorigenicity. Co-immunoprecipitation (Co-IP), shotgun and bioinformatic analysis identified 256 proteins and 225 of them were annotated by gene ontology categories. This analysis revealed 71 proteins involved in cell apoptosis or proliferation of biological processes and signaling pathways. Moreover, the effect of MLAA-34 apoptosis may be through interaction with the Ras, Wnt, calcium and chemokine signaling pathways and thirteen of the annotated proteins may interact with MLAA-34 and participate in carcinogenesis directly. This study provides a basis for a better understanding of the molecular mechanism and proteomics in the inhibition of apoptosis by MLAA-34 in U937 cells and indicates that MLAA-34 may be a potential candidate for the early diagnosis and therapeutic application of M5.
Collapse
Affiliation(s)
- Wen-Juan Zhang
- Department of Clinical Hematology, Affiliated No. 2 Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710004, PR China
| | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
This review traces the historical origins and conceptual developments leading to the current state of knowledge of the three superfamilies of protein Ser/Thr phosphatases. 'PR enzyme' was identified as an enzyme that inactivates glycogen phosphorylase, although it took 10 years before this ugly duckling was recognized for its true identity as a protein Ser/Thr phosphatase. Ethanol denaturation for purification in the 1970s yielded a phosphatase that exhibited broad specificity, which was resolved into type-1 and type-2 phosphatases in the 1980s. More recent developments show that regulation and specificity are achieved through assembly of multisubunit holoenzymes, transient phosphorylation and the action of inhibitor proteins. Still not widely appreciated, there are hundreds of discrete protein Ser/Thr phosphatases available to counteract protein kinases, offering potential therapeutic targets. Signalling networks and modelling schemes need to incorporate the full gamut of protein Ser/Thr phosphatases and their interconnections.
Collapse
Affiliation(s)
- David L Brautigan
- Department of Microbiology, Immunology and Cancer Biology, Center for Cell Signaling, University of Virginia, School of Medicine, Charlottesville, VA 22908, USA.
| |
Collapse
|
32
|
Shohat M, Ben-Meir D, Lavi S. Protein phosphatase magnesium dependent 1A (PPM1A) plays a role in the differentiation and survival processes of nerve cells. PLoS One 2012; 7:e32438. [PMID: 22384250 PMCID: PMC3288098 DOI: 10.1371/journal.pone.0032438] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 01/27/2012] [Indexed: 12/22/2022] Open
Abstract
The serine/threonine phosphatase type 2C (PPM1A) has a broad range of substrates, and its role in regulating stress response is well established. We have investigated the involvement of PPM1A in the survival and differentiation processes of PC6-3 cells, a subclone of the PC12 cell line. This cell line can differentiate into neuron like cells upon exposure to nerve growth factor (NGF). Overexpression of PPM1A in naive PC6-3 cells caused cell cycle arrest at the G2/M phase followed by apoptosis. Interestingly, PPM1A overexpression did not affect fully differentiated cells. Using PPM1A overexpressing cells and PPM1A knockdown cells, we show that this phosphatase affects NGF signaling in PC6-3 cells and is engaged in neurite outgrowth. In addition, the ablation of PPM1A interferes with NGF-induced growth arrest during differentiation of PC6-3 cells.
Collapse
Affiliation(s)
| | | | - Sara Lavi
- Department of Cell Research and Immunology, Tel Aviv University, Tel Aviv, Israel
- * E-mail:
| |
Collapse
|
33
|
Activation of protein serine/threonine phosphatase PP2Cα efficiently prevents liver fibrosis. PLoS One 2010; 5:e14230. [PMID: 21151953 PMCID: PMC2997772 DOI: 10.1371/journal.pone.0014230] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Accepted: 11/15/2010] [Indexed: 01/22/2023] Open
Abstract
Background Over-activation of TGFβ signaling pathway and uncontrolled cell proliferation of hepatic stellate cells (HSCs) play pivotal roles in liver fibrogenesis, while the protein serine/threonine phosphatase PP2Cα was reported to negatively regulate TGFβ signaling pathway and cell cycle. Our study aimed to investigate the role of PP2Cα in liver fibrogenesis. Methodology/Principal Findings The effects of PP2Cα activation on liver fibrosis were investigated in human HSCs and primary rat HSCs in vitro using western blotting, real-time PCR, nuclear translocation, cell viability and cell cycle analyses. The antifibrogenic effects in carbon tetrachloride (CCl4)- and bile duct ligation (BDL)-induced mice in vivo were assessed using biochemical, histological and immunohistochemical analyses. The results demonstrated that activation of PP2Cα by overexpression or the new discovered small molecular activator NPLC0393 terminated TGFβ-Smad3 and TGFβ-p38 signaling pathways, induced cell cycle arrest in HSCs and decreased α-smooth muscle actin (α-SMA) expression, collagen deposition and hepatic hydroxyproline (HYP) level in CCl4- and BDL-induced mice. Conclusions/Significance Our findings suggested that PP2Cα activation might be an attractive new strategy for treating liver fibrosis while the small molecular activator NPLC0393 might represent a lead compound for antifibrogenic drug development. Moreover, our study might provide the first evidence for the role of PP2C family members in the fibrotic disease.
Collapse
|
34
|
p53-mediated apoptosis requires inositol hexakisphosphate kinase-2. Proc Natl Acad Sci U S A 2010; 107:20947-51. [PMID: 21078964 DOI: 10.1073/pnas.1015671107] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Inositol pyrophosphates have been implicated in numerous biological processes. Inositol hexakisphosphate kinase-2 (IP6K2), which generates the inositol pyrophosphate, diphosphoinositol pentakisphosphate (IP7), influences apoptotic cell death. The tumor suppressor p53 responds to genotoxic stress by engaging a transcriptional program leading to cell-cycle arrest or apoptosis. We demonstrate that IP6K2 is required for p53-mediated apoptosis and modulates the outcome of the p53 response. Gene disruption of IP6K2 in colorectal cancer cells selectively impairs p53-mediated apoptosis, instead favoring cell-cycle arrest. IP6K2 acts by binding directly to p53 and decreasing expression of proarrest gene targets such as the cyclin-dependent kinase inhibitor p21.
Collapse
|
35
|
Abstract
Type 2C Ser/Thr phosphatases are a remarkable class of protein phosphatases, which are conserved in eukaryotes and involved in a large variety of functional processes. Unlike in other Ser/Thr phosphatases, the catalytic polypeptide is not usually associated with regulatory subunits, and functional specificity is achieved by encoding multiple isoforms. For fungi, most information comes from the study of type 2C protein phosphatase (PP2C) enzymes in Saccharomyces cerevisiae, where seven PP2C-encoding genes (PTC1 to -7) with diverse functions can be found. More recently, data on several Candida albicans PP2C proteins became available, suggesting that some of them can be involved in virulence. In this work we review the available literature on fungal PP2Cs and explore sequence databases to provide a comprehensive overview of these enzymes in fungi.
Collapse
|
36
|
Wang M, Wang D, Lin L, Hong H. Protein profiles in zebrafish (Danio rerio) brains exposed to chronic microcystin-LR. CHEMOSPHERE 2010; 81:716-724. [PMID: 20800265 DOI: 10.1016/j.chemosphere.2010.07.061] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 07/20/2010] [Accepted: 07/28/2010] [Indexed: 05/29/2023]
Abstract
Microcystin-LR (MCLR) is a commonly encountered blue-green algal hepatotoxin and a known inhibitor of cellular protein phosphatase (PP), however, little is known about its neurotoxicity. This study investigated the protein profiles of zebrafish (Danio rerio) brains chronically exposed to MCLR concentrations (2 or 20 μg L(-1)) using the proteomic approach. The results showed that MCLR strikingly enhanced toxin accumulation and the PP activity in zebrafish brains after 30 d exposure. Comparison of two-dimensional electrophoresis protein profiles of MCLR exposed and non-exposed zebrafish brains revealed that the abundance of 30 protein spots was remarkably altered in response to MCLR exposure. These proteins are involved in cytoskeleton assembly, macromolecule metabolism, oxidative stress, signal transduction, and other functions (e.g. transporting, protein degradation, apoptosis and translation), indicating that MCLR toxicity in the fish brain is complex and diverse. The chronic neurotoxicity of MCLR might initiate the PP pathway via an upregulation of PP2C in the zebrafish brain, in addition to the reactive oxygen species pathway. Additionally, the increase of vitellogenin abundance in MCLR exposed zebrafish brains suggested that MCLR might mimic the effects of endocrine disrupting chemicals. This study demonstrated that MCLR causes neurotoxicity in zebrafish at the proteomic level, which provides a new insight into MCLR toxicity in aquatic organisms and human beings.
Collapse
Affiliation(s)
- Minghua Wang
- State Key Laboratory of Marine Environmental Science/Environmental Science Research Center, Xiamen University, Xiamen 361005, People's Republic of China
| | | | | | | |
Collapse
|
37
|
Aburai N, Yoshida M, Ohnishi M, Kimura K. Pisiferdiol and pisiferic acid isolated from Chamaecyparis pisifera activate protein phosphatase 2C in vitro and induce caspase-3/7-dependent apoptosis via dephosphorylation of Bad in HL60 cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2010; 17:782-788. [PMID: 20153620 DOI: 10.1016/j.phymed.2009.12.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Revised: 11/25/2009] [Accepted: 12/17/2009] [Indexed: 05/28/2023]
Abstract
Protein phosphatase 2C (PP2C) dephosphorylates a broad range of substrates and regulates apoptosis, stress response and growth-related pathways. In the course of screening for PP2C activators from natural sources, we isolated abietane-type diterpenes, pisiferdiol and pisiferic acid from Chamaecyparis pisifera. Pisiferdiol having a unique seven-membered ring showed more specific PP2C activation activity (1.3-fold at 100 microM) than pisiferic acid having a normal six-membered ring and oleic acid, which is known to activate PP2C. Pisiferdiol and pisiferic acid showed mixed-type activation with respect to alpha-casein, and this differed from the non-competitive activation of oleic acid in vitro. In vivo, the cytotoxicity of pisiferdiol toward human promyelocytic leukemia cell line HL60 with an IC(50) value of 18.3 microM was 2-fold and 7-fold stronger than those of pisiferic acid and oleic acid, and pisiferdiol induced apoptosis through a caspase 3/7-dependent mechanism involving the dephosphorylation of Bad(1), which is a PP2C substrate. We thus conclude that pisiferdiol and pisiferic acid are novel PP2C activators, and the more specific activator, pisiferdiol, may be a useful chemical probe to study PP2C-mediated signaling pathways, and a lead compound for pharmaceutical agents.
Collapse
Affiliation(s)
- N Aburai
- Laboratory of Chemical Biology, The United Graduate School of Agricultural Sciences, Iwate University, Morioka, Iwate 020-8550, Japan
| | | | | | | |
Collapse
|
38
|
Faherty CS, Merrell DS, Semino-Mora C, Dubois A, Ramaswamy AV, Maurelli AT. Microarray analysis of Shigella flexneri-infected epithelial cells identifies host factors important for apoptosis inhibition. BMC Genomics 2010; 11:272. [PMID: 20429941 PMCID: PMC2996966 DOI: 10.1186/1471-2164-11-272] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Accepted: 04/29/2010] [Indexed: 01/05/2023] Open
Abstract
Background Shigella flexneri inhibits apoptosis in infected epithelial cells. In order to understand the pro-survival effects induced by the bacteria, we utilized apoptosis-specific microarrays to analyze the changes in eukaryotic gene expression in both infected and uninfected cells in the presence and absence of staurosporine, a chemical inducer of the intrinsic pathway of apoptosis. The goal of this research was to identify host factors that contribute to apoptosis inhibition in infected cells. Results The microarray analysis revealed distinct expression profiles in uninfected and infected cells, and these changes were altered in the presence of staurosporine. These profiles allowed us to make comparisons between the treatment groups. Compared to uninfected cells, Shigella-infected epithelial cells, both in the presence and absence of staurosporine, showed significant induced expression of JUN, several members of the inhibitor of apoptosis gene family, nuclear factor κB and related genes, genes involving tumor protein 53 and the retinoblastoma protein, and surprisingly, genes important for the inhibition of the extrinsic pathway of apoptosis. We confirmed the microarray results for a selection of genes using in situ hybridization analysis. Conclusion Infection of epithelial cells with S. flexneri induces a pro-survival state in the cell that results in apoptosis inhibition in the presence and absence of staurosporine. The bacteria may target these host factors directly while some induced genes may represent downstream effects due to the presence of the bacteria. Our results indicate that the bacteria block apoptosis at multiple checkpoints along both pathways so that even if a cell fails to prevent apoptosis at an early step, Shigella will block apoptosis at the level of caspase-3. Apoptosis inhibition is most likely vital to the survival of the bacteria in vivo. Future characterization of these host factors is required to fully understand how S. flexneri inhibits apoptosis in epithelial cells.
Collapse
Affiliation(s)
- Christina S Faherty
- Department of Microbiology and Immunology, F, Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | | | | | | | | | | |
Collapse
|
39
|
Rajkumar R, Konishi K, Richards TJ, Ishizawar DC, Wiechert AC, Kaminski N, Ahmad F. Genomewide RNA expression profiling in lung identifies distinct signatures in idiopathic pulmonary arterial hypertension and secondary pulmonary hypertension. Am J Physiol Heart Circ Physiol 2010; 298:H1235-48. [PMID: 20081107 DOI: 10.1152/ajpheart.00254.2009] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Idiopathic pulmonary arterial hypertension (PAH) is a life-threatening condition characterized by pulmonary arteriolar remodeling. This investigation aimed to identify genes involved specifically in the pathogenesis of PAH and not other forms of pulmonary hypertension (PH). Using genomewide microarray analysis, we generated the largest data set to date of RNA expression profiles from lung tissue specimens from 1) 18 PAH subjects and 2) 8 subjects with PH secondary to idiopathic pulmonary fibrosis (IPF) and 3) 13 normal subjects. A molecular signature of 4,734 genes discriminated among these three cohorts. We identified significant novel biological changes that were likely to contribute to the pathogenesis of PAH, including regulation of actin-based motility, protein ubiquitination, and cAMP, transforming growth factor-beta, MAPK, estrogen receptor, nitric oxide, and PDGF signaling. Bone morphogenic protein receptor type II expression was downregulated, even in subjects without a mutation in this gene. Women with PAH had higher expression levels of estrogen receptor 1 than normal women. Real-time quantitative PCR confirmed differential expression of the following genes in PAH relative to both normal controls and PH secondary to IPF: a disintegrin-like and metalloprotease with thrombospondin type 1 motif 9, cell adhesion molecule with homology to L1CAM, cytochrome b(558) and beta-polypeptide, coagulation factor II receptor-like 3, A-myb myeloblastosis viral oncogene homolog 1, nuclear receptor coactivator 2, purinergic receptor P2Y, platelet factor 4, phospholamban, and tropomodulin 3. This study shows that PAH and PH secondary to IPF are characterized by distinct gene expression signatures, implying distinct pathophysiological mechanisms.
Collapse
Affiliation(s)
- Revathi Rajkumar
- Cardiovascular Institute, Univ. of Pittsburgh, PA 15213-2582, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Jan G, Delorme V, Saksouk N, Abrivard M, Gonzalez V, Cayla X, Hakimi MA, Tardieux I. A Toxoplasma type 2C serine-threonine phosphatase is involved in parasite growth in the mammalian host cell. Microbes Infect 2009; 11:935-45. [PMID: 19563907 DOI: 10.1016/j.micinf.2009.06.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2009] [Revised: 06/09/2009] [Accepted: 06/16/2009] [Indexed: 01/03/2023]
Abstract
Toxoplasma gondii is a human protozoan parasite that belongs to the phylum of Apicomplexa and causes toxoplasmosis. As the other members of this phylum, T. gondii obligatory multiplies within a host cell by a peculiar type of mitosis that leads to daughter cell assembly within a mother cell. Although parasite growth and virulence have been linked for years, few molecules controlling mitosis have been yet identified and they include a couple of kinases but not the counteracting phosphatases. Here, we report that in contrast to other animal cells, type 2C is by far the major type of serine threonine phosphatase activity both in extracellular and in intracellular dividing parasites. Using wild type and transgenic parasites, we characterized the 37kDa TgPP2C molecule as an abundant cytoplasmic and nuclear enzyme with activity being under tight regulation. In addition, we showed that the increase in TgPP2C activity significantly affected parasite growth by impairing cytokinesis while nuclear division still occurred. This study supports for the first time that type 2C protein phosphatase is an important regulator of cell growth in T. gondii.
Collapse
Affiliation(s)
- Gaelle Jan
- Institut Cochin, Université Paris Descartes, CNRS UMR 8104, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Krieglstein J, Hufnagel B, Dworak M, Schwarz S, Kewitz T, Reinbold M, Klumpp S. Influence of various fatty acids on the activity of protein phosphatase type 2C and apoptosis of endothelial cells and macrophages. Eur J Pharm Sci 2008; 35:397-403. [DOI: 10.1016/j.ejps.2008.08.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Revised: 08/20/2008] [Accepted: 08/20/2008] [Indexed: 10/21/2022]
|
42
|
Abstract
Multigenes and multigene interaction are involved in the development, recurrence and metastasis of hepatocellular carcinoma, while aberrant gene expression is the significant cause of recurrence and metastasis. Researches on gene changes and gene interaction in hepatitis, liver cirrhosis and hepatocellular carcinoma are important for identifying the development, elucidating the pathogenesis, and guiding the prognosis and therapy of hepatocellular carcinoma.
Collapse
|
43
|
Stany MP, Bonome T, Wamunyokoli F, Zorn K, Ozbun L, Park DC, Hao K, Boyd J, Sood AK, Gershenson DM, Berkowitz RS, Mok SC, Birrer MJ. Classification of ovarian cancer: a genomic analysis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 622:23-33. [PMID: 18546616 DOI: 10.1007/978-0-387-68969-2_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
44
|
Yan M, Song Y, Wong CP, Hardin K, Ho E. Zinc deficiency alters DNA damage response genes in normal human prostate epithelial cells. J Nutr 2008; 138:667-73. [PMID: 18356318 PMCID: PMC4152237 DOI: 10.1093/jn/138.4.667] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Zinc is an essential trace element for human health and is a critical component of many proteins and transcription factors involved in DNA damage response and repair. The prostate is known to accumulate high levels of zinc, but levels are markedly decreased with cancer development. We hypothesized that zinc plays a critical role in maintaining DNA integrity in the prostate and zinc deficiency would lead to increased DNA damage and altered DNA damage response mechanisms. To test this hypothesis, the goal of this study was to determine the effects of zinc deficiency on DNA damage and DNA repair mechanisms by examining changes in global gene expression and transcription factor binding abilities in normal prostate epithelial cells (PrEC). Increased single-strand DNA breaks (Comet assay) were observed in PrEC grown in zinc-deficient media compared with cells grown in zinc-adequate media for 7 d. Using Affymetrix HG-U133A gene chips, differential expression of genes involved in cell cycle, apoptosis, transcription, and DNA damage response and repair were identified with low cellular zinc. Among genes involved in DNA damage response and repair, tumor protein p73, MRE11 meiotic recombination 11 homolog A, X-ray repair complementing defective repair in Chinese hamster cells 4, and breast cancer 2, early onset were down-regulated and TP53 was up-regulated. Additionally, western blotting showed increased nuclear p53 protein expression with zinc deficiency. Despite increased p53 gene and nuclear protein expression, there was no significant change in p53 binding activity. Zinc deficiency also induced an increase in binding activity of transcription factors involved in regulating cell proliferation and apoptosis. Thus, zinc deficiency may compromise DNA integrity in the prostate by impairing the function of zinc-containing proteins.
Collapse
Affiliation(s)
| | | | | | | | - Emily Ho
- To whom correspondence should be addressed.
| |
Collapse
|
45
|
Lammers T, Lavi S. Role of type 2C protein phosphatases in growth regulation and in cellular stress signaling. Crit Rev Biochem Mol Biol 2008; 42:437-61. [PMID: 18066953 DOI: 10.1080/10409230701693342] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A number of interesting features, phenotypes, and potential clinical applications have recently been ascribed to the type 2C family of protein phosphatases. Thus far, 16 different PP2C genes have been identified in the human genome, encoding (by means of alternative splicing) for at least 22 different isozymes. Virtually ever since their discovery, type 2C phosphatases have been predominantly linked to cell growth and to cellular stress signaling. Here, we provide an overview of the involvement of type 2C phosphatases in these two processes, and we show that four of them (PP2Calpha, PP2Cbeta, ILKAP, and PHLPP) can be expected to function as tumor suppressor proteins, and one as an oncoprotein (PP2Cdelta /Wip1). In addition, we demonstrate that in virtually all cases in which they have been linked to the stress response, PP2Cs act as inhibitors of cellular stress signaling. Based on the vast amount of experimental evidence obtained thus far, it therefore seems justified to conclude that type 2C protein phosphatases are important physiological regulators of cell growth and of cellular stress signaling.
Collapse
Affiliation(s)
- Twan Lammers
- Department of Innovative Cancer Diagnosis and Therapy, German Cancer Research Center, Heidelberg, Germany.
| | | |
Collapse
|
46
|
Lammers T, Peschke P, Ehemann V, Debus J, Slobodin B, Lavi S, Huber P. Role of PP2Calpha in cell growth, in radio- and chemosensitivity, and in tumorigenicity. Mol Cancer 2007; 6:65. [PMID: 17941990 PMCID: PMC2100065 DOI: 10.1186/1476-4598-6-65] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2007] [Accepted: 10/17/2007] [Indexed: 12/19/2022] Open
Abstract
Background PP2Cα is the representative member of the type 2C family of protein phosphatases, and it has recently been implicated in the regulation of p53-, TGFβ-, cyclin-dependent kinase- and apoptosis-signaling. To investigate the role of PP2Cα in cell growth and in radio- and chemosensitivity, wild type and PP2Cα siRNA-expressing MCF7 cells were subjected to several different viability and cell cycle analyses, both under basal conditions and upon treatment with radio- and chemotherapy. By comparing the growth of tumors established from both types of cells, we also evaluated the involvement of PP2Cα in tumorigenesis. Results It was found that knockdown of PP2Cα did not affect the proliferation, the clonogenic survival and the membrane integrity of MCF7 cells. In addition, it did not alter their radio- and chemosensitivity. For PP2Cα siRNA-expressing MCF7 cells, the number of cells in the G0/G1 phase of the cell cycle was reduced, the induction of the G1 block was attenuated, the number of cells in G2/M was increased, and the induction of the G2 block was enhanced. The tumorigenic potential of PP2Cα siRNA-expressing MCF7 cells was found to be higher than that of wild type MCF7 cells, and the in vivo proliferation of these cells was found to be increased. Conclusion Based on these findings, we conclude that PP2Cα is not involved in controlling cell growth and radio- and chemosensitivity in vitro. It does, however, play a role in the regulation of the cell cycle, in the induction of cell cycle checkpoints and in tumorigenesis. The latter notion implies that PP2Cα may possess tumor-suppressing properties, and it thereby sets the stage for more elaborate analyses on its involvement in the development and progression of cancer.
Collapse
Affiliation(s)
- Twan Lammers
- Department of Innovative Cancer Diagnosis and Therapy, Clinical Cooperation Unit Radiotherapeutic Oncology, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | | | | | | | | | | | | |
Collapse
|
47
|
Lu X, Ma O, Nguyen TA, Jones SN, Oren M, Donehower LA. The Wip1 Phosphatase acts as a gatekeeper in the p53-Mdm2 autoregulatory loop. Cancer Cell 2007; 12:342-54. [PMID: 17936559 DOI: 10.1016/j.ccr.2007.08.033] [Citation(s) in RCA: 215] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2006] [Revised: 07/23/2007] [Accepted: 08/29/2007] [Indexed: 12/11/2022]
Abstract
The tumor suppressor p53 is a transcription factor that responds to cellular stresses by initiating cell cycle arrest or apoptosis. One transcriptional target of p53 is Mdm2, an E3 ubiquitin ligase that interacts with p53 to promote its proteasomal degradation in a negative feedback regulatory loop. Here we show that the wild-type p53-induced phosphatase 1 (Wip1), or PPM1D, downregulates p53 protein levels by stabilizing Mdm2 and facilitating its access to p53. Wip1 interacts with and dephosphorylates Mdm2 at serine 395, a site phosphorylated by the ATM kinase. Dephosphorylated Mdm2 has increased stability and affinity for p53, facilitating p53 ubiquitination and degradation. Thus, Wip1 acts as a gatekeeper in the Mdm2-p53 regulatory loop by stabilizing Mdm2 and promoting Mdm2-mediated proteolysis of p53.
Collapse
Affiliation(s)
- Xiongbin Lu
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Arden N, Majors BS, Ahn SH, Oyler G, Betenbaugh MJ. Inhibiting the apoptosis pathway using MDM2 in mammalian cell cultures. Biotechnol Bioeng 2007; 97:601-14. [PMID: 17149774 DOI: 10.1002/bit.21254] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The ability to regulate apoptosis in mammalian cell cultures represents one approach to developing more economical and efficient processes. Genetic modification of cells using anti-apoptotic genes is one method that may be used to improve cellular performance. This study investigates a method to inhibit upstream apoptosis pathways through the overexpression of MDM2, an E3 ubiquitin ligase for p53. Both 293 and CHO cells expressing MDM2 were examined under both batch and spent media conditions. For batch cultures, MDM2 overexpression increased viable cell densities and viabilities over control cells with the largest enhancements observed in CHO cells. When CHO cells were passaged without medium exchange, cells expressing MDM2 reached a viable cell density that was nearly double the control and survived for an extra day in culture. When exposed to spent media initially, both 293-MDM2 and CHO-MDM2 cells continued to grow for 2 days while the control cells stopped growing after the first day. DNA analysis using flow cytometry confirmed that while CHO controls were found to be undergoing DNA fragmentation, CHO-MDM2 cells exhibit DNA degradation at a much slower rate. When compared to Bcl-2-expressing cells, MDM2 expression showed greater protection against apoptosis in passaged culture, spent medium, and following transient p53 overexpression. However, expression of the RING sequence of MDM2 responsible for E3 ligase activity without the other components of the protein was found to be toxic to 293 cells in culture. These results suggest that the overexpression of heterologous MDM2 represents a promising method to delay apoptosis in mammalian cell cultures.
Collapse
Affiliation(s)
- Nilou Arden
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, USA
| | | | | | | | | |
Collapse
|
49
|
Stern A, Privman E, Rasis M, Lavi S, Pupko T. Evolution of the Metazoan Protein Phosphatase 2C Superfamily. J Mol Evol 2006; 64:61-70. [PMID: 17160364 DOI: 10.1007/s00239-006-0033-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2006] [Accepted: 10/04/2006] [Indexed: 01/22/2023]
Abstract
Members of the protein phosphatase 2C (PP2C) superfamily are Mg(2+)/Mn(2+)-dependent serine/threonine phosphatases, which are essential for regulation of cell cycle and stress signaling pathways in cells. In this study, a comprehensive genomic analysis of all available metazoan PP2C sequences was conducted. The phylogeny of PP2C was reconstructed, revealing the existence of 15 vertebrate families which arose following a series of gene duplication events. Relative dating of these duplications showed that they occurred in two active periods: before the divergence of bilaterians and before vertebrate diversification. PP2C families which duplicated during the first period take part in different signaling pathways, whereas PP2C families which diverged in the second period display tissue expression differences yet participate in similar signaling pathways. These differences were found to involve variation of expression in tissues which show higher complexity in vertebrates, such as skeletal muscle and the nervous system. Further analysis was performed with the aim of identifying the functional domains of PP2C. The conservation pattern across the entire PP2C superfamily revealed an extensive domain of more than 50 amino acids which is highly conserved throughout all PP2C members. Several insertion or deletion events were found which may have led to the specialization of each PP2C family.
Collapse
Affiliation(s)
- Adi Stern
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | | | | | |
Collapse
|
50
|
Klumpp S, Thissen MC, Krieglstein J. Protein phosphatases types 2Cα and 2Cβ in apoptosis. Biochem Soc Trans 2006; 34:1370-5. [PMID: 17073821 DOI: 10.1042/bst0341370] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This mini-review highlights the involvement of PP2C (protein phosphatase type 2C) family members α and β in apoptosis. The activity of these isoenzymes can be stimulated by unsaturated fatty acids with special structural features, e.g. oleic acid. Those fatty acids capable of activating PP2Cα and PP2Cβ in vitro induce apoptosis in various cell types as shown here for neurons and endothelial cells. Using RNA interference to reduce the amount of PP2Cα and PP2Cβ results in cells significantly less susceptible to the apoptotic effect of oleic acid. Increased endothelial cell death is considered to be an initial step of atherogenesis. Thus activation of PP2C by physiological unbound (‘free’) unsaturated fatty acids (liberated from lipoproteins) could represent a crucial mechanism in the development of atherosclerosis.
Collapse
Affiliation(s)
- S Klumpp
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität, Münster, Germany.
| | | | | |
Collapse
|