1
|
Farias JO, Pacheco DRDCG, Magalhaes YT, Russo LC, Boell VK, Hilares DJF, Forti FL. Knockdown of dual-specificity phosphatase 3 drives differentiation and polarization of myeloid leukemia cells into macrophages with reduced proliferative and DNA repair fitness. Tissue Cell 2025; 96:102947. [PMID: 40334397 DOI: 10.1016/j.tice.2025.102947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/08/2025] [Accepted: 04/28/2025] [Indexed: 05/09/2025]
Abstract
Dual-specificity phosphatase 3 (DUSP3) regulates key cellular processes, including the cell cycle, proliferation, and differentiation. Recently, we demonstrated its crucial role in maintaining genomic stability by interacting with and dephosphorylating nucleophosmin (NPM), thereby modulating nuclear p53 activity under genotoxic stress. Given the frequent mutations in both p53 and NPM in acute myeloid leukemia (AML), this study aimed to investigate the impact of DUSP3 knockdown in two p53-deficient AML cell lines and explore potential correlations with NPM expression. THP-1 cells exhibited higher basal levels of DUSP3 and NPM compared to HL-60 cells, while DUSP3 knockdown reduced NPM expression in HL-60 cells. Upon phorbol 12-myristate 13-acetate (PMA)-induced differentiation into macrophage-like cells, only HL-60 cells displayed decreased levels of both DUSP3 and NPM. DUSP3 knockdown enhanced differentiation in THP-1 and HL-60 cells and promoted non-classical M2 macrophage polarization following additional PMA exposure, as indicated by increased expression of CD11b and CD206. Bioinformatics analysis revealed significant correlations between DUSP3 and NPM gene expression, AML patient survival, and the maturation stage of myeloid cells. Furthermore, DUSP3 knockdown in undifferentiated HL-60 cells impaired proliferation and compromised genomic stability under genotoxic stress induced by doxorubicin. These findings suggest that DUSP3 plays a regulatory role in the differentiation, polarization, and proliferation of myeloid cells. Through the modulation of NPM expression and activity, DUSP3 may contribute to a deeper understanding of leukemia pathophysiology and mechanisms of chemotherapy resistance.
Collapse
Affiliation(s)
- Jessica O Farias
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Sao Paulo, SP, Brazil
| | - Diana R D C G Pacheco
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Sao Paulo, SP, Brazil
| | - Yuli T Magalhaes
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Sao Paulo, SP, Brazil
| | - Lilian C Russo
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Sao Paulo, SP, Brazil
| | - Viktor K Boell
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Sao Paulo, SP, Brazil
| | - Donna J F Hilares
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Sao Paulo, SP, Brazil
| | - Fabio L Forti
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
2
|
Patysheva MR, Prostakishina EA, Budnitskaya AA, Bragina OD, Kzhyshkowska JG. Dual-Specificity Phosphatases in Regulation of Tumor-Associated Macrophage Activity. Int J Mol Sci 2023; 24:17542. [PMID: 38139370 PMCID: PMC10743672 DOI: 10.3390/ijms242417542] [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: 11/22/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
The regulation of protein kinases by dephosphorylation is a key mechanism that defines the activity of immune cells. A balanced process of the phosphorylation/dephosphorylation of key protein kinases by dual-specificity phosphatases is required for the realization of the antitumor immune response. The family of dual-specificity phosphatases is represented by several isoforms found in both resting and activated macrophages. The main substrate of dual-specificity phosphatases are three components of mitogen-activated kinase signaling cascades: the extracellular signal-regulated kinase ERK1/2, p38, and Janus kinase family. The results of the study of model tumor-associated macrophages supported the assumption of the crucial role of dual-specificity phosphatases in the formation and determination of the outcome of the immune response against tumor cells through the selective suppression of mitogen-activated kinase signaling cascades. Since mitogen-activated kinases mostly activate the production of pro-inflammatory mediators and the antitumor function of macrophages, the excess activity of dual-specificity phosphatases suppresses the ability of tumor-associated macrophages to activate the antitumor immune response. Nowadays, the fundamental research in tumor immunology is focused on the search for novel molecular targets to activate the antitumor immune response. However, to date, dual-specificity phosphatases received limited discussion as key targets of the immune system to activate the antitumor immune response. This review discusses the importance of dual-specificity phosphatases as key regulators of the tumor-associated macrophage function.
Collapse
Affiliation(s)
- Marina R. Patysheva
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia; (M.R.P.); (E.A.P.); (A.A.B.)
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia;
| | - Elizaveta A. Prostakishina
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia; (M.R.P.); (E.A.P.); (A.A.B.)
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia;
| | - Arina A. Budnitskaya
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia; (M.R.P.); (E.A.P.); (A.A.B.)
- Laboratory of Genetic Technologies, Siberian State Medical University, 634050 Tomsk, Russia
| | - Olga D. Bragina
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia;
| | - Julia G. Kzhyshkowska
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia; (M.R.P.); (E.A.P.); (A.A.B.)
- Laboratory of Genetic Technologies, Siberian State Medical University, 634050 Tomsk, Russia
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Mannheim Institute of Innate Immunosciences (MI3), University of Heidelberg, 68167 Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg—Hessen, 69117 Mannheim, Germany
| |
Collapse
|
3
|
Sprenkle NT, Winn NC, Bunn KE, Zhao Y, Park DJ, Giese BG, Karijolich JJ, Ansel KM, Serezani CH, Hasty AH, Pua HH. The miR-23-27-24 clusters drive lipid-associated macrophage proliferation in obese adipose tissue. Cell Rep 2023; 42:112928. [PMID: 37542720 PMCID: PMC10712211 DOI: 10.1016/j.celrep.2023.112928] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/19/2023] [Accepted: 07/17/2023] [Indexed: 08/07/2023] Open
Abstract
Identifying molecular circuits that control adipose tissue macrophage (ATM) function is necessary to understand how ATMs contribute to tissue homeostasis and obesity-induced insulin resistance. In this study, we find that mice with a myeloid-specific knockout of the miR-23-27-24 clusters of microRNAs (miRNAs) gain less weight on a high-fat diet but exhibit worsened glucose and insulin tolerance. Analysis of ATMs from these mice shows selectively reduced numbers and proliferation of a recently reported subset of lipid-associated CD9+Trem2+ ATMs (lipid-associated macrophages [LAMs]). Leveraging the role of miRNAs to control networks of genes, we use RNA sequencing (RNA-seq), functional screens, and biochemical assays to identify candidate target transcripts that regulate proliferation-associated signaling. We determine that miR-23 directly targets the mRNA of Eif4ebp2, a gene that restricts protein synthesis and proliferation in macrophages. Altogether, our study demonstrates that control of proliferation of a protective subset of LAMs by noncoding RNAs contributes to protection against diet-induced obesity metabolic dysfunction.
Collapse
Affiliation(s)
- Neil T Sprenkle
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nathan C Winn
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Kaitlyn E Bunn
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yang Zhao
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Deborah J Park
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Brenna G Giese
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John J Karijolich
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Center for Immunobiology and Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanerbilt-Ingram Cancer Center, Nashville, TN, USA
| | - K Mark Ansel
- Department of Microbiology and Immunology and Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA, USA
| | - C Henrique Serezani
- Vanderbilt Center for Immunobiology and Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Immunobiology and Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA; Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Heather H Pua
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Center for Immunobiology and Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
4
|
Zhang Z, Chen Y, Zheng L, Du J, Wei S, Zhu X, Xiong JW. A DUSP6 inhibitor suppresses inflammatory cardiac remodeling and improves heart function after myocardial infarction. Dis Model Mech 2023; 16:285836. [PMID: 36478044 PMCID: PMC9789401 DOI: 10.1242/dmm.049662] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 11/13/2022] [Indexed: 12/12/2022] Open
Abstract
Acute myocardial infarction (MI) results in loss of cardiomyocytes and abnormal cardiac remodeling with severe inflammation and fibrosis. However, how cardiac repair can be achieved by timely resolution of inflammation and cardiac fibrosis remains incompletely understood. Our previous findings have shown that dual-specificity phosphatase 6 (DUSP6) is a regeneration repressor from zebrafish to rats. In this study, we found that intravenous administration of the DUSP6 inhibitor (E)-2-benzylidene-3-(cyclohexylamino)-2,3-dihydro-1H-inden-1-one (BCI) improved heart function and reduced cardiac fibrosis in MI rats. Mechanistic analysis revealed that BCI attenuated macrophage inflammation through NF-κB and p38 signaling, independent of DUSP6 inhibition, leading to the downregulation of various cytokines and chemokines. In addition, BCI suppressed differentiation-related signaling pathways and decreased bone-marrow cell differentiation into macrophages through inhibiting DUSP6. Furthermore, intramyocardial injection of poly (D, L-lactic-co-glycolic acid)-loaded BCI after MI had a notable effect on cardiac repair. In summary, BCI improves heart function and reduces abnormal cardiac remodeling by inhibiting macrophage formation and inflammation post-MI, thus providing a promising pro-drug candidate for the treatment of MI and related heart diseases. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Zongwang Zhang
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
- Peking University-Nanjing Institute of Translational Medicine, Nanjing 211800, China
| | - Yang Chen
- Peking University-Nanjing Institute of Translational Medicine, Nanjing 211800, China
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Lixia Zheng
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
- Peking University-Nanjing Institute of Translational Medicine, Nanjing 211800, China
| | - Jianyong Du
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
- Peking University-Nanjing Institute of Translational Medicine, Nanjing 211800, China
| | - Shicheng Wei
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Xiaojun Zhu
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
- Peking University-Nanjing Institute of Translational Medicine, Nanjing 211800, China
- Authors for correspondence (; )
| | - Jing-Wei Xiong
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
- Peking University-Nanjing Institute of Translational Medicine, Nanjing 211800, China
- Authors for correspondence (; )
| |
Collapse
|
5
|
Huang W, Zheng S, Xiao J, Liu C, Du T, Wu K. Parental exposure to bisphenol A affects pharyngeal cartilage development and causes global transcriptomic changes in zebrafish (Danio rerio) offspring. CHEMOSPHERE 2020; 249:126537. [PMID: 32208220 DOI: 10.1016/j.chemosphere.2020.126537] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/14/2020] [Accepted: 03/16/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND As one of the most common endocrine-disrupting chemicals (EDCs), bisphenol A (BPA) is a threat to aquatic ecosystems. Despite a rich literature addressing the adverse effects of BPA on various systems in fish models, the potential impact of parental BPA exposure on offspring pharyngeal cartilage development is poorly understood. METHODS Adult zebrafish (F0) were exposed to BPA (1.0 μM) or control for 7 days. Eggs (F1) were collected and exposed to BPA (control, 0.05, 0.1, 1, 10 μM) until 120 h post-fertilization. Histomorphometrical essay was used to quantitatively and qualitatively assess the effects of BPA on pharyngeal cartilage development. RNA sequencing (RNA-seq) was used to discover differentially expressed genes (DEGs), and KEGG pathway and GO enrichment analysis were performed to interpret functional ontology. RESULTS Parental BPA exposure affected hatchability and heart rates of F1 progeny. By pathology analysis, parental BPA exposure caused craniofacial deformity, characterized by wider angles of cartilage elements, disrupted pharyngeal chondrocytes and promoted apoptosis and elongation of head length. RNA-seq suggested that many DEGs were involved in multiple biological processes and signaling pathways; defense responses, reactive oxygen species metabolic process, apoptosis, p53 signaling pathway and MAPK signaling pathway were closely associated with the toxicity of parental BPA exposure. CONCLUSIONS Parental BPA exposure affected chondrogenesis in the viscerocranium of zebrafish offspring and led to global transcriptomic changes involved in apoptosis, hyperplasia and oxidative stress. These newly identified gene expression patterns, pathways and gene networks of zebrafish eleutheroembryos after early-life waterborne BPA exposure, may lead to severe and permanent morphological and functional consequences.
Collapse
Affiliation(s)
- Wenlong Huang
- Department of Preventive Medicine, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Shukai Zheng
- Department of Preventive Medicine, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Jiefeng Xiao
- Department of Preventive Medicine, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Caixia Liu
- Department of Preventive Medicine, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Taifeng Du
- Department of Preventive Medicine, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Kusheng Wu
- Department of Preventive Medicine, Shantou University Medical College, Shantou, 515041, Guangdong, China.
| |
Collapse
|
6
|
Lang R, Raffi FAM. Dual-Specificity Phosphatases in Immunity and Infection: An Update. Int J Mol Sci 2019; 20:ijms20112710. [PMID: 31159473 PMCID: PMC6600418 DOI: 10.3390/ijms20112710] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 05/28/2019] [Accepted: 05/30/2019] [Indexed: 12/26/2022] Open
Abstract
Kinase activation and phosphorylation cascades are key to initiate immune cell activation in response to recognition of antigen and sensing of microbial danger. However, for balanced and controlled immune responses, the intensity and duration of phospho-signaling has to be regulated. The dual-specificity phosphatase (DUSP) gene family has many members that are differentially expressed in resting and activated immune cells. Here, we review the progress made in the field of DUSP gene function in regulation of the immune system during the last decade. Studies in knockout mice have confirmed the essential functions of several DUSP-MAPK phosphatases (DUSP-MKP) in controlling inflammatory and anti-microbial immune responses and support the concept that individual DUSP-MKP shape and determine the outcome of innate immune responses due to context-dependent expression and selective inhibition of different mitogen-activated protein kinases (MAPK). In addition to the canonical DUSP-MKP, several small-size atypical DUSP proteins regulate immune cells and are therefore also reviewed here. Unexpected and complex findings in DUSP knockout mice pose new questions regarding cell type-specific and redundant functions. Another emerging question concerns the interaction of DUSP-MKP with non-MAPK binding partners and substrate proteins. Finally, the pharmacological targeting of DUSPs is desirable to modulate immune and inflammatory responses.
Collapse
Affiliation(s)
- Roland Lang
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Faizal A M Raffi
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany.
| |
Collapse
|
7
|
Smith SR, Schaaf K, Rajabalee N, Wagner F, Duverger A, Kutsch O, Sun J. The phosphatase PPM1A controls monocyte-to-macrophage differentiation. Sci Rep 2018; 8:902. [PMID: 29343725 PMCID: PMC5772551 DOI: 10.1038/s41598-017-18832-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/18/2017] [Indexed: 02/06/2023] Open
Abstract
Differentiation of circulating monocytes into tissue-bound or tissue-resident macrophages is a critical regulatory process affecting host defense and inflammation. However, the regulatory signaling pathways that control the differentiation of monocytes into specific and distinct functional macrophage subsets are poorly understood. Herein, we demonstrate that monocyte-to-macrophage differentiation is controlled by the Protein Phosphatase, Mg2+/Mn2+-dependent 1A (PPM1A). Genetic manipulation experiments demonstrated that overexpression of PPM1A attenuated the macrophage differentiation program, while knockdown of PPM1A expression accelerated the ability of monocytes to differentiate into macrophages. We identify imiquimod and Pam3CSK4 as two Toll-like receptor agonists that induce PPM1A expression, and show that increased expression of PPM1A at the onset of differentiation impairs cellular adherence, reduces expression of inflammatory (M1) macrophage-specific markers, and inhibits the production of inflammatory cytokines. Our findings reveal PPM1A as a negative threshold regulator of M1-type monocyte-to-macrophage differentiation, establishing it as a key phosphatase that orchestrates this program.
Collapse
Affiliation(s)
- Samuel R Smith
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kaitlyn Schaaf
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nusrah Rajabalee
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Frederic Wagner
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Alexandra Duverger
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Olaf Kutsch
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jim Sun
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
8
|
Haque N, Abdullah BJJ, Kasim NHA. Secretome: Pharmaceuticals for Cell-Free Regenerative Therapy. STEM CELL DRUGS - A NEW GENERATION OF BIOPHARMACEUTICALS 2018. [DOI: 10.1007/978-3-319-99328-7_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
9
|
Seo H, Cho YC, Ju A, Lee S, Park BC, Park SG, Kim JH, Kim K, Cho S. Dual-specificity phosphatase 5 acts as an anti-inflammatory regulator by inhibiting the ERK and NF-κB signaling pathways. Sci Rep 2017; 7:17348. [PMID: 29229953 PMCID: PMC5725455 DOI: 10.1038/s41598-017-17591-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 11/29/2017] [Indexed: 01/01/2023] Open
Abstract
Although dual-specificity phosphatase 5 (DUSP5), which inactivates extracellular signal-regulated kinase (ERK), suppresses tumors in several types of cancer, its functional roles remain largely unknown. Here, we show that DUSP5 is induced during lipopolysaccharide (LPS)-mediated inflammation and inhibits nuclear factor-κB (NF-κB) activity. DUSP5 mRNA and protein expression increased transiently in LPS-stimulated RAW 264.7 cells and then returned to basal levels. DUSP5 overexpression in RAW 264.7 cells suppressed the production of pro-inflammatory tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), whereas knockdown of DUSP5 increased their expression. Investigation of two major inflammatory signaling pathways, mitogen-activated protein kinase (MAPK) and NF-κB, using activator protein-1 (AP-1) and NF-κB reporter plasmids, respectively, showed that NF-κB transcription activity was downregulated by DUSP5 in a phosphatase activity-independent manner whereas AP-1 activity was inhibited by DUSP5 phosphatase activity towards ERK,. Further investigation showed that DUSP5 directly interacts with transforming growth factor beta-activated kinase 1 (TAK1) and inhibitor of κB (IκB) kinases (IKKs) but not with IκBα. DUSP5 binding to IKKs interfered with the association of TAK1 with IKKs, suggesting that DUSP5 might act as a competitive inhibitor of TAK1-IKKs association. Therefore, we propose that DUSP5 negatively regulates ERK and NF-κB in a phosphatase activity-dependent and -independent manner, respectively.
Collapse
Affiliation(s)
- Huiyun Seo
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Young-Chang Cho
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Anna Ju
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Sewoong Lee
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Byoung Chul Park
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Sung Goo Park
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Jeong-Hoon Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Kwonseop Kim
- College of Pharmacy and Research Institute for Drug Development, Chonnam National University, Gwang-ju, 61186, Republic of Korea
| | - Sayeon Cho
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
10
|
Wu M, Liu S, Hu L, Qu H, Pan C, Lei P, Shen Y, Yang M. Global transcriptomic analysis of zebrafish in response to embryonic exposure to three antidepressants, amitriptyline, fluoxetine and mianserin. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2017; 192:274-283. [PMID: 28992598 DOI: 10.1016/j.aquatox.2017.09.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 08/26/2017] [Accepted: 09/27/2017] [Indexed: 06/07/2023]
Abstract
Antidepressants are among the most commonly detected pharmaceuticals in aqueous systems, and, as emerging organic pollutants, may exert negative effects on non-target aquatic organisms. Previously, it has been revealed that antidepressant exposure significantly inhibits the growth and development of fish during their early developmental stages. Thus, in the present study, we aimed to identify and compare the underlying mechanisms of action of different antidepressants at the transcriptional level using zebrafish (Danio rerio) embryos. Through high-throughput RNA sequencing (RNA-Seq) data analysis, 32, 34, and 130 differentially expressed genes (DEGs) were obtained from zebrafish larvae after 120h of embryonic exposure to sublethal concentrations of amitriptyline, fluoxetine, and mianserin, respectively. The expression profiles of the identified DEGs showed similar trends in response to the three antidepressant treatments, suggesting consistent toxic effects of low concentrations of these three drugs on the regulation of gene expression in fish. Several metabolic and signaling pathways, including glycolysis/gluconeogenesis and the insulin pathway, were affected in the exposed fish larvae. The expression profiles of selected DEGs were then verified by the qRT-PCR method, which indicated significant positive correlations with the RNA-Seq results. Next, we determined the concentration-dependent expression patterns of 6 selected DEGs in fish larvae exposed to three antidepressants at a series of environmentally relevant concentrations. The results revealed a significant concentration-dependent reduction in the levels of dual-specificity phosphatase 5 (dusp5) mRNA, as well as a non-concentration-dependent gene expression inhibition of prostaglandin D2 synthase b (ptgdsb); the circadian rhythm-related genes, i.e. those encoding nuclear receptor subfamily 1, group D, member 1 (nr1d1) and period 2 (per2); and genes encoding early growth response factors (egr1 and egr4), in the antidepressant-treated fish larvae. In summary, to our knowledge, our findings demonstrate, for the first time, that the three different categories of antidepressants have common effects on the gene expression involved in multiple biological processes and signaling pathways during the early development of fish and thus provide information for characterizing the adverse outcome pathways and on the ecological risk assessment of these pharmaceutical pollutants in the aquatic environment.
Collapse
Affiliation(s)
- Minghong Wu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444 China.
| | - Shuai Liu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444 China; College of the Environment & Ecology, Xiamen University, Xiamen, Fujian 361005, China.
| | - Lei Hu
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Haidong Qu
- College of the Environment & Ecology, Xiamen University, Xiamen, Fujian 361005, China.
| | - Chenyuan Pan
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444 China.
| | - Penghui Lei
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Yingjia Shen
- College of the Environment & Ecology, Xiamen University, Xiamen, Fujian 361005, China.
| | - Ming Yang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444 China.
| |
Collapse
|
11
|
Zhang H, Qian PY, Ravasi T. Selective phosphorylation during early macrophage differentiation. Proteomics 2015; 15:3731-43. [PMID: 26307563 DOI: 10.1002/pmic.201400511] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 07/08/2015] [Accepted: 08/19/2015] [Indexed: 12/27/2022]
Abstract
The differentiation of macrophages from monocytes is a tightly controlled and complex biological process. Although numerous studies have been conducted using biochemical approaches or global gene/protein profiling, the mechanisms of the early stages of differentiation remain unclear. Here we used SILAC-based quantitative proteomics approach to perform temporal phosphoproteome profiling of early macrophage differentiation. We identified a large set of phosphoproteins and grouped them as PMA-regulated and non-regulated phosphoproteins in the early stages of differentiation. Further analysis of the PMA-regulated phosphoproteins revealed that transcriptional suppression, cytoskeletal reorganization and cell adhesion were among the most significantly activated pathways. Some key involved regulators of these pathways are mTOR, MYB, STAT1 and CTNNB. Moreover, we were able to classify the roles and activities of several transcriptional factors during different differentiation stages and found that E2F is likely to be an important regulator during the relatively late stages of differentiation. This study provides the first comprehensive picture of the dynamic phosphoproteome during myeloid cells differentiation, and identifies potential molecular targets in leukemic cells.
Collapse
Affiliation(s)
- Huoming Zhang
- Division of Biological and Environmental Sciences & Engineering, Division of Applied Mathematics and Computer Sciences, King Abdullah University of Science & Technology, Thuwal, Kingdom of Saudi Arabia.,Bioscience Core Laboratory, King Abdullah University of Science and Technology, Thuwal, Kingdom of Saudi Arabia
| | - Pei-Yuan Qian
- School of Science, Hong Kong University of Science and Technology, Hong Kong, P. R. China
| | - Timothy Ravasi
- Division of Biological and Environmental Sciences & Engineering, Division of Applied Mathematics and Computer Sciences, King Abdullah University of Science & Technology, Thuwal, Kingdom of Saudi Arabia
| |
Collapse
|
12
|
Carras S, Valayer A, Moratal C, Weiss-Gayet M, Pages G, Morlé F, Mouchiroud G, Gobert S. Instructive role of M-CSF on commitment of bipotent myeloid cells involves ERK-dependent positive and negative signaling. J Leukoc Biol 2015; 99:311-9. [PMID: 26336156 DOI: 10.1189/jlb.2a1214-619r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 08/17/2015] [Indexed: 12/20/2022] Open
Abstract
M-CSF and G-CSF are instructive cytokines that specifically induce differentiation of bipotent myeloid progenitors into macrophages and granulocytes, respectively. Through morphology and colony assay studies, flow cytometry analysis of specific markers, and expression of myeloid transcription factors, we show here that the Eger/Fms cell line is composed of cells whose differentiation fate is instructed by M-CSF and G-CSF, thus representing a good in vitro model of myeloid bipotent progenitors. Consistent with the essential role of ERK1/2 during macrophage differentiation and defects of macrophagic differentiation in native ERK1(-/-) progenitors, ERK signaling is strongly activated in Eger/Fms cells upon M-CSF-induced macrophagic differentiation but only to a very small extent during G-CSF-induced granulocytic differentiation. Previous in vivo studies indicated a key role of Fli-1 in myeloid differentiation and demonstrated its weak expression during macrophagic differentiation with a strong expression during granulocytic differentiation. Here, we demonstrated that this effect could be mediated by a differential regulation of protein kinase Cδ (PKCd) on Fli-1 expression in response to M-CSF and G-CSF. With the use of knockdown of PKCd by small interfering RNA, we demonstrated that M-CSF activates PKCd, which in turn, inhibits Fli-1 expression and granulocytic differentiation. Finally, we studied the connection between ERK and PKCd and showed that in the presence of the MEK inhibitor U0126, PKCd expression is decreased, and Fli-1 expression is increased in response to M-CSF. Altogether, we demonstrated that in bipotent myeloid cells, M-CSF promotes macrophagic over granulocytic differentiation by inducing ERK activation but also PKCd expression, which in turn, down-regulates Fli-1 expression and prevents granulocytic differentiation.
Collapse
Affiliation(s)
- Sylvain Carras
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique, Unités Mixtes de Recherche 5534, Université de Lyon, Villeurbanne Cedex, France
| | - Alexandre Valayer
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique, Unités Mixtes de Recherche 5534, Université de Lyon, Villeurbanne Cedex, France
| | - Claudine Moratal
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique, Unités Mixtes de Recherche 5534, Université de Lyon, Villeurbanne Cedex, France
| | - Michèle Weiss-Gayet
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique, Unités Mixtes de Recherche 5534, Université de Lyon, Villeurbanne Cedex, France
| | - Gilles Pages
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique, Unités Mixtes de Recherche 5534, Université de Lyon, Villeurbanne Cedex, France
| | - François Morlé
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique, Unités Mixtes de Recherche 5534, Université de Lyon, Villeurbanne Cedex, France
| | - Guy Mouchiroud
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique, Unités Mixtes de Recherche 5534, Université de Lyon, Villeurbanne Cedex, France
| | - Stéphanie Gobert
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique, Unités Mixtes de Recherche 5534, Université de Lyon, Villeurbanne Cedex, France
| |
Collapse
|
13
|
Hu N, Qiu Y, Dong F. Role of Erk1/2 signaling in the regulation of neutrophil versus monocyte development in response to G-CSF and M-CSF. J Biol Chem 2015; 290:24561-73. [PMID: 26296889 DOI: 10.1074/jbc.m115.668871] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Indexed: 01/11/2023] Open
Abstract
Lineage specification in the hematopoietic system depends on the expression of lineage specific transcription factors. However, the role of hematopoietic cytokines in this process has been controversial and little is known about the intracellular signaling mechanisms by which cytokines instruct lineage choice. G-CSF and M-CSF are two lineage-specific cytokines that play a dominant role in granulopoiesis and monopoiesis, respectively. We show here that a G-CSFR mutant in which tyrosine 729 had been mutated to phenylalanine (Y729F) promoted monocyte rather than neutrophil development in myeloid precursors, which was associated with prolonged activation of Erk1/2 and augmented activation of downstream targets c-Fos and Egr1. Inhibition of Erk1/2 activation or knockdown of c-Fos or Egr1 largely rescued neutrophil development in cells expressing G-CSFR Y729F. We also show that M-CSF, but not G-CSF, stimulated strong and sustained activation of Erk1/2 in mouse lineage marker negative (Lin(-)) bone marrow cells. Significantly, inhibition of Erk1/2 signaling in these cells favored neutrophil over monocyte development in response to M-CSF. Thus, prolonged Erk1/2 activation resulted in monocyte development following G-CSF induction whereas inhibition of Erk1/2 signaling promoted neutrophil development at the expense of monocyte formation in response to M-CSF. These results reveal an important mechanism by which G-CSF and M-CSF instruct neutrophil versus monocyte lineage choice, i.e. differential activation of Erk1/2 pathway.
Collapse
Affiliation(s)
- Nan Hu
- From the Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Yaling Qiu
- From the Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Fan Dong
- From the Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| |
Collapse
|
14
|
Holmes DA, Yeh JH, Yan D, Xu M, Chan AC. Dusp5 negatively regulates IL-33-mediated eosinophil survival and function. EMBO J 2014; 34:218-35. [PMID: 25398911 DOI: 10.15252/embj.201489456] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mitogen-activated protein kinase (MAPK) activation controls diverse cellular functions including cellular survival, proliferation, and apoptosis. Tuning of MAPK activation is counter-regulated by a family of dual-specificity phosphatases (DUSPs). IL-33 is a recently described cytokine that initiates Th2 immune responses through binding to a heterodimeric IL-33Rα (ST2L)/IL-1α accessory protein (IL-1RAcP) receptor that coordinates activation of ERK and NF-κB pathways. We demonstrate here that DUSP5 is expressed in eosinophils, is upregulated following IL-33 stimulation and regulates IL-33 signaling. Dusp5(-/-) mice have prolonged eosinophil survival and enhanced eosinophil effector functions following infection with the helminth Nippostrongylus brasiliensis. IL-33-activated Dusp5(-/-) eosinophils exhibit increased cellular ERK1/2 activation and BCL-XL expression that results in enhanced eosinophil survival. In addition, Dusp5(-/-) eosinophils demonstrate enhanced IL-33-mediated activation and effector functions. Together, these data support a role for DUSP5 as a novel negative regulator of IL-33-dependent eosinophil function and survival.
Collapse
Affiliation(s)
- Derek A Holmes
- Department of Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Jung-Hua Yeh
- Department of Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Donghong Yan
- Department of Translational Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Min Xu
- Department of Translational Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Andrew C Chan
- Department of Immunology, Genentech, Inc., South San Francisco, CA, USA
| |
Collapse
|
15
|
Abstract
The CSF-1 receptor (CSF-1R) is activated by the homodimeric growth factors colony-stimulating factor-1 (CSF-1) and interleukin-34 (IL-34). It plays important roles in development and in innate immunity by regulating the development of most tissue macrophages and osteoclasts, of Langerhans cells of the skin, of Paneth cells of the small intestine, and of brain microglia. It also regulates the differentiation of neural progenitor cells and controls functions of oocytes and trophoblastic cells in the female reproductive tract. Owing to this broad tissue expression pattern, it plays a central role in neoplastic, inflammatory, and neurological diseases. In this review we summarize the evolution, structure, and regulation of expression of the CSF-1R gene. We discuss the structures of CSF-1, IL-34, and the CSF-1R and the mechanism of ligand binding to and activation of the receptor. We further describe the pathways regulating macrophage survival, proliferation, differentiation, and chemotaxis downstream from the CSF-1R.
Collapse
Affiliation(s)
- E Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
16
|
Yang SK, Hong M, Zhao W, Jung Y, Baek J, Tayebi N, Kim KM, Ye BD, Kim KJ, Park SH, Lee I, Lee EJ, Kim WH, Cheon JH, Kim YH, Jang BI, Kim HS, Choi JH, Koo JS, Lee JH, Jung SA, Lee YJ, Jang JY, Shin HD, Kang D, Youn HS, Liu J, Song K. Genome-wide association study of Crohn's disease in Koreans revealed three new susceptibility loci and common attributes of genetic susceptibility across ethnic populations. Gut 2014; 63:80-7. [PMID: 23850713 DOI: 10.1136/gutjnl-2013-305193] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Crohn's disease (CD) is an intractable inflammatory bowel disease (IBD) of unknown cause. Recent meta-analysis of the genome-wide association studies (GWAS) and Immunochip data identified 163 susceptibility loci to IBD in Caucasians, however there are limited studies in other populations. METHODS We performed a GWAS and two validation studies in the Korean population comprising a total of 2311 patients with CD and 2442 controls. RESULTS We confirmed four previously reported loci: TNFSF15, IL23R, the major histocompatibility complex region, and the RNASET2-FGFR1OP-CCR6 region. We identified three new susceptibility loci at genome-wide significance: rs6856616 at 4p14 (OR=1.43, combined p=3.60×10(-14)), rs11195128 at 10q25 (OR=1.42, combined p=1.55×10(-10)) and rs11235667 at 11q13 (OR=1.46, combined p=7.15×10(-9)), implicating ATG16L2 and/or FCHSD2 as novel susceptibility genes for CD. Further analysis of the 11q13 locus revealed a non-synonymous single nucleotide polymorphism (SNP) (R220W/rs11235604) in the evolutionarily conserved region of ATG16L2 with stronger association (OR=1.61, combined p=2.44×10(-12)) than rs11235667, suggesting ATG16L2 as a novel susceptibility gene for CD and rs11235604 to be a potential causal variant of the association. Two of the three SNPs (rs6856616 (p=0.00024) and rs11195128 (p=5.32×10(-5))) showed consistent patterns of association in the International IBD Genetics Consortium dataset. Together, the novel and replicated loci accounted for 5.31% of the total genetic variance for CD risk in Koreans. CONCLUSIONS Our study provides new biological insight to CD and supports the complementary value of genetic studies in different populations.
Collapse
Affiliation(s)
- Suk-Kyun Yang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, , Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Following pathogen infection or tissue damage, the stimulation of pattern recognition receptors on the cell surface and in the cytoplasm of innate immune cells activates members of each of the major mitogen-activated protein kinase (MAPK) subfamilies--the extracellular signal-regulated kinase (ERK), p38 and Jun N-terminal kinase (JNK) subfamilies. In conjunction with the activation of nuclear factor-κB and interferon-regulatory factor transcription factors, MAPK activation induces the expression of multiple genes that together regulate the inflammatory response. In this Review, we discuss our current knowledge about the regulation and the function of MAPKs in innate immunity, as well as the importance of negative feedback loops in limiting MAPK activity to prevent host tissue damage. We also examine how pathogens have evolved complex mechanisms to manipulate MAPK activation to increase their virulence. Finally, we consider the potential of the pharmacological targeting of MAPK pathways to treat autoimmune and inflammatory diseases.
Collapse
|
18
|
Transcriptional profiling and pathway analysis of CSF-1 and IL-34 effects on human monocyte differentiation. Cytokine 2013; 63:10-17. [PMID: 23684409 DOI: 10.1016/j.cyto.2013.04.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2012] [Revised: 10/29/2012] [Accepted: 04/11/2013] [Indexed: 01/05/2023]
Abstract
CSF-1 is the well-known ligand for CSF-1R, which plays a vital role in monocyte-macrophage generation, survival, and function. IL-34 is a newly discovered cytokine that also signals through CSF-1R. Although there are limited data for downstream signaling and pathway activation for CSF-1, none are published, to date, for expression profiles of IL-34. The objective of this study was to characterize and compare the signaling pathways downstream of the CSF-1R receptor, based on these two ligands. This was accomplished through transcriptional profiling and pathway analysis of CD14(+) human monocytes differentiated with each ligand. Additionally, cells were treated with a CSF-1R inhibitor GW2580 to establish that observations associated with each ligand were CSF-1R mediated. Gene expression profiles were generated for each condition using Agilent 4x44K Whole Human Genome Microarrays. Overall profiles generated by each cytokine were similar (~75% of genes) with a dampened effect noted on some pathways (~25% of genes) with IL-34. One key difference observed, between the two cytokines was in the repression of CCR2 message. A similar divergence in protein level was established by FACS analysis. The differential effect on CCR2 expression has major implications for monocyte/macrophage biology including homeostasis and function. Further study of IL-34 effects on monocyte/macrophage biology will shed light on the specific role each ligand plays and the context in which these roles are important. To our knowledge, this study is the first to illustrate downstream transcriptional profiles and pathways of IL-34 in comparison with CSF-1 and identify notable differences in CCR2 expression.
Collapse
|
19
|
Abstract
Antigen-presenting cells (APCs) are critical targets of Ebola virus (EBOV) infection in vivo. However, the susceptibility of monocytes to infection is controversial. Studies indicate productive monocyte infection, and yet monocytes are also reported to be resistant to EBOV GP-mediated entry. In contrast, monocyte-derived macrophages and dendritic cells are permissive for both EBOV entry and replication. Here, freshly isolated monocytes are demonstrated to indeed be refractory to EBOV entry. However, EBOV binds monocytes, and delayed entry occurs during monocyte differentiation. Cultured monocytes spontaneously downregulate the expression of viral entry restriction factors such as interferon-inducible transmembrane proteins, while upregulating the expression of critical EBOV entry factors cathepsin B and NPC1. Moreover, these processes are accelerated by EBOV infection. Finally, ectopic expression of NPC1 is sufficient to rescue entry into an undifferentiated, normally nonpermissive monocytic cell line. These results define the molecular basis for infection of APCs and suggest means to limit APC infection.
Collapse
|
20
|
Leander R, Dai S, Schlesinger LS, Friedman A. A mathematical model of CR3/TLR2 crosstalk in the context of Francisella tularensis infection. PLoS Comput Biol 2012; 8:e1002757. [PMID: 23133361 PMCID: PMC3486853 DOI: 10.1371/journal.pcbi.1002757] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 09/05/2012] [Indexed: 02/04/2023] Open
Abstract
Complement Receptor 3 (CR3) and Toll-like Receptor 2 (TLR2) are pattern recognition receptors expressed on the surface of human macrophages. Although these receptors are essential components for recognition by the innate immune system, pathogen coordinated crosstalk between them can suppress the production of protective cytokines and promote infection. Recognition of the virulent Schu S4 strain of the intracellular pathogen Francisella tularensis by host macrophages involves CR3/TLR2 crosstalk. Although experimental data provide evidence that Lyn kinase and PI3K are essential components of the CR3 pathway that influences TLR2 activity, additional responsible upstream signaling components remain unknown. In this paper we construct a mathematical model of CR3 and TLR2 signaling in response to F. tularensis. After demonstrating that the model is consistent with experimental results we perform numerical simulations to evaluate the contributions that Akt and Ras-GAP make to ERK inhibition. The model confirms that phagocytosis-associated changes in the composition of the cell membrane can inhibit ERK activity and predicts that Akt and Ras-GAP synergize to inhibit ERK.
Collapse
Affiliation(s)
- Rachel Leander
- Mathematical Biosciences Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Shipan Dai
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
| | - Larry S. Schlesinger
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
| | - Avner Friedman
- Mathematical Biosciences Institute, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
21
|
Le Goff A, Ji Z, Leclercq B, Bourette RP, Mougel A, Guerardel C, de Launoit Y, Vicogne J, Goormachtigh G, Fafeur V. Anti-apoptotic role of caspase-cleaved GAB1 adaptor protein in hepatocyte growth factor/scatter factor-MET receptor protein signaling. J Biol Chem 2012; 287:35382-35396. [PMID: 22915589 PMCID: PMC3471683 DOI: 10.1074/jbc.m112.409797] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 08/20/2012] [Indexed: 11/06/2022] Open
Abstract
The GRB2-associated binder 1 (GAB1) docking/scaffold protein is a key mediator of the MET-tyrosine kinase receptor activated by hepatocyte growth factor/scatter factor (HGF/SF). Activated MET promotes recruitment and tyrosine phosphorylation of GAB1, which in turn recruits multiple proteins and mediates MET signaling leading to cell survival, motility, and morphogenesis. We previously reported that, without its ligand, MET is a functional caspase target during apoptosis, allowing the generation of a p40-MET fragment that amplifies apoptosis. In this study we established that GAB1 is also a functional caspase target by evidencing a caspase-cleaved p35-GAB1 fragment that contains the MET binding domain. GAB1 is cleaved by caspases before MET, and the resulting p35-GAB1 fragment is phosphorylated by MET upon HGF/SF binding and can interact with a subset of GAB1 partners, PI3K, and GRB2 but not with SHP2. This p35-GAB1 fragment favors cell survival by maintaining HGF/SF-induced MET activation of AKT and by hindering p40-MET pro-apoptotic function. These data demonstrate an anti-apoptotic role of caspase-cleaved GAB1 in HGF/SF-MET signaling.
Collapse
Affiliation(s)
- Arnaud Le Goff
- CNRS UMR 8161, Institut de Biologie de Lille, Université Lille-Nord de France, Institut Pasteur de Lille, IFR142, Lille, France
| | - Zongling Ji
- CNRS UMR 8161, Institut de Biologie de Lille, Université Lille-Nord de France, Institut Pasteur de Lille, IFR142, Lille, France; Faculty of Life Sciences, C2222 Michael Smith Building, University of Manchester, Manchester, United Kingdom
| | - Bérénice Leclercq
- CNRS UMR 8161, Institut de Biologie de Lille, Université Lille-Nord de France, Institut Pasteur de Lille, IFR142, Lille, France
| | - Roland P Bourette
- CNRS UMR 8161, Institut de Biologie de Lille, Université Lille-Nord de France, Institut Pasteur de Lille, IFR142, Lille, France
| | - Alexandra Mougel
- CNRS UMR 8161, Institut de Biologie de Lille, Université Lille-Nord de France, Institut Pasteur de Lille, IFR142, Lille, France
| | - Cateline Guerardel
- CNRS UMR 8161, Institut de Biologie de Lille, Université Lille-Nord de France, Institut Pasteur de Lille, IFR142, Lille, France
| | - Yvan de Launoit
- CNRS UMR 8161, Institut de Biologie de Lille, Université Lille-Nord de France, Institut Pasteur de Lille, IFR142, Lille, France
| | - Jérôme Vicogne
- CNRS UMR 8161, Institut de Biologie de Lille, Université Lille-Nord de France, Institut Pasteur de Lille, IFR142, Lille, France
| | - Gautier Goormachtigh
- CNRS UMR 8161, Institut de Biologie de Lille, Université Lille-Nord de France, Institut Pasteur de Lille, IFR142, Lille, France
| | - Véronique Fafeur
- CNRS UMR 8161, Institut de Biologie de Lille, Université Lille-Nord de France, Institut Pasteur de Lille, IFR142, Lille, France.
| |
Collapse
|
22
|
Gab2 promotes colony-stimulating factor 1-regulated macrophage expansion via alternate effectors at different stages of development. Mol Cell Biol 2011; 31:4563-81. [PMID: 21930791 DOI: 10.1128/mcb.05706-11] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Colony-stimulating factor 1 (CSF-1) receptor (CSF-1R, or macrophage CSF receptor [M-CSFR]) is the primary regulator of the proliferation, survival, and differentiation of mononuclear phagocytes (MNPs), but the critical CSF-1 signals for these functions are unclear. The scaffold protein Gab2 is a major tyrosyl phosphoprotein in the CSF-1R signaling network. Here we demonstrate that Gab2 deficiency results in profoundly defective expansion of CSF-1R-dependent MNP progenitors in the bone marrow, through decreased proliferation and survival. Reconstitution and phospho-flow studies show that downstream of CSF-1R, Gab2 uses phosphatidylinositol 3-kinase (PI3K)-Akt and extracellular signal-regulated kinase (Erk) to regulate MNP progenitor expansion. Unexpectedly, Gab2 ablation enhances Jun N-terminal protein kinase 1 (JNK1) phosphorylation in differentiated MNPs but reduces their proliferation; inhibition of JNK signaling or reduction of JNK1 levels restores proliferation. MNP recruitment to inflammatory sites and the corresponding bone marrow response is strongly impaired in Gab2-deficient mice. Our data provide genetic and biochemical evidence that CSF-1R, through Gab2, utilizes different effectors at different stages of MNP development to promote their expansion.
Collapse
|
23
|
Ehnert S, Schyschka L, Noss A, Knobeloch D, Kleeff J, Büchler P, Gillen S, Stöckle U, Burkhart J, Fabian E, Nussler AK. Further characterization of autologous NeoHepatocytes for in vitro toxicity testing. Toxicol In Vitro 2011; 25:1203-1208. [PMID: 21621600 DOI: 10.1016/j.tiv.2011.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 05/13/2011] [Accepted: 05/13/2011] [Indexed: 10/18/2022]
Abstract
Gold standard for in vitro toxicity tests and drug screenings is primary human hepatocytes (hHeps). Because of their limited availability efforts have been made to provide alternatives, e.g., monocyte-derived NeoHepatocytes. In the past years it has been critically discussed if gaining hepatocyte features is associated with trans-differentiation of monocytes or their activation towards a macrophage phenotype. Generating NeoHepatocytes in the presence of six different human AB sera, fetal calf serum (FCS) or autologous serum showed that yield and quality of NeoHepatocytes is inversely correlated to macrophage activation. Using autologous serum constantly the highest amount of cells with the best metabolic capacity was obtained. Focus of this study was to further analyze bio-transformation capacity of the optimized NeoHepatocytes for use as in vitro toxicity test-system. Treatment of the optimized NeoHepatocytes with two different pro-teratogenic substances with corresponding metabolites and eight known hepatotoxins showed comparable toxicity to hHeps. Bio-transformation rates, assessed by testosterone metabolism, were comparable in both cell types. Our data reveal that use of autologous serum reduced macrophage activation which improved yield and function of NeoHepatocytes resulting in bio-transformation and toxicity profiles comparable to hHeps. Thus, their easy accessibility makes them an ideal candidate for in vitro toxicity studies.
Collapse
Affiliation(s)
- S Ehnert
- Dept. of Traumatology, MRI, Technische Universität München, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Olsnes C, Olofsson J, Aarstad HJ. MAPKs ERK and p38, but not JNK Phosphorylation, Modulate IL-6 and TNF-α Secretion Following OK-432 In Vitro Stimulation of Purified Human Monocytes. Scand J Immunol 2011; 74:114-25. [DOI: 10.1111/j.1365-3083.2011.02555.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
25
|
Popp T, Egea V, Kehe K, Steinritz D, Schmidt A, Jochum M, Ries C. Sulfur mustard induces differentiation in human primary keratinocytes: opposite roles of p38 and ERK1/2 MAPK. Toxicol Lett 2011; 204:43-51. [PMID: 21524694 DOI: 10.1016/j.toxlet.2011.04.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 04/01/2011] [Accepted: 04/11/2011] [Indexed: 02/03/2023]
Abstract
The chemical warfare agent sulfur mustard (SM) severely affects the regeneration capacity of skin. The underlying molecular and cellular mechanisms, however, are far from clear. Here, we demonstrate that normal human epidermal keratinocytes (NHEK) after exposure to SM strongly upregulated expression of keratin-1, involucrin, and loricrin, thus indicating premature epidermal differentiation. Furthermore, proliferation was repressed after treatment with SM. Analysis of intracellular signaling in NHEK revealed that SM enhances phosphorylation, nuclear translocation, and activity of the mitogen-activated protein kinases (MAPK) p38 and ERK1/2. Inhibition of p38 activity downregulated expression of keratin-1 and loricrin, whereas blockage of ERK1/2 significantly stimulated biosynthesis of these markers, pointing to opposite roles of p38 and ERK1/2 in the differentiation process. Simultaneous interruption of p38 and ERK1/2 activity led to a decreased expression of keratin-1 and loricrin. This suggests that NHEK differentiation is essentially controlled by p38 activity which may be negatively influenced by ERK1/2 activity. Functional analysis demonstrated that SM affects NHEK in their ability to migrate through extracellular matrix which can be rescued upon application of an inhibitor of p38 activity. Thus, our findings indicate that SM triggers premature differentiation in keratinocytes via p38 activity which may contribute to impaired regeneration of SM-injured skin.
Collapse
Affiliation(s)
- Tanja Popp
- Division of Clinical Chemistry and Clinical Biochemistry, Ludwig-Maximilians-University of Munich, Nussbaumstrasse 20, 80336 Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
26
|
Ehnert S, Seeliger C, Vester H, Schmitt A, Saidy-Rad S, Lin J, Neumaier M, Gillen S, Kleeff J, Friess H, Burkhart J, Stöckle U, Nüssler AK. Autologous serum improves yield and metabolic capacity of monocyte-derived hepatocyte-like cells: possible implication for cell transplantation. Cell Transplant 2011; 20:1465-1477. [PMID: 21294943 DOI: 10.3727/096368910x550224] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hepatocyte-transplantation is a therapeutic approach for diverse acute and chronic liver diseases. As availability of primary cells is limited, there is an increasing demand for hepatocyte-like cells (e.g., neohepatocytes generated from peripheral blood monocytes). The aim of this study was to evaluate the effects of six different human AB sera, fetal calf serum, or autologous serum on production of neohepatocytes. The yield and quality of neohepatocytes varied considerably depending on the different sera. Using autologous sera for the whole production process we constantly generated the highest amount of cells with the highest metabolic activity for phase I (e.g., CYP1A1/2, CYP3A4) and phase II enzymes (e.g., glutathione-S-transferase). Moreover, similar effects were seen examining glucose and urea metabolism. Especially, glucose-6-phosphatase and PAS staining showed distinct serum-dependent differences. The role of macrophage activation was investigated by measuring the secretion of TNF-α, TGF-β, and RANKL, MMP activity, as well as mRNA levels of different interleukins in programmable cells of monocytic origin (PCMO). Our data clearly demonstrate that the use of autologous serum reduced initial macrophage activation in PCMOs and subsequently improved both yield and function of differentiated neohepatocytes. The autologous approach presented here might also be useful in other stem cell preparation processes where cell activation during generation shall be kept to a minimum.
Collapse
Affiliation(s)
- S Ehnert
- Department of Traumatology, MRI, Technische Universität München, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|