1
|
Runyan LA, Kudryashova E, Agrawal R, Mohamed M, Kudryashov DS. Human plastins are novel cytoskeletal pH sensors with a reduced F-actin bundling capacity at basic pH. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645573. [PMID: 40196613 PMCID: PMC11974883 DOI: 10.1101/2025.03.26.645573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Intracellular pH (pH i ) is a fundamental component of cell homeostasis. Controlled elevations in pH i precede and accompany cell polarization, cytokinesis, and directional migration. pH dysregulation contributes to cancer, neurodegenerative diseases, diabetes, and other metabolic disorders. While cytoskeletal rearrangements are crucial for these processes, only a few cytoskeletal proteins, namely Cdc42, cofilin, talin, cortactin, α-actinin, and AIP1 have been documented as pH sensors. Here, we report that actin-bundling proteins plastin 2 (PLS2, aka LCP1) and plastin 3 (PLS3) respond to physiological scale pH fluctuations by a reduced F-actin bundling at alkaline pH. The inhibition of PLS2 actin-bundling activity at elevated pH stems from the reduced affinity of the N-terminal actin-binding domain (ABD1) to actin. In fibroblast cells, elevated cytosolic pH caused the dissociation of ectopically expressed PLS2 from actin structures, whereas acidic conditions promoted its tighter association with focal adhesions and stress fibers. We identified His207 as one of the pH-sensing residues whose mutation to Lys and Tyr reduces pH sensitivity by enhancing and inhibiting the bundling ability, respectively. Our results suggest that weaker actin bundling by plastin isoforms at alkaline pH favors higher dynamics of the actin cytoskeleton. Therefore, like other cytoskeleton pH sensors, plastins promote disassembly and faster dynamics of cytoskeletal components during cytokinesis and cell migration. Since both plastins are implemented in cancer, their pH sensitivity may contribute to the accelerated proliferation and enhanced invasive and metastatic potentials of cancer cells at alkaline pH i . Abstract Figure
Collapse
|
2
|
Haap‐Hoff A, Freeley M, Dempsey E, Dunican D, Bennett E, Triglia D, Skubis‐Zegadlo J, Mitchell Davies A, Kelleher D, Long A. RNAi library screening reveals Gβ1, Casein Kinase 2 and ICAP-1 as novel regulators of LFA-1-mediated T cell polarity and migration. Immunol Cell Biol 2025; 103:73-92. [PMID: 39607284 PMCID: PMC11688611 DOI: 10.1111/imcb.12838] [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: 06/25/2024] [Revised: 10/28/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024]
Abstract
The αLβ2 integrin LFA-1 plays a key role in T-cell adhesion to the endothelial vasculature and migration into both secondary lymphoid organs and peripheral tissues via interactions with its target protein ICAM-1, but the pathways that regulate LFA-1-mediated T-cell polarity and migration are not fully understood. In this study we screened two RNAi libraries targeting G protein-coupled receptors (GPCR)/GPCR-associated proteins and kinases in a HuT 78 T cell line model of LFA-1-stimulated T-cell migration. Based on staining of the actin cytoskeleton, multiple parameters to measure cell morphology were used to assess the contribution of 1109 genes to LFA-1-mediated T-cell polarity and migration. These RNAi screens identified a number of both novel and previously identified genes that either increased or decreased the polarity and migratory capacity of these cells. Following multiparametric analysis, hierarchical clustering and pathway analysis, three of these genes were characterized in further detail using primary human T cells, revealing novel roles for the heterotrimeric G protein subunit Gβ1 and Casein Kinase 2 in LFA-1-mediated T-cell polarity and migration in vitro. Our studies also highlighted a new role for ICAP-1, an adaptor protein previously described to be associated with β1 integrins, in β2 integrin LFA-1-directed migration in T cells. Knockdown of ICAP-1 expression in primary T cells revealed a role in cell polarity, cell velocity and transmigration towards SDF-1 for this adaptor protein. This study therefore uncovers new roles for GPCR/GPCR-associated proteins and kinases in T-cell migration and provides potential novel targets for modulation of the T-cell immune response.
Collapse
Affiliation(s)
- Antje Haap‐Hoff
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
| | - Michael Freeley
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
- Present address:
School of BiotechnologyDublin City UniversityDublinIreland
| | - Eugene Dempsey
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
| | - Dara Dunican
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
| | - Emily Bennett
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
| | - Denise Triglia
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
| | - Joanna Skubis‐Zegadlo
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
| | - Anthony Mitchell Davies
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
| | - Dermot Kelleher
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
- Present address:
Faculty of MedicineUniversity of British ColumbiaVancouverBCCanada
| | - Aideen Long
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
| |
Collapse
|
3
|
Ceccherini E, Morlando A, Norelli F, Coco B, Bellini M, Brunetto MR, Cecchettini A, Rocchiccioli S. Cytoskeleton Remodeling-Related Proteins Represent a Specific Salivary Signature in PSC Patients. Molecules 2024; 29:5783. [PMID: 39683940 DOI: 10.3390/molecules29235783] [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: 10/02/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Primary sclerosing cholangitis (PSC) and Primary biliary cholangitis (PBC) are chronic inflammatory biliary diseases characterized by progressive damage of the bile ducts, resulting in hepatobiliary fibrosis and cirrhosis. Currently, specific biomarkers that allow to distinguish between PSC and PBC do not exist. In this study, we examined the salivary proteome by carrying out a comprehensive and non-invasive screening aimed at highlighting possible quali-quantitative protein deregulations that could be the starting point for the identification of effective biomarkers in future. Saliva samples collected from 6 PBC patients were analyzed using a liquid chromatography-tandem mass spectrometry technique, and the results were compared with those previously obtained in the PSC group. We identified 40 proteins as significantly deregulated in PSC patients compared to the PBC group. The Gene Ontology and pathway analyses highlighted that several proteins (e.g., small integral membrane protein 22, cofilin-1, macrophage-capping protein, plastin-2, and biliverdin reductase A) were linked to innate immune responses and actin cytoskeleton remodeling, which is a critical event in liver fibrosis and cancer progression. These findings provide new foundations for a deeper understanding of the pathophysiology of PSC and demonstrate that saliva is a suitable biological sample for obtaining proteomic fingerprints useful in the search for biomarkers capable of discriminating between the two cholestatic diseases.
Collapse
Affiliation(s)
- Elisa Ceccherini
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
| | - Antonio Morlando
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
| | - Francesco Norelli
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
| | - Barbara Coco
- Hepatology Unit, Reference Centre of the Tuscany Region for Chronic Liver Disease and Cancer, University Hospital of Pisa, 56124 Pisa, Italy
| | - Massimo Bellini
- Gastrointestinal Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy
| | - Maurizia Rossana Brunetto
- Hepatology Unit, Reference Centre of the Tuscany Region for Chronic Liver Disease and Cancer, University Hospital of Pisa, 56124 Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Antonella Cecchettini
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Silvia Rocchiccioli
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
| |
Collapse
|
4
|
Pan S, Wan M, Jin H, Ning R, Zhang J, Han X. LCP1 correlates with immune infiltration: a prognostic marker for triple-negative breast cancer. BMC Immunol 2024; 25:42. [PMID: 38977952 PMCID: PMC11229261 DOI: 10.1186/s12865-024-00635-x] [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: 03/13/2024] [Accepted: 06/25/2024] [Indexed: 07/10/2024] Open
Abstract
OBJECTIVE Triple-Negative Breast Cancer (TNBC) is known for its aggressiveness and treatment challenges due to the absence of ER, PR, and HER2 receptors. Our work emphasizes the prognostic value of LCP1 (Lymphocyte cytosolic protein 1), which plays a crucial role in cell processes and immune cell activity, to predict outcomes and guide treatments in TNBC. METHODS We explored LCP1 as a potential biomarker in TNBC and investigated the mRNA and protein expression levels of LCP1. We investigated different databases, including GTEX, TCGA, GEO, cBioPortal and Kaplan-Meier Plotter. Immunohistochemistry on TNBC and benign tumor samples was performed to examine LCP1's relationship with patient clinical characteristics and macrophage markers. We also assessed survival rates, immune cell infiltration, and drug sensitivity related to LCP1 using various bioinformatics tools. RESULTS The results indicated that LCP1 expression was higher in TNBC tissues compared to adjacent normal tissues. However, high expression of LCP1 was significantly associated with favorable survival outcomes in patients with TNBC. Enrichment analysis revealed that genes co-expressed with LCP1 were significantly enriched in various immune processes. LCP1 showed a positive correlation with the infiltration of resting dendritic cells, M1 macrophages, and memory CD4 T cells, and a negative correlation with M2 macrophages. Further analysis suggested a link between high levels of LCP1 and increased survival outcomes in cancer patients receiving immunotherapy. CONCLUSION LCP1 may serve as a potential diagnostic and prognostic biomarker for TNBC, which was closely associated with immune cell infiltration, particularly M1 and M2 macrophages. Our findings may provide valuable insights into immunotherapeutic strategies for TNBC patients.
Collapse
Affiliation(s)
- Shuaikang Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Wan Nan Medical College, Wuhu, China
| | - Mengting Wan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongwei Jin
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Ran Ning
- Department of Pathology, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, 238000, Anhui, China
| | - Jinguo Zhang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Xinghua Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
5
|
Wang Y, Liu Z, Qi Y, Wu J, Liu B, Cui X. Activin A, a Novel Chemokine, Induces Mouse NK Cell Migration via AKT and Calcium Signaling. Cells 2024; 13:728. [PMID: 38727264 PMCID: PMC11083611 DOI: 10.3390/cells13090728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
Natural killer (NK) cells can migrate quickly to the tumor site to exert cytotoxic effects on tumors, and some chemokines, including CXCL8, CXCL10 or and CXCL12, can regulate the migration of NK cells. Activin A, a member of the transforming growth factor β (TGF-β) superfamily, is highly expressed in tumor tissues and involved in tumor development and immune cell activation. In this study, we focus on the effects of activin A on NK cell migration. In vitro, activin A induced NK cell migration and invasion, promoted cell polarization and inhibited cell adhesion. Moreover, activin A increased Ca2+, p-SMAD3 and p-AKT levels in NK cells. An AKT inhibitor and Ca2+ chelator partially blocked activin A-induced NK cell migration. In vivo, exogenous activin A increased tumor-infiltrating NK cells in NS-1 cell solid tumors and inhibited tumor growth, and blocking endogenous activin A with anti-activin A antibody reduced tumor-infiltrating NK cells in 4T-1 cell solid tumors. These results suggest that activin A induces NK cell migration through AKT signaling and calcium signaling and may enhance the antitumor effect of NK cells by increasing tumor-infiltrating NK cells.
Collapse
Affiliation(s)
- Yunfeng Wang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (Y.W.); (Z.L.); (Y.Q.)
- Key Laboratory of Neuroimmunology and Clinical Immunology, Changchun 130021, China
| | - Zhonghui Liu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (Y.W.); (Z.L.); (Y.Q.)
- Key Laboratory of Neuroimmunology and Clinical Immunology, Changchun 130021, China
| | - Yan Qi
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (Y.W.); (Z.L.); (Y.Q.)
- Key Laboratory of Neuroimmunology and Clinical Immunology, Changchun 130021, China
| | - Jiandong Wu
- Bionic Sensing and Intelligence Center, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China;
| | - Boyang Liu
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
- Department of Scientific Research, Jilin Jianzhu University, Changchun 130118, China
| | - Xueling Cui
- Key Laboratory of Neuroimmunology and Clinical Immunology, Changchun 130021, China
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| |
Collapse
|
6
|
Ozulumba T, Montalbine AN, Ortiz-Cárdenas JE, Pompano RR. New tools for immunologists: models of lymph node function from cells to tissues. Front Immunol 2023; 14:1183286. [PMID: 37234163 PMCID: PMC10206051 DOI: 10.3389/fimmu.2023.1183286] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
The lymph node is a highly structured organ that mediates the body's adaptive immune response to antigens and other foreign particles. Central to its function is the distinct spatial assortment of lymphocytes and stromal cells, as well as chemokines that drive the signaling cascades which underpin immune responses. Investigations of lymph node biology were historically explored in vivo in animal models, using technologies that were breakthroughs in their time such as immunofluorescence with monoclonal antibodies, genetic reporters, in vivo two-photon imaging, and, more recently spatial biology techniques. However, new approaches are needed to enable tests of cell behavior and spatiotemporal dynamics under well controlled experimental perturbation, particularly for human immunity. This review presents a suite of technologies, comprising in vitro, ex vivo and in silico models, developed to study the lymph node or its components. We discuss the use of these tools to model cell behaviors in increasing order of complexity, from cell motility, to cell-cell interactions, to organ-level functions such as vaccination. Next, we identify current challenges regarding cell sourcing and culture, real time measurements of lymph node behavior in vivo and tool development for analysis and control of engineered cultures. Finally, we propose new research directions and offer our perspective on the future of this rapidly growing field. We anticipate that this review will be especially beneficial to immunologists looking to expand their toolkit for probing lymph node structure and function.
Collapse
Affiliation(s)
- Tochukwu Ozulumba
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Alyssa N. Montalbine
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States
| | - Jennifer E. Ortiz-Cárdenas
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Rebecca R. Pompano
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
- Carter Immunology Center and University of Virginia (UVA) Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
7
|
Lin X, Krishnamoorthy P, Walker EC, Joshi H, Morley SC. Expression of non-phosphorylatable S5A-L-plastin exerts phenotypes distinct from L-plastin deficiency during podosome formation and phagocytosis. Front Cell Dev Biol 2023; 11:1020091. [PMID: 37138794 PMCID: PMC10150066 DOI: 10.3389/fcell.2023.1020091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 04/03/2023] [Indexed: 05/05/2023] Open
Abstract
Introduction: The actin cytoskeleton remodels to enable diverse processes essential to immunity, such as cell adhesion, migration and phagocytosis. A panoply of actin-binding proteins regulate these rapid rearrangements to induce actin-based shape changes and to generate force. L-plastin (LPL) is a leukocyte-specific, actin-bundling protein that is regulated in part by phosphorylation of the Ser-5 residue. LPL deficiency in macrophages impairs motility, but not phagocytosis; we recently found that expression of LPL in which the S5 residue is converted to a non-phosphorylatable alanine (S5A-LPL) resulted in diminished phagocytosis, but unimpaired motility. Methods: To provide mechanistic insight into these findings, we now compare the formation of podosomes (an adhesive structure) and phagosomes in alveolar macrophages derived from wild-type (WT), LPL-deficient, or S5A-LPL mice. Both podosomes and phagosomes require rapid remodeling of actin, and both are force-transmitting. Actin rearrangement, force generation, and signaling rely upon recruitment of many actin-binding proteins, including the adaptor protein vinculin and the integrin-associated kinase Pyk2. Prior work suggested that vinculin localization to podosomes was independent of LPL, while Pyk2 was displaced by LPL deficiency. We therefore chose to compare vinculin and Pyk2 co-localization with F-actin at sites of adhesion of phagocytosis in AMs derived from WT, S5A-LPL or LPL-/- mice, using Airyscan confocal microscopy. Results: As described previously, podosome stability was significantly disrupted by LPL deficiency. In contrast, LPL was dispensable for phagocytosis and was not recruited to phagosomes. Recruitment of vinculin to sites of phagocytosis was significantly enhanced in cells lacking LPL. Expression of S5A-LPL impeded phagocytosis, with reduced appearance of ingested bacteria-vinculin aggregates. Discussion: Our systematic analysis of the regulation of LPL during podosome vs. phagosome formation illuminates essential remodeling of actin during key immune processes.
Collapse
Affiliation(s)
- Xue Lin
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Praveen Krishnamoorthy
- Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, MO, United States
| | - Emma C. Walker
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Hemant Joshi
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Sharon Celeste Morley
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, United States
- *Correspondence: Sharon Celeste Morley,
| |
Collapse
|
8
|
Joshi H, Morley SC. Efficient T Cell Migration and Activation Require L-Plastin. Front Immunol 2022; 13:916137. [PMID: 35844504 PMCID: PMC9277003 DOI: 10.3389/fimmu.2022.916137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/02/2022] [Indexed: 11/28/2022] Open
Abstract
Rapid re-organization of the actin cytoskeleton supports T-cell trafficking towards immune sites and interaction with antigen presenting cells (APCs). F-actin rearrangement enables T-cell trafficking by stabilizing adhesion to vascular endothelial cells and promoting transendothelial migration. T-cell/APC immune synapse (IS) maturation also relies upon f-actin-anchored LFA-1:ICAM-1 ligation. Therefore, efficient T-cell responses require tight regulation of f-actin dynamics. In this review, we summarize how the actin-bundling protein L-plastin (LPL) regulates T-cell activation and migration. LPL enhances f-actin polymerization and also directly binds to the β2 chain of the integrin LFA-1 to support intercellular adhesion and IS formation in human and murine T cells. LPL- deficient T cells migrate slowly in response to chemo-attractants such as CXCL12, CCL19, and poorly polarize towards ICAM-1. Loss of LPL impairs thymic egress and intranodal motility. LPL is also required for T-cell IS maturation with APCs, and therefore for efficient cytokine production and proliferation. LPL-/- mice are less susceptible to T-cell mediated pathologies, such as allograft rejection and experimental autoimmune encephalomyelitis (EAE). LPL activity is regulated by its N-terminal “headpiece”, which contains serine and threonine phosphorylation and calcium- and calmodulin-binding sites. LPL phosphorylation is required for lamellipodia formation during adhesion and migration, and also for LFA-1 clustering during IS formation. However, the precise molecular interactions by which LPL supports T-cell functional responses remain unclear. Future studies elucidating LPL-mediated regulation of T-cell migration and/or activation may illuminate pathways for therapeutic targeting in T-cell-mediated diseases.
Collapse
Affiliation(s)
- Hemant Joshi
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
- Division of Immunobiology, Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO, United States
| | - Sharon Celeste Morley
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
- Division of Immunobiology, Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO, United States
- *Correspondence: Sharon Celeste Morley,
| |
Collapse
|
9
|
Du W, Nair P, Johnston A, Wu PH, Wirtz D. Cell Trafficking at the Intersection of the Tumor-Immune Compartments. Annu Rev Biomed Eng 2022; 24:275-305. [PMID: 35385679 PMCID: PMC9811395 DOI: 10.1146/annurev-bioeng-110320-110749] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Migration is an essential cellular process that regulates human organ development and homeostasis as well as disease initiation and progression. In cancer, immune and tumor cell migration is strongly associated with immune cell infiltration, immune escape, and tumor cell metastasis, which ultimately account for more than 90% of cancer deaths. The biophysics and molecular regulation of the migration of cancer and immune cells have been extensively studied separately. However, accumulating evidence indicates that, in the tumor microenvironment, the motilities of immune and cancer cells are highly interdependent via secreted factors such as cytokines and chemokines. Tumor and immune cells constantly express these soluble factors, which produce a tightly intertwined regulatory network for these cells' respective migration. A mechanistic understanding of the reciprocal regulation of soluble factor-mediated cell migration can provide critical information for the development of new biomarkers of tumor progression and of tumor response to immuno-oncological treatments. We review the biophysical andbiomolecular basis for the migration of immune and tumor cells and their associated reciprocal regulatory network. We also describe ongoing attempts to translate this knowledge into the clinic.
Collapse
Affiliation(s)
- Wenxuan Du
- Institute for NanoBiotechnology Department of Chemical and Biomolecular Engineering, and Johns Hopkins Physical Sciences Oncology Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Praful Nair
- Institute for NanoBiotechnology Department of Chemical and Biomolecular Engineering, and Johns Hopkins Physical Sciences Oncology Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Adrian Johnston
- Institute for NanoBiotechnology Department of Chemical and Biomolecular Engineering, and Johns Hopkins Physical Sciences Oncology Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Pei-Hsun Wu
- Institute for NanoBiotechnology Department of Chemical and Biomolecular Engineering, and Johns Hopkins Physical Sciences Oncology Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Denis Wirtz
- Institute for NanoBiotechnology Department of Chemical and Biomolecular Engineering, and Johns Hopkins Physical Sciences Oncology Center, Johns Hopkins University, Baltimore, Maryland, USA,Department of Oncology, Department of Pathology, and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
10
|
Lymphocyte cytosolic protein 1 (L-plastin) I232F mutation impairs granulocytic proliferation and causes neutropenia. Blood Adv 2022; 6:2581-2594. [PMID: 34991157 PMCID: PMC9043934 DOI: 10.1182/bloodadvances.2021006398] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/17/2021] [Indexed: 11/20/2022] Open
Abstract
Neutrophils migrate into inflamed tissue, engage in phagocytosis, and clear pathogens or apoptotic cells. These processes require well-coordinated events involving the actin cytoskeleton. We describe a child with severe neutropenia and episodes of soft tissue infections and pneumonia. Bone marrow examination showed granulocytic hypoplasia with dysplasia. Whole-exome sequencing revealed a de novo heterozygous missense mutation in LCP1, which encodes the F-actin-binding protein Lymphocyte Cytosolic Protein 1. To determine its pathophysiological significance, we stably transduced cells with doxycycline-inducible wild-type LCP1 and LCP1 I232F lentiviral constructs. We observed dysplastic granulocytic 32D cells expressing LCP1 I232F cells. These cells showed decreased proliferation without a block in differentiation. In addition, expression of LCP1 I232F resulted in a cell cycle arrest at the G2/M phase, but it did not lead to increased levels of genes involved in apoptosis or the unfolded protein response. Both 32D and HeLa cells expressing mutant LCP1 displayed impaired cell motility and invasiveness. Flow cytometry showed increased F-actin. However, mutant LCP1-expressing 32D cells exhibited normal oxidative burst upon stimulation. Confocal imaging and subcellular fractionation revealed diffuse intracellular localization of LCP1, but only the mutant form was found in the nucleus. We conclude that LCP1 is a new gene involved in granulopoiesis, and the missense variant LCP1 I232F leads to neutropenia and granulocytic dysplasia with aberrant actin dynamics. Our work supports a model of neutropenia due to aberrant actin regulation.
Collapse
|
11
|
Joshi H, Almgren-Bell A, Anaya EP, Todd EM, Van Dyken SJ, Seth A, McIntire KM, Singamaneni S, Sutterwala F, Morley SC. L-plastin enhances NLRP3 inflammasome assembly and bleomycin-induced lung fibrosis. Cell Rep 2022; 38:110507. [PMID: 35294888 PMCID: PMC8998782 DOI: 10.1016/j.celrep.2022.110507] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 01/06/2022] [Accepted: 02/16/2022] [Indexed: 12/12/2022] Open
Abstract
Macrophage adhesion and stretching have been shown to induce interleukin (IL)-1β production, but the mechanism of this mechanotransduction remains unclear. Here we specify the molecular link between mechanical tension on tissue-resident macrophages and activation of the NLRP3 inflammasome, which governs IL-1β production. NLRP3 activation enhances antimicrobial defense, but excessive NLRP3 activity causes inflammatory tissue damage in conditions such as pulmonary fibrosis and acute respiratory distress syndrome. We find that the actin-bundling protein L-plastin (LPL) significantly enhances NLRP3 assembly. Specifically, LPL enables apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC) oligomerization during NLRP3 assembly by stabilizing ASC interactions with the kinase Pyk2, a component of cell-surface adhesive structures called podosomes. Upon treatment with exogenous NLRP3 activators, lung-resident alveolar macrophages (AMs) lacking LPL exhibit reduced caspase-1 activity, IL-1β cleavage, and gasdermin-D processing. LPL−/− mice display resistance to bleomycin-induced lung injury and fibrosis. These findings identify the LPL-Pyk2-ASC pathway as a target for modulation in NLRP3-mediated inflammatory conditions. In this study, Joshi et al. identify a crucial modulator, L-plastin, in lung inflammation. L-plastin supports the macrophage inflammatory response to enhance lung fibrosis during lung injury by connecting inflammation and mechanical stimuli in a process called mechanotransduction. The findings from this study will help determine efficient targets for diagnosis and treatment of lung inflammatory diseases.
Collapse
Affiliation(s)
- Hemant Joshi
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Division of Immunobiology, Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alison Almgren-Bell
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Division of Immunobiology, Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Edgar P Anaya
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Division of Immunobiology, Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Elizabeth M Todd
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Division of Immunobiology, Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven J Van Dyken
- Division of Immunobiology, Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anushree Seth
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Katherine M McIntire
- Division of Immunobiology, Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Srikanth Singamaneni
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Fayyaz Sutterwala
- Division of Infectious Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sharon C Morley
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Division of Immunobiology, Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
12
|
Fazil MHUT, Chirumamilla CS, Perez-Novo C, Wong BHS, Kumar S, Sze SK, Vanden Berghe W, Verma NK. The steroidal lactone withaferin A impedes T-cell motility by inhibiting the kinase ZAP70 and subsequent kinome signaling. J Biol Chem 2021; 297:101377. [PMID: 34742736 PMCID: PMC8637146 DOI: 10.1016/j.jbc.2021.101377] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 02/07/2023] Open
Abstract
The steroidal lactone withaferin A (WFA) is a dietary phytochemical, derived from Withania somnifera. It exhibits a wide range of biological properties, including immunomodulatory, anti-inflammatory, antistress, and anticancer activities. Here we investigated the effect of WFA on T-cell motility, which is crucial for adaptive immune responses as well as autoimmune reactions. We found that WFA dose-dependently (within the concentration range of 0.3–1.25 μM) inhibited the ability of human T-cells to migrate via cross-linking of the lymphocyte function-associated antigen-1 (LFA-1) integrin with its ligand, intercellular adhesion molecule 1 (ICAM-1). Coimmunoprecipitation of WFA interacting proteins and subsequent tandem mass spectrometry identified a WFA-interactome consisting of 273 proteins in motile T-cells. In particular, our data revealed significant enrichment of the zeta-chain-associated protein kinase 70 (ZAP70) and cytoskeletal actin protein interaction networks upon stimulation. Phospho-peptide mapping and kinome analysis substantiated kinase signaling downstream of ZAP70 as a key WFA target, which was further confirmed by bait-pulldown and Western immunoblotting assays. The WFA-ZAP70 interaction was disrupted by a disulfide reducing agent dithiothreitol, suggesting an involvement of cysteine covalent binding interface. In silico docking predicted WFA binding to ZAP70 at cystine 560 and 564 residues. These findings provide a mechanistic insight whereby WFA binds to and inhibits the ZAP70 kinase and impedes T-cell motility. We therefore conclude that WFA may be exploited to pharmacologically control host immune responses and potentially prevent autoimmune-mediated pathologies.
Collapse
Affiliation(s)
| | - Chandra Sekhar Chirumamilla
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signaling (PPES) and Integrated Personalized and Precision Oncology Network (IPPON), Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Claudina Perez-Novo
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signaling (PPES) and Integrated Personalized and Precision Oncology Network (IPPON), Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Brandon Han Siang Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, Singapore; NTU Institute for Health Technologies (HealthTech NTU), Interdisciplinary Graduate Programme, Nanyang Technological University Singapore, Singapore
| | - Sunil Kumar
- Indian Council of Agricultural Research-National Bureau of Agriculturally Important Microorganisms, Kushmaur, Mau, Uttar Pradesh, India
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore
| | - Wim Vanden Berghe
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signaling (PPES) and Integrated Personalized and Precision Oncology Network (IPPON), Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium.
| | - Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, Singapore.
| |
Collapse
|
13
|
Chellaiah MA. L-Plastin Phosphorylation: Possible Regulation by a TNFR1 Signaling Cascade in Osteoclasts. Cells 2021; 10:2432. [PMID: 34572081 PMCID: PMC8464874 DOI: 10.3390/cells10092432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/25/2021] [Accepted: 09/09/2021] [Indexed: 12/30/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF-α) signaling regulates phosphorylation of L-plastin, which is involved in forming the nascent sealing zone, a precursor zone for the matured sealing ring. This study aimed to illustrate the molecular mechanisms of L-plastin phosphorylation and the subsequent formation of the nascent sealing zone in osteoclasts treated with TNF-α. Here, we report that anti-TNF-receptor 1, inhibitors of signaling proteins (Src, PI3-K, Rho, and Rho-kinase), and siRNA of TRAF-6 attenuated the phosphorylation of LPL and filamentous actin content significantly in the presence of TNF-α. An inhibitor of integrin αvβ3, PKC, or PKA did not inhibit TNF-α-induced L-plastin phosphorylation. Inhibitors of Src and PI3-K and not Rho or Rho-kinase reduced tyrosine phosphorylation of TRAF-6, suggesting that Src and PI3-K regulate TRAF-6 phosphorylation, and Rho and Rho-kinase are downstream of TRAF-6 regulation. Osteoclasts expressing constitutively active or kinase-defective Src proteins were used to determine the role of Src on L-plastin phosphorylation; similarly, the effect of Rho was confirmed by transducing TAT-fused constitutively active (V14) or dominant-negative (N19) Rho proteins into osteoclasts. Pull-down analysis with glutathione S-transferase-fused SH2 and SH3 domains of Src and PI3-K demonstrated coprecipitation of L-plastin and TRAF-6 with the SH3 and SH2 domains of the PI3-K and Src proteins. However, the actual order of the interaction of proteins requires further elucidation; a comprehensive screening should corroborate the initial findings of protein interactions via the SH2/SH3 domains. Ultimately, inhibition of the interaction of proteins with SH2/SH3 could reduce L-plastin phosphorylation and affect NSZ formation and bone resorption in conditions that display osteoclast activation and bone loss.
Collapse
Affiliation(s)
- Meenakshi A Chellaiah
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA
| |
Collapse
|
14
|
Anaya EP, Lin X, Todd EM, Szasz TP, Morley SC. Novel Mouse Model Reveals That Serine Phosphorylation of L-Plastin Is Essential for Effective Splenic Clearance of Pneumococcus. THE JOURNAL OF IMMUNOLOGY 2021; 206:2135-2145. [PMID: 33858961 DOI: 10.4049/jimmunol.2000899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 02/19/2021] [Indexed: 01/04/2023]
Abstract
Asplenia imparts susceptibility to life-threatening sepsis with encapsulated bacteria, such as the pneumococcus. However, the cellular components within the splenic environment that guard against pneumococcal bacteremia have not been defined. The actin-bundling protein L-plastin (LPL) is essential for the generation of marginal zone B cells and for anti-pneumococcal host defense, as revealed by a mouse model of genetic LPL deficiency. In independent studies, serine phosphorylation of LPL at residue 5 (S5) has been described as a key "switch" in regulating LPL actin binding and subsequent cell motility, although much of the data are correlative. To test the importance of S5 phosphorylation in LPL function, and to specifically assess the requirement of LPL S5 phosphorylation in anti-pneumococcal host defense, we generated the "S5A" mouse, expressing endogenous LPL bearing a serine-to-alanine mutation at this position. S5A mice were bred to homozygosity, and LPL was expressed at levels equivalent to wild-type, but S5 phosphorylation was absent. S5A mice exhibited specific impairment in clearance of pneumococci following i.v. challenge, with 10-fold-higher bacterial bloodstream burden 24 h after challenge compared with wild-type or fully LPL-deficient animals. Defective bloodstream clearance correlated with diminished population of marginal zone macrophages and with reduced phagocytic capacity of multiple innate immune cells. Development and function of other tested leukocyte lineages, such as T and B cell motility and activation, were normal in S5A mice. The S5A mouse thus provides a novel system in which to elucidate the precise molecular control of critical immune cell functions in specific host-pathogen defense interactions.
Collapse
Affiliation(s)
- Edgar P Anaya
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO; and
| | - Xue Lin
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO; and
| | - Elizabeth M Todd
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO; and
| | - Taylor P Szasz
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO; and
| | - S Celeste Morley
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO; and .,Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
15
|
Aljohani H, Stains JP, Majumdar S, Srinivasan D, Senbanjo L, Chellaiah MA. Peptidomimetic inhibitor of L-plastin reduces osteoclastic bone resorption in aging female mice. Bone Res 2021; 9:22. [PMID: 33837180 PMCID: PMC8035201 DOI: 10.1038/s41413-020-00135-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 11/22/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022] Open
Abstract
L-plastin (LPL) was identified as a potential regulator of the actin-bundling process involved in forming nascent sealing zones (NSZs), which are precursor zones for mature sealing zones. TAT-fused cell-penetrating small molecular weight LPL peptide (TAT- MARGSVSDEE, denoted as an inhibitory LPL peptide) attenuated the formation of NSZs and impaired bone resorption in vitro in osteoclasts. Also, the genetic deletion of LPL in mice demonstrated decreased eroded perimeters and increased trabecular bone density. In the present study, we hypothesized that targeting LPL with the inhibitory LPL peptide in vivo could reduce osteoclast function and increase bone density in a mice model of low bone mass. We injected aging C57BL/6 female mice (36 weeks old) subcutaneously with the inhibitory and scrambled peptides of LPL for 14 weeks. Micro-CT and histomorphometry analyses demonstrated an increase in trabecular bone density of femoral and tibial bones with no change in cortical thickness in mice injected with the inhibitory LPL peptide. A reduction in the serum levels of CTX-1 peptide suggests that the increase in bone density is associated with a decrease in osteoclast function. No changes in bone formation rate and mineral apposition rate, and the serum levels of P1NP indicate that the inhibitory LPL peptide does not affect osteoblast function. Our study shows that the inhibitory LPL peptide can block osteoclast function without impairing the function of osteoblasts. LPL peptide could be developed as a prospective therapeutic agent to treat osteoporosis.
Collapse
Affiliation(s)
- Hanan Aljohani
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
- Department of Oral Medicine and Diagnostics Sciences, King Saud University, School of Dentistry, Riyadh, Kingdom of Saudi Arabia
| | - Joseph P Stains
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sunipa Majumdar
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Deepa Srinivasan
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Linda Senbanjo
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Meenakshi A Chellaiah
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
16
|
L-plastin Ser5 phosphorylation is modulated by the PI3K/SGK pathway and promotes breast cancer cell invasiveness. Cell Commun Signal 2021; 19:22. [PMID: 33618712 PMCID: PMC7898450 DOI: 10.1186/s12964-021-00710-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/18/2021] [Indexed: 01/15/2023] Open
Abstract
Background Metastasis is the predominant cause for cancer morbidity and mortality accounting for approximatively 90% of cancer deaths. The actin-bundling protein L-plastin has been proposed as a metastatic marker and phosphorylation on its residue Ser5 is known to increase its actin-bundling activity. We recently showed that activation of the ERK/MAPK signalling pathway leads to L-plastin Ser5 phosphorylation and that the downstream kinases RSK1 and RSK2 are able to directly phosphorylate Ser5. Here we investigate the involvement of the PI3K pathway in L-plastin Ser5 phosphorylation and the functional effect of this phosphorylation event in breast cancer cells. Methods To unravel the signal transduction network upstream of L-plastin Ser5 phosphorylation, we performed computational modelling based on immunoblot analysis data, followed by experimental validation through inhibition/overexpression studies and in vitro kinase assays. To assess the functional impact of L-plastin expression/Ser5 phosphorylation in breast cancer cells, we either silenced L-plastin in cell lines initially expressing endogenous L-plastin or neoexpressed L-plastin wild type and phosphovariants in cell lines devoid of endogenous L-plastin. The established cell lines were used for cell biology experiments and confocal microscopy analysis. Results Our modelling approach revealed that, in addition to the ERK/MAPK pathway and depending on the cellular context, the PI3K pathway contributes to L-plastin Ser5 phosphorylation through its downstream kinase SGK3. The results of the transwell invasion/migration assays showed that shRNA-mediated knockdown of L-plastin in BT-20 or HCC38 cells significantly reduced cell invasion, whereas stable expression of the phosphomimetic L-plastin Ser5Glu variant led to increased migration and invasion of BT-549 and MDA-MB-231 cells. Finally, confocal image analysis combined with zymography experiments and gelatin degradation assays provided evidence that L-plastin Ser5 phosphorylation promotes L-plastin recruitment to invadopodia, MMP-9 activity and concomitant extracellular matrix degradation. Conclusion Altogether, our results demonstrate that L-plastin Ser5 phosphorylation increases breast cancer cell invasiveness. Being a downstream molecule of both ERK/MAPK and PI3K/SGK pathways, L-plastin is proposed here as a potential target for therapeutic approaches that are aimed at blocking dysregulated signalling outcome of both pathways and, thus, at impairing cancer cell invasion and metastasis formation. Video abstract
Collapse
|
17
|
Bakke FK, Monte MM, Stead DA, Causey DR, Douglas A, Macqueen DJ, Dooley H. Plasma Proteome Responses in Salmonid Fish Following Immunization. Front Immunol 2020; 11:581070. [PMID: 33133099 PMCID: PMC7579410 DOI: 10.3389/fimmu.2020.581070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022] Open
Abstract
Vaccination plays a critical role in the protection of humans and other animals from infectious diseases. However, the same vaccine often confers different protection levels among individuals due to variation in genetics and/or immunological histories. While this represents a well-recognized issue in humans, it has received little attention in fish. Here we address this knowledge gap in a proteomic study of rainbow trout (Oncorhynchus mykiss, Walbaum), using non-lethal repeated blood sampling to establish the plasma protein response of individual fish following immunization. Six trout were immunized with adjuvanted hen egg-white lysozyme (HEL) and peripheral blood sampled at ten time points from day 0 to day 84 post-injection. We confirm that an antigen-specific antibody response to HEL was raised, showing differences in timing and magnitude among individuals. Using label-free liquid chromatography-mass spectrometry, we quantified the abundance of 278 plasma proteins across the timecourse. As part of the analysis, we show that this approach can distinguish many (but not all) duplicated plasma proteins encoded by paralogous genes retained from the salmonid-specific whole genome duplication event. Global variation in the plasma proteome was predominantly explained by individual differences among fish. However, sampling day explained a major component of variation in abundance for a statistically defined subset of 41 proteins, representing 15% of those detected. These proteins clustered into five groups showing distinct temporal responses to HEL immunization at the population level, and include classical immune (e.g. complement system members) and acute phase molecules (e.g. apolipoproteins, haptoglobins), several enzymes and other proteins supporting the immune response, in addition to evolutionarily conserved molecules that are as yet uncharacterized. Overall, this study improves our understanding of the fish plasma proteome, provides valuable marker proteins for different phases of the immune response, and has implications for vaccine development and the design of immune challenge experiments.
Collapse
Affiliation(s)
- Fiona K Bakke
- School of Biological Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Milena M Monte
- School of Biological Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - David A Stead
- Aberdeen Proteomics, The Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom
| | - Dwight R Causey
- School of Biological Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Alex Douglas
- School of Biological Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Daniel J Macqueen
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Helen Dooley
- Department of Microbiology and Immunology, Institute of Marine and Environmental Technology (IMET), University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
18
|
Bhatlekar S, Manne BK, Basak I, Edelstein LC, Tugolukova E, Stoller ML, Cody MJ, Morley SC, Nagalla S, Weyrich AS, Rowley JW, O'Connell RM, Rondina MT, Campbell RA, Bray PF. miR-125a-5p regulates megakaryocyte proplatelet formation via the actin-bundling protein L-plastin. Blood 2020; 136:1760-1772. [PMID: 32844999 PMCID: PMC7544541 DOI: 10.1182/blood.2020005230] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/24/2020] [Indexed: 12/17/2022] Open
Abstract
There is heritability to interindividual variation in platelet count, and better understanding of the regulating genetic factors may provide insights for thrombopoiesis. MicroRNAs (miRs) regulate gene expression in health and disease, and megakaryocytes (MKs) deficient in miRs have lower platelet counts, but information about the role of miRs in normal human MK and platelet production is limited. Using genome-wide miR profiling, we observed strong correlations among human bone marrow MKs, platelets, and differentiating cord blood-derived MK cultures, and identified MK miR-125a-5p as associated with human platelet number but not leukocyte or hemoglobin levels. Overexpression and knockdown studies showed that miR-125a-5p positively regulated human MK proplatelet (PP) formation in vitro. Inhibition of miR-125a-5p in vivo lowered murine platelet counts. Analyses of MK and platelet transcriptomes identified LCP1 as a miR-125a-5p target. LCP1 encodes the actin-bundling protein, L-plastin, not previously studied in MKs. We show that miR-125a-5p directly targets and reduces expression of MK L-plastin. Overexpression and knockdown studies show that L-plastin promotes MK progenitor migration, but negatively correlates with human platelet count and inhibits MK PP formation (PPF). This work provides the first evidence for the actin-bundling protein, L-plastin, as a regulator of human MK PPF via inhibition of the late-stage MK invagination system, podosome and PPF, and PP branching. We also provide resources of primary and differentiating MK transcriptomes and miRs associated with platelet counts. miR-125a-5p and L-plastin may be relevant targets for increasing in vitro platelet manufacturing and for managing quantitative platelet disorders.
Collapse
Affiliation(s)
- Seema Bhatlekar
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
| | - Bhanu K Manne
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
| | - Indranil Basak
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
| | - Leonard C Edelstein
- Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA
| | - Emilia Tugolukova
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
| | | | - Mark J Cody
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
| | - Sharon C Morley
- Division of Infectious Diseases, Department of Pediatrics and
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Srikanth Nagalla
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Andrew S Weyrich
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
- Division of Pulmonary, Department of Internal Medicine
| | - Jesse W Rowley
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
- Division of Pulmonary, Department of Internal Medicine
| | - Ryan M O'Connell
- Division of Microbiology and Immunology, Department of Pathology, and
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Matthew T Rondina
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
- Geriatric Research, Education and Clinical Center, George E. Wahlen VAMC GRECC, Salt Lake City, UT; and
- Division of General Internal Medicine and
| | - Robert A Campbell
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
- Division of General Internal Medicine and
| | - Paul F Bray
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
- Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, UT
| |
Collapse
|
19
|
Zeng Q, Li L, Feng Z, Luo L, Xiong J, Jie Z, Cao Y, Li Z. LCP1 is a prognostic biomarker correlated with immune infiltrates in gastric cancer. Cancer Biomark 2020; 30:105-125. [PMID: 32986657 DOI: 10.3233/cbm-200006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Previous studies have identified LCP1 as a diagnostic and prognostic marker in several cancers. However, the role of LCP1 in gastric cancer (GC) and its effect on tumor immune infiltration remain unclear. OBJECTIVE The aim was to explore the role of LCP1 in GC and its effect on tumor immune infiltration. METHODS We explored the expression of LCP1 relative to clinicopathology in GC patients by bioinformatics analysis and immunohistochemistry. Using cBioportal database, we analyzed the characteristic genetic variations of LCP1 in GC. In addition, we evaluated the correlation between LCP1 expression and tumor-infiltrating lymphocytes (TILs) using R software, TIMER and TISIDB databases. Finally, we analyzed the biological functions in which LCP1 may participate and the signaling pathways it may regulate. RESULTS Here, we showed that LCP1 expression is significantly correlated with tumor aggressiveness and poor prognosis in GC patients. Additionally, the results indicated that LCP1 was associated with TILs, including both immunosuppressive and immunosupportive cells, and was strongly correlated with various immune marker sets in GC. GSEA analysis demonstrated that LCP1 expression played an important role in lymphocyte formation and immune reaction. CONCLUSIONS LCP1 may be a potential prognostic biomarker for GC patients and a marker for tumor immunotherapy.
Collapse
Affiliation(s)
- Qingwen Zeng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China.,Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Leyan Li
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.,Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Zongfeng Feng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Lianghua Luo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Jianbo Xiong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Zhigang Jie
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Yi Cao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Zhengrong Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
20
|
Schaffner-Reckinger E, Machado RAC. The actin-bundling protein L-plastin-A double-edged sword: Beneficial for the immune response, maleficent in cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 355:109-154. [PMID: 32859369 DOI: 10.1016/bs.ircmb.2020.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The dynamic organization of the actin cytoskeleton into bundles and networks is orchestrated by a large variety of actin-binding proteins. Among them, the actin-bundling protein L-plastin is normally expressed in hematopoietic cells, where it is involved in the immune response. However, L-plastin is also often ectopically expressed in malignant cancer cells of non-hematopoietic origin and is even considered as a marker for cancer progression. Post-translational modification modulates L-plastin activity. In particular, L-plastin Ser5 phosphorylation has been shown to be important for the immune response in leukocytes as well as for invasion and metastasis formation of carcinoma cells. This chapter discusses the physiological and pathological role of L-plastin with a special focus on the importance of L-plastin Ser5 phosphorylation for the protein functions. The potential use of Ser5 phosphorylated L-plastin as a biomarker and/or therapeutic target will be evoked.
Collapse
Affiliation(s)
- Elisabeth Schaffner-Reckinger
- Cancer Cell Biology and Drug Discovery Group, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| | - Raquel A C Machado
- Cancer Cell Biology and Drug Discovery Group, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
21
|
Wang Y, Luo Y, Yao Y, Ji Y, Feng L, Du F, Zheng X, Tao T, Zhai X, Li Y, Han P, Xu B, Zhao H. Silencing the lncRNA Maclpil in pro-inflammatory macrophages attenuates acute experimental ischemic stroke via LCP1 in mice. J Cereb Blood Flow Metab 2020; 40:747-759. [PMID: 30895879 PMCID: PMC7168792 DOI: 10.1177/0271678x19836118] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Long noncoding RNAs (lncRNA) expression profiles change in the ischemic brain after stroke, but their roles in specific cell types after stroke have not been studied. We tested the hypothesis that lncRNA modulates brain injury by altering macrophage functions. Using RNA deep sequencing, we identified 73 lncRNAs that were differentially expressed in monocyte-derived macrophages (MoDMs) and microglia-derived macrophages (MiDMs) isolated in the ischemic brain three days after stroke. Among these, the lncRNA, GM15628, is highly expressed in pro-inflammatory MoDMs but not in MiDMs, and are functionally related to its neighbor gene, lymphocyte cytosolic protein 1 (LCP1), which plays a role in maintaining cell shape and cell migration. We termed this lncRNA as Macrophage contained LCP1 related pro-inflammatory lncRNA, Maclpil. Using cultured macrophages polarized by LPS, M(LPS), we found that downregulation of Maclpil in M(LPS) decreased pro-inflammatory gene expression while promoting anti-inflammatory gene expression. Maclpil inhibition also reduced the migration and phagocytosis ability of MoDMs by inhibiting LCP1. Furthermore, adoptive transfer of Maclpil silenced M(LPS), reduced ischemic brain infarction, improved behavioral performance and attenuated penetration of MoDMs in the ischemic hemisphere. We conclude that by blocking macrophage, Maclpil protects against acute ischemic stroke by inhibiting neuroinflammation.
Collapse
Affiliation(s)
- Yan Wang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ying Luo
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yang Yao
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuhua Ji
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Liangshu Feng
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Fang Du
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaoya Zheng
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Tao Tao
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Xuan Zhai
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yaning Li
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Pei Han
- Department of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Heng Zhao
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
22
|
Ma G, Gezer D, Herrmann O, Feldberg K, Schemionek M, Jawhar M, Reiter A, Brümmendorf TH, Koschmieder S, Chatain N. LCP1 triggers mTORC2/AKT activity and is pharmacologically targeted by enzastaurin in hypereosinophilia. Mol Carcinog 2019; 59:87-103. [DOI: 10.1002/mc.23131] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/14/2019] [Accepted: 10/21/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Guangxin Ma
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine RWTH Aachen University Aachen Germany
- Hematology and Oncology Unit, Department of Geriatrics Qilu Hospital of Shandong University Jinan Shandong China
| | - Deniz Gezer
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine RWTH Aachen University Aachen Germany
| | - Oliver Herrmann
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine RWTH Aachen University Aachen Germany
| | - Kristina Feldberg
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine RWTH Aachen University Aachen Germany
| | - Mirle Schemionek
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine RWTH Aachen University Aachen Germany
| | - Mohamad Jawhar
- Department of Hematology and Oncology University Medical Centre Mannheim, Heidelberg University Mannheim Germany
| | - Andreas Reiter
- Department of Hematology and Oncology University Medical Centre Mannheim, Heidelberg University Mannheim Germany
| | - Tim H. Brümmendorf
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine RWTH Aachen University Aachen Germany
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine RWTH Aachen University Aachen Germany
| | - Nicolas Chatain
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine RWTH Aachen University Aachen Germany
| |
Collapse
|
23
|
Spatial oxidation of L-plastin downmodulates actin-based functions of tumor cells. Nat Commun 2019; 10:4073. [PMID: 31501427 PMCID: PMC6733871 DOI: 10.1038/s41467-019-11909-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 08/06/2019] [Indexed: 01/15/2023] Open
Abstract
Several antitumor therapies work by increasing reactive oxygen species (ROS) within the tumor micromilieu. Here, we reveal that L-plastin (LPL), an established tumor marker, is reversibly regulated by ROS-induced thiol oxidation on Cys101, which forms a disulfide bridge with Cys42. LPL reduction is mediated by the Thioredoxin1 (TRX1) system, as shown by TRX1 trapping, TRX1 knockdown and blockade of Thioredoxin1 reductase (TRXR1) with auranofin. LPL oxidation diminishes its actin-bundling capacity. Ratiometric imaging using an LPL-roGFP-Orp1 fusion protein and a dimedone-based proximity ligation assay (PLA) reveal that LPL oxidation occurs primarily in actin-based cellular extrusions and strongly inhibits cell spreading and filopodial extension formation in tumor cells. This effect is accompanied by decreased tumor cell migration, invasion and extracellular matrix (ECM) degradation. Since LPL oxidation occurs following treatment of tumors with auranofin or γ-irradiation, it may be a molecular mechanism contributing to the effectiveness of tumor treatment with redox-altering therapies. The actin-remodelling protein L-plastin promotes tumour migration and invasion. Here, the authors show that L-plastin is regulated spatially by ROS-induced thiol oxidation which inhibits its actin-bundling function and cell spreading and filopodial extension formation in tumor cells.
Collapse
|
24
|
Takabayashi T, Tanaka Y, Susuki D, Yoshida K, Tomita K, Sakashita M, Imoto Y, Kato Y, Narita N, Nakayama T, Haruna S, Schleimer RP, Fujieda S. Increased expression of L-plastin in nasal polyp of patients with nonsteroidal anti-inflammatory drug-exacerbated respiratory disease. Allergy 2019; 74:1307-1316. [PMID: 30479022 DOI: 10.1111/all.13677] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 09/04/2018] [Accepted: 09/21/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Most patients with nonsteroidal anti-inflammatory drug-exacerbated respiratory disease (NERD) suffer from recurrence of nasal polyps. However, little is known about the specific cellular and molecular mechanisms contributing to the pathogenesis of nasal polyp development in patients with NERD in particular, especially at baseline when cyclooxygenase 1 inhibitors are not present. The objectives of this study were to identify proteins involved in the pathogenesis of nasal polyps in patients with NERD. METHODS We collected nasal polyp tissue from patients with NERD and from patients with aspirin-tolerant chronic rhinosinusitis with nasal polyps (CRSwNP). Protein profiles were analyzed by 2-dimensional electrophoresis and identified several proteins, including L-plastin, as highly expressed. We examined L-plastin and tissue factor (TF) expression by immunohistochemical and immunofluorescence analyses. To examine the role of L-plastin in eosinophils, we knocked down L-plastin expression in Eol-1 cells by using siRNA transfection. RESULTS L-plastin protein levels in nasal polyp tissue were increased in patients with NERD relative to those in patients with aspirin tolerant CRSwNP. Immunofluorescence analysis revealed that L-plastin was dominantly expressed in eosinophils and L-plastin and TF were co-expressed in eosinophils in NERD nasal polyp tissue. Knockdown of L-plastin in Eol-1 cells disrupted the cell surface distribution of TF by stimulation with granulocyte macrophage colony-stimulating factor. CONCLUSION Increased expression of L-plastin by eosinophils may contribute to abnormal fibrin deposition through TF translocation to the eosinophil cell surface in NERD nasal polyp tissue, which in turn may contribute to the pathogenesis of NERD.
Collapse
Affiliation(s)
- Tetsuji Takabayashi
- Division of Otorhinolaryngology Head and Neck Surgery Department of Sensory and Locomotor Medicine University of Fukui Fukui Japan
| | - Yukie Tanaka
- Department of Molecular Biology and Chemistry University of Fukui Fukui Japan
| | - Dai Susuki
- Division of Otorhinolaryngology Head and Neck Surgery Department of Sensory and Locomotor Medicine University of Fukui Fukui Japan
| | - Kanako Yoshida
- Division of Otorhinolaryngology Head and Neck Surgery Department of Sensory and Locomotor Medicine University of Fukui Fukui Japan
| | - Kaori Tomita
- Division of Otorhinolaryngology Head and Neck Surgery Department of Sensory and Locomotor Medicine University of Fukui Fukui Japan
| | - Masafumi Sakashita
- Division of Otorhinolaryngology Head and Neck Surgery Department of Sensory and Locomotor Medicine University of Fukui Fukui Japan
| | - Yoshimasa Imoto
- Division of Otorhinolaryngology Head and Neck Surgery Department of Sensory and Locomotor Medicine University of Fukui Fukui Japan
| | - Yukinori Kato
- Division of Otorhinolaryngology Head and Neck Surgery Department of Sensory and Locomotor Medicine University of Fukui Fukui Japan
| | - Norihiko Narita
- Division of Otorhinolaryngology Head and Neck Surgery Department of Sensory and Locomotor Medicine University of Fukui Fukui Japan
| | - Tsugihisa Nakayama
- Department of Otorhinolaryngology, Head and Neck Surgery Dokkyo Medical University Shimotsuga‐gun Japan
| | - Shinichi Haruna
- Department of Otorhinolaryngology, Head and Neck Surgery Dokkyo Medical University Shimotsuga‐gun Japan
| | - Robert P. Schleimer
- Division of Allergy and Immunology Department of Medicine Northwestern University Feinberg School of Medicine Chicago Illinois
| | - Shigeharu Fujieda
- Division of Otorhinolaryngology Head and Neck Surgery Department of Sensory and Locomotor Medicine University of Fukui Fukui Japan
| |
Collapse
|
25
|
Baumgartner EA, Compton ZJ, Evans S, Topczewski J, LeClair EE. Identification of regulatory elements recapitulating early expression of L-plastin in the zebrafish enveloping layer and embryonic periderm. Gene Expr Patterns 2019; 32:53-66. [PMID: 30940554 PMCID: PMC6655599 DOI: 10.1016/j.gep.2019.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/22/2019] [Accepted: 03/24/2019] [Indexed: 12/18/2022]
Abstract
We have cloned and characterized an intronic fragment of zebrafish lymphocyte cytosolic protein 1 (lcp1, also called L-plastin) that drives expression to the zebrafish enveloping layer (EVL). L-plastin is a calcium-dependent actin-bundling protein belonging to the plastin/fimbrin family of proteins, and is necessary for the proper migration and attachment of several adult cell types, including leukocytes and osteoclasts. However, in zebrafish lcp1 is abundantly expressed much earlier, during differentiation of the EVL. The cells of this epithelial layer migrate collectively, spreading vegetally over the yolk. L-plastin expression persists into the larval periderm, a transient epithelial tissue that forms the first larval skin. This finding establishes that L-plastin is activated in two different embryonic waves, with a distinct regulatory switch between the early EVL and the later leukocyte. To better study L-plastin expressing cells we attempted CRISPR/Cas9 homology-driven recombination (HDR) to insert a self-cleaving peptide (Cre-P2A-EGFP-CAAX) downstream of the native lcp1 promoter. This produced a stable zebrafish line expressing Cre recombinase in EVL nuclei and green fluorescence in EVL cell membranes. In vivo tracking of these labeled cells provided enhanced views of EVL migration behavior, membrane extensions, and mitotic events. Finally, we experimentally dissected key elements of the targeted lcp1 locus, discovering a ∼300 bp intronic sequence sufficient to drive EVL expression. The lcp1: Cre-P2A-EGFP-CAAX zebrafish should be useful for studying enveloping layer specification, gastrulation movements and periderm development in this widely used vertebrate model. In addition, the conserved regulatory sequences we have isolated predict that L-plastin orthologs may have a similar early expression pattern in other vertebrate embryos.
Collapse
Affiliation(s)
| | | | - Spencer Evans
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, USA
| | - Jacek Topczewski
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, USA; Department of Pediatrics, Northwestern University Feinberg School of Medicine, USA; Department of Biochemistry and Molecular Biology, Medical University of Lublin, Poland
| | | |
Collapse
|
26
|
Plasma-based protein biomarkers can predict the risk of acute graft-versus-host disease and non-relapse mortality in patients undergoing allogeneic hematopoietic stem cell transplantation. Blood Cells Mol Dis 2018; 74:5-12. [PMID: 30344086 DOI: 10.1016/j.bcmd.2018.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 10/01/2018] [Indexed: 12/19/2022]
Abstract
Predictive biomarkers for acute graft-versus-host disease (aGVHD) is currently lacking. In this study, we employed an unbiased proteome profiling method to prospectively collected plasma samples from allogeneic hematopoietic stem cell transplantation (alloHSCT) recipients to identify protein biomarkers that predict the risk of aGVHD and non-relapse mortality (NRM). In the discovery set, including five aGVHD patients and five controls, we identified seven candidate proteins. Patients with high levels of these proteins tended to exhibit a higher risk of aGVHD and NRM compared to patients with low levels in post-engraftment plasma samples from an independent validation set (n = 89). Tissue inhibitor of metalloproteinase 1, plastin-2, and regenerating islet-derived protein 3-α were selected as the most-predictive biomarkers via an exhaustive variable screening algorithm and were collectively used to develop a biomarker panel score ranging from 0 to 3. The biomarker panel score correlated significantly with aGVHD and NRM risk in univariable and multivariable Cox models. Furthermore, using the biomarker panel score in conjunction with clinical predictors significantly improved the discriminatory performance of the Cox model in predicting aGVHD and NRM risk. Our findings suggest that plasma-derived protein biomarkers can be used to predict aGVHD and NRM before the onset of clinical manifestations.
Collapse
|
27
|
Chellaiah MA, Majumdar S, Aljohani H. Peptidomimetic inhibitors of L-plastin reduce the resorptive activity of osteoclast but not the bone forming activity of osteoblasts in vitro. PLoS One 2018; 13:e0204209. [PMID: 30248139 PMCID: PMC6152981 DOI: 10.1371/journal.pone.0204209] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/05/2018] [Indexed: 12/19/2022] Open
Abstract
Sealing ring formation is a requirement for osteoclast function. We have recently identified the role of an actin-bundling protein L-plastin in the assembly of nascent sealing zones (NSZs) at the early phase of sealing ring formation in osteoclasts. TNF-α signaling regulates this actin assembly by the phosphorylation of L-plastin on serine -5 and -7 residues at the amino-terminal end. These NSZs function as a core for integrin localization and coordinating integrin signaling required for maturation into fully functional sealing rings. Our goal is to elucidate the essential function of L-plastin phosphorylation in actin bundling, a process required for NSZs formation. The present study was undertaken to determine whether targeting serine phosphorylation of cellular L-plastin would be the appropriate approach to attenuate the formation of NSZs. Our approach is to use TAT-fused small molecular weight amino-terminal L-plastin peptides (10 amino acids) containing phospho- Ser-5 and Ser-7. We used peptides unsubstituted (P1) and substituted (P2- P4) at serine-to-alanine residues. Immunoblotting, actin staining, and dentine resorption analyses were done to determine cellular L-plastin phosphorylation, NSZ or sealing ring formation, and osteoclast function, respectively. Immunoblotting for bone formation markers, Alizarin red staining and alkaline phosphatase activity assay have been done to determine the effect of peptides on the mineralization process mediated by osteoblasts. Transduction of unsubstituted (P1) and substituted peptides at either Serine 5 or Serine 7 with Alanine (P3 and P4) demonstrated variable inhibitory effects on the phosphorylation of cellular L-plastin protein. Peptide P1 reduces the following processes substantially: 1) cellular L-plastin phosphorylation; 2) formation of nascent sealing zones and sealing rings; 3) bone resorption. Substitution of both Serine-5 and -7 with Alanine (P2) had no effects on the inhibitory activities described above. Furthermore, either the L-plastin (P1-P5) or (P6) control peptides had a little or no impact on the a) assembly/disassembly of podosomes and migration of osteoclasts; b) mineralization process mediated by osteoblasts in vitro. Small molecular weight peptidomimetics of L-plastin inhibits bone resorption by osteoclasts via attenuation of NSZ and sealing ring formation but not bone formation by osteoblasts in vitro. The L-plastin may be a valuable therapeutic target to treat and prevent diseases associated with bone loss without affecting bone formation.
Collapse
Affiliation(s)
- Meenakshi A. Chellaiah
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, Maryland, United States of America
| | - Sunipa Majumdar
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, Maryland, United States of America
| | - Hanan Aljohani
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, Maryland, United States of America
| |
Collapse
|
28
|
Chellaiah MA, Ma T, Majumdar S. L-plastin phosphorylation regulates the early phase of sealing ring formation by actin bundling process in mouse osteoclasts. Exp Cell Res 2018; 372:73-82. [PMID: 30244178 DOI: 10.1016/j.yexcr.2018.09.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/05/2018] [Accepted: 09/19/2018] [Indexed: 10/28/2022]
Abstract
The process of sealing ring formation requires major actin filament reorganization. We previously demonstrated that an actin-bundling protein L-plastin has a role in the cross-linking of actin filaments into tight bundles and forms actin aggregates (denoted as nascent sealing zones). These nascent sealing zones mature into fully functional sealing rings. We have shown here that TNF-alpha signaling regulates the phosphorylation of serine-5 and -7 in L-plastin which increases the actin bundling capacity of L-plastin and hence the formation of nascent sealing zones in mouse osteoclasts. Using the TAT-mediated transduction method, we confirmed the role of L-plastin in nascent sealing zones formation at the early phase of the sealing ring assembly. Transduction of TAT-fused full-length L-plastin peptide significantly increases the number of nascent sealing zones and therefore sealing rings. But, transduction of amino-terminal L-plastin peptides consisting of the serine-5 and -7 reduces the formation of both nascent sealing zones and sealing rings. Therefore, bone resorption in vitro was reduced considerably. The decrease was associated with the selective inhibition of cellular L-plastin phosphorylation by the transduced peptides. Neither the formation of podosomes nor the migration was affected in these osteoclasts. Phosphorylation of L- plastin on serine 5 and -7 residues increases the F-actin bundling capacity. The significance of our studies stands on laying the groundwork for a better understanding of L-plastin as a potential regulator at the early phase of sealing ring formation and could be a new therapeutic target to treat bone loss.
Collapse
Affiliation(s)
- Meenakshi A Chellaiah
- Department of Oncology and Diagnostic Sciences Dental School, University of Maryland, Baltimore, Maryland.
| | - Tao Ma
- Department of Oncology and Diagnostic Sciences Dental School, University of Maryland, Baltimore, Maryland
| | - Sunipa Majumdar
- Department of Oncology and Diagnostic Sciences Dental School, University of Maryland, Baltimore, Maryland
| |
Collapse
|
29
|
Day JP, Whiteley E, Freeley M, Long A, Malacrida B, Kiely P, Baillie GS. RAB40C regulates RACK1 stability via the ubiquitin-proteasome system. Future Sci OA 2018; 4:FSO317. [PMID: 30112187 PMCID: PMC6088270 DOI: 10.4155/fsoa-2018-0022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/15/2018] [Indexed: 11/17/2022] Open
Abstract
AIM RACK1 is a multifunctional scaffolding protein that is expressed in many cellular compartments, orchestrating a number of signaling processes. RACK1 acts as a signaling hub to localize active enzymes to discrete locations; therefore tight control of RACK1 is vital to cellular homeostasis. Our aim was to identify the mechanisms responsible for RACK1 turnover and show that degradation is directed by the ubiquitin proteasome system. RESULTS Using siRNA screening, we identified RAB40C as the ubiquitin E3 ligase responsible for ubiquitination of RACK1, and that the action of RAB40C in controlling RACK1 levels is crucial to both cancer cell growth and migration of T cells. CONCLUSION Our data suggest that manipulation of RACK1 levels in this way may provide a novel strategy to explore RACK1 function.
Collapse
Affiliation(s)
- Jon P Day
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Ellanor Whiteley
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Michael Freeley
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College, Dublin, D08 W9RT, Ireland
| | - Aideen Long
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College, Dublin, D08 W9RT, Ireland
| | - Beatrice Malacrida
- Materials & Surface Science Institute & Health Research Institute, University of Limerick, Limerick, Ireland
| | - Patrick Kiely
- Materials & Surface Science Institute & Health Research Institute, University of Limerick, Limerick, Ireland
| | - George S Baillie
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
30
|
Stewart-Hutchinson PJ, Szasz TP, Jaeger ER, Onken MD, Cooper JA, Morley SC. Technical Advance: New in vitro method for assaying the migration of primary B cells using an endothelial monolayer as substrate. J Leukoc Biol 2017. [PMID: 28637896 DOI: 10.1189/jlb.1ta0117-008r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Migration of B cells supports their development and recruitment into functional niches. Therefore, defining factors that control B cell migration will lead to a better understanding of adaptive immunity. In vitro cell migration assays with B cells have been limited by poor adhesion of cells to glass coated with adhesion molecules. We have developed a technique using monolayers of endothelial cells as the substrate for B cell migration and used this technique to establish a robust in vitro assay for B cell migration. We use TNF-α to up-regulate surface expression of the adhesion molecule VCAM-1 on endothelial cells. The ligand VLA-4 is expressed on B cells, allowing them to interact with the endothelial monolayer and migrate on its surface. We tested our new method by examining the role of L-plastin (LPL), an F-actin-bundling protein, in B cell migration. LPL-deficient (LPL-/-) B cells displayed decreased speed and increased arrest coefficient compared with wild-type (WT) B cells, following chemokine stimulation. However, the confinement ratios for WT and LPL-/- B cells were similar. Thus, we demonstrate how the use of endothelial monolayers as a substrate will support future interrogation of molecular pathways essential to B cell migration.
Collapse
Affiliation(s)
- Phillip J Stewart-Hutchinson
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Taylor P Szasz
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Emily R Jaeger
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael D Onken
- Departments of Biochemistry and Molecular Biophysics and Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA; and
| | - John A Cooper
- Departments of Biochemistry and Molecular Biophysics and Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA; and
| | - Sharon Celeste Morley
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA; .,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
31
|
Ding WY, Ong HT, Hara Y, Wongsantichon J, Toyama Y, Robinson RC, Nédélec F, Zaidel-Bar R. Plastin increases cortical connectivity to facilitate robust polarization and timely cytokinesis. J Cell Biol 2017; 216:1371-1386. [PMID: 28400443 PMCID: PMC5412556 DOI: 10.1083/jcb.201603070] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 01/11/2017] [Accepted: 03/08/2017] [Indexed: 01/23/2023] Open
Abstract
The cell cortex is essential to maintain animal cell shape, and contractile forces generated within it by nonmuscle myosin II (NMY-2) drive cellular morphogenetic processes such as cytokinesis. The role of actin cross-linking proteins in cortical dynamics is still incompletely understood. Here, we show that the evolutionarily conserved actin bundling/cross-linking protein plastin is instrumental for the generation of potent cortical actomyosin contractility in the Caenorhabditis elegans zygote. PLST-1 was enriched in contractile structures and was required for effective coalescence of NMY-2 filaments into large contractile foci and for long-range coordinated contractility in the cortex. In the absence of PLST-1, polarization was compromised, cytokinesis was delayed or failed, and 50% of embryos died during development. Moreover, mathematical modeling showed that an optimal amount of bundling agents enhanced the ability of a network to contract. We propose that by increasing the connectivity of the F-actin meshwork, plastin enables the cortex to generate stronger and more coordinated forces to accomplish cellular morphogenesis.
Collapse
Affiliation(s)
- Wei Yung Ding
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Hui Ting Ong
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Yusuke Hara
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604, Singapore
| | - Jantana Wongsantichon
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology, and Research), Singapore 138673, Singapore
| | - Yusuke Toyama
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Robert C. Robinson
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology, and Research), Singapore 138673, Singapore
- Department of Biochemistry, National University of Singapore, Singapore 117597, Singapore
| | - François Nédélec
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Ronen Zaidel-Bar
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore 117583, Singapore
| |
Collapse
|
32
|
Wabnitz G, Balta E, Samstag Y. L-plastin regulates the stability of the immune synapse of naive and effector T-cells. Adv Biol Regul 2017; 63:107-114. [PMID: 27720134 DOI: 10.1016/j.jbior.2016.09.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 09/22/2016] [Accepted: 09/25/2016] [Indexed: 06/06/2023]
Abstract
T-cells need to be tightly regulated during their activation and effector phase to assure an appropriate defence against cancer or pathogens and - vice versa - to avoid autoimmune reactions. Regulatory signals are provided via the immune synapse between T-cells and antigen-presenting cells (APCs) or target cells. The stability and kinetics of immune synapse formation is critical for proper T-cell functions. It requires dynamic rearrangements of the actin cytoskeleton necessary for organized spatio-temporal redistribution of receptors and adhesion molecules. We identified glucocorticoid-sensitive phosphorylation of serine 5 on the actin-bundling protein L-plastin as one important signalling event for this regulation. Using imaging flow cytometry as well as confocal and super-resolution microscopy we showed that L-plastin relocalizes to the immune synapse upon antigen encounter, where it associates with the β2-subunit of LFA-1 (CD11a/CD18). Interfering with L-plastin expression or activation leads to a defective LFA-1 recruitment and unstable T-cell/APC contacts. Consequently, the lack of L-plastin diminishes T-cell activation, proliferation and proximal effector responses such as cytokine production. On the other hand, a pro-oxidative milieu leads to prolonged activation of L-plastin resulting in a stronger enrichment of LFA-1 in the cytolytic immune synapse. Concomitant stabilization of conjugates formed by cytotoxic T-cells (CTLs) and their target cells impairs the ability of CTLs to kill more than one target cells (serial killing), which de facto leads to a downregulation of T-cell cytotoxicity. Together, we demonstrate that activation and spacial distribution of L-plastin regulates the maturation and stability of activating and cytolytic immune synapses important for T-cell activation and effector functions.
Collapse
Affiliation(s)
- Guido Wabnitz
- Institute of Immunology, Section Molecular Immunology, Ruprecht-Karls-University Heidelberg, Im Neuenheimer Feld 305, D-69120 Heidelberg, Germany.
| | - Emre Balta
- Institute of Immunology, Section Molecular Immunology, Ruprecht-Karls-University Heidelberg, Im Neuenheimer Feld 305, D-69120 Heidelberg, Germany
| | - Yvonne Samstag
- Institute of Immunology, Section Molecular Immunology, Ruprecht-Karls-University Heidelberg, Im Neuenheimer Feld 305, D-69120 Heidelberg, Germany
| |
Collapse
|
33
|
|
34
|
Alveolar macrophage development in mice requires L-plastin for cellular localization in alveoli. Blood 2016; 128:2785-2796. [PMID: 27758872 DOI: 10.1182/blood-2016-03-705962] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 09/13/2016] [Indexed: 12/18/2022] Open
Abstract
Alveolar macrophages are lung-resident sentinel cells that develop perinatally and protect against pulmonary infection. Molecular mechanisms controlling alveolar macrophage generation have not been fully defined. Here, we show that the actin-bundling protein L-plastin (LPL) is required for the perinatal development of alveolar macrophages. Mice expressing a conditional allele of LPL (CD11c.Crepos-LPLfl/fl) exhibited significant reductions in alveolar macrophages and failed to effectively clear pulmonary pneumococcal infection, showing that immunodeficiency results from reduced alveolar macrophage numbers. We next identified the phase of alveolar macrophage development requiring LPL. In mice, fetal monocytes arrive in the lungs during a late fetal stage, maturing to alveolar macrophages through a prealveolar macrophage intermediate. LPL was required for the transition from prealveolar macrophages to mature alveolar macrophages. The transition from prealveolar macrophage to alveolar macrophage requires the upregulation of the transcription factor peroxisome proliferator-activated receptor-γ (PPAR-γ), which is induced by exposure to granulocyte-macrophage colony-stimulating factor (GM-CSF). Despite abundant lung GM-CSF and intact GM-CSF receptor signaling, PPAR-γ was not sufficiently upregulated in developing alveolar macrophages in LPL-/- pups, suggesting that precursor cells were not correctly localized to the alveoli, where GM-CSF is produced. We found that LPL supports 2 actin-based processes essential for correct localization of alveolar macrophage precursors: (1) transmigration into the alveoli, and (2) engraftment in the alveoli. We thus identify a molecular pathway governing neonatal alveolar macrophage development and show that genetic disruption of alveolar macrophage development results in immunodeficiency.
Collapse
|
35
|
Zhou JY, Szasz TP, Stewart-Hutchinson PJ, Sivapalan J, Todd EM, Deady LE, Cooper JA, Onken MD, Morley SC. L-Plastin promotes podosome longevity and supports macrophage motility. Mol Immunol 2016; 78:79-88. [PMID: 27614263 PMCID: PMC5205537 DOI: 10.1016/j.molimm.2016.08.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 08/12/2016] [Accepted: 08/22/2016] [Indexed: 10/24/2022]
Abstract
Elucidating the molecular regulation of macrophage migration is essential for understanding the pathophysiology of multiple human diseases, including host responses to infection and autoimmune disorders. Macrophage migration is supported by dynamic rearrangements of the actin cytoskeleton, with formation of actin-based structures such as podosomes and lamellipodia. Here we provide novel insights into the function of the actin-bundling protein l-plastin (LPL) in primary macrophages. We found that podosome stability is disrupted in primary resident peritoneal macrophages from LPL-/- mice. Live-cell imaging of F-actin using resident peritoneal macrophages from LifeACT-RFP+ mice demonstrated that loss of LPL led to decreased longevity of podosomes, without reducing the number of podosomes initiated. Additionally, macrophages from LPL-/- mice failed to elongate in response to chemotactic stimulation. These deficiencies in podosome stabilization and in macrophage elongation correlated with impaired macrophage transmigration in culture and decreased monocyte migration into murine peritoneum. Thus, we have identified a role for LPL in stabilizing long-lived podosomes and in enabling macrophage motility.
Collapse
Affiliation(s)
- Julie Y Zhou
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, 63110, United States
| | - Taylor P Szasz
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, 63110, United States
| | - Phillip J Stewart-Hutchinson
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, 63110, United States
| | - Janardan Sivapalan
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, 63110, United States
| | - Elizabeth M Todd
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, 63110, United States
| | - Lauren E Deady
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, 63110, United States
| | - John A Cooper
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, United States
| | - Michael D Onken
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, United States
| | - S Celeste Morley
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, 63110, United States; Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, 63110, United States.
| |
Collapse
|
36
|
Li D, Liu Y, Zhang X, Lv H, Pang W, Sun X, Gan LM, Hammock BD, Ai D, Zhu Y. Inhibition of soluble epoxide hydrolase alleviated atherosclerosis by reducing monocyte infiltration in Ldlr(-/-) mice. J Mol Cell Cardiol 2016; 98:128-37. [PMID: 27496380 DOI: 10.1016/j.yjmcc.2016.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 07/05/2016] [Accepted: 08/01/2016] [Indexed: 01/08/2023]
Abstract
RATIONALE Circulating monocytes play pivotal roles in chronic inflammatory diseases. Epoxyeicosatrienoic acids (EETs), metabolites of arachidonic acid, are known to have anti-inflammatory effects and are hydrolyzed by soluble epoxide hydrolase (sEH). OBJECTIVE We aimed to investigate the effect of sEH inhibition in atherogenesis. METHODS AND RESULTS Mice with low-density lipoprotein receptor deficiency (Ldlr(-/-)) with or without sEH inhibitor, and Ldlr/sEH double-knockout (DK) mice were fed a Western-type diet (WTD) for 6weeks to induce arteriosclerosis. Both sEH inhibition and gene depletion decreased the WTD-induced hyperlipidemia, plaque area and macrophage infiltration in mice arterial wall. Ly6C(hi) infiltration of monocytes remained similar in blood, spleen and bone marrow of DK mice, but was decreased in aortic lesions. To further assess the role of sEH or EETs in monocyte/macrophage infiltration in atherogenesis, we transplanted DK bone marrow into Ldlr(-/-) recipients, and then fed mice the WTD. Aortic lesions and Ly6C(hi) monocyte infiltration were reduced in mice with transplanted bone marrow of DK mice without diminishing the cholesterol level. Furthermore, sEH inhibition or gene depletion increased the ratio of EETs/DHETs and diminished the expression of P-selectin glycoprotein ligand 1 (PSGL-1) in mice peripheral-blood mononuclear cells. Monocyte adhesion to P-selectin and to tumor necrosis factor α-activated endothelial cells was also diminished by sEH inhibition. CONCLUSION sEH inhibition and gene depletion attenuated atherosclerosis in mice by decreasing the infiltration of monocytes into the artery wall. EET and PSGL-1 may play pivotal roles in monocyte/macrophage infiltration and atherogenesis.
Collapse
Affiliation(s)
- Dan Li
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - Yajin Liu
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Xu Zhang
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Huizhen Lv
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Wei Pang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - Xiaoli Sun
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - Li-Ming Gan
- Innovative Medicines & Early Development, AstraZeneca R&D, SE-431 83 Mölndal, Sweden
| | - Bruce D Hammock
- Department of Entomology and Nematology UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA 95616, USA
| | - Ding Ai
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Yi Zhu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China; Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
37
|
Xu X, Wang X, Todd EM, Jaeger ER, Vella JL, Mooren OL, Feng Y, Hu J, Cooper JA, Morley SC, Huang YH. Mst1 Kinase Regulates the Actin-Bundling Protein L-Plastin To Promote T Cell Migration. THE JOURNAL OF IMMUNOLOGY 2016; 197:1683-91. [PMID: 27465533 DOI: 10.4049/jimmunol.1600874] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/27/2016] [Indexed: 12/31/2022]
Abstract
Exploring the mechanisms controlling lymphocyte trafficking is essential for understanding the function of the immune system and the pathophysiology of immunodeficiencies. The mammalian Ste20-like kinase 1 (Mst1) has been identified as a critical signaling mediator of T cell migration, and loss of Mst1 results in immunodeficiency disease. Although Mst1 is known to support T cell migration through induction of cell polarization and lamellipodial formation, the downstream effectors of Mst1 are incompletely defined. Mice deficient for the actin-bundling protein L-plastin (LPL) have phenotypes similar to mice lacking Mst1, including decreased T cell polarization, lamellipodial formation, and cell migration. We therefore asked whether LPL functions downstream of Mst1. The regulatory N-terminal domain of LPL contains a consensus Mst1 phosphorylation site at Thr(89) We found that Mst1 can phosphorylate LPL in vitro and that Mst1 can interact with LPL in cells. Removal of the Mst1 phosphorylation site by mutating Thr(89) to Ala impaired localization of LPL to the actin-rich lamellipodia of T cells. Expression of the T89A LPL mutant failed to restore migration of LPL-deficient T cells in vitro. Furthermore, expression of T89A LPL in LPL-deficient hematopoietic cells, using bone marrow chimeras, failed to rescue the phenotype of decreased thymic egress. These results identify LPL as a key effector of Mst1 and establish a novel mechanism linking a signaling intermediate to an actin-binding protein critical to T cell migration.
Collapse
Affiliation(s)
- Xiaolu Xu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Xinxin Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Elizabeth M Todd
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110
| | - Emily R Jaeger
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Jennifer L Vella
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - Olivia L Mooren
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110; Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110; and
| | - Yunfeng Feng
- Department of Pathology, The Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - Jiancheng Hu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - John A Cooper
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110; Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110; and
| | - Sharon Celeste Morley
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110; Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110;
| | - Yina H Huang
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, NH 03756; Department of Pathology, The Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| |
Collapse
|
38
|
Dorris ER, Blackshields G, Sommerville G, Alhashemi M, Dias A, McEneaney V, Smyth P, O'Leary JJ, Sheils O. Pluripotency markers are differentially induced by MEK inhibition in thyroid and melanoma BRAFV600E cell lines. Cancer Biol Ther 2016; 17:526-42. [PMID: 26828826 PMCID: PMC4910922 DOI: 10.1080/15384047.2016.1139230] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Oncogenic mutations in BRAF are common in melanoma and thyroid carcinoma and drive constitutive activation of the MAPK pathway. Molecularly targeted therapies of this pathway improves survival compared to chemotherapy; however, responses tend to be short-lived as resistance invariably occursCell line models of melanoma and thyroid carcinoma, +/− BRAFV600E activating mutation, were treated with the MEK inhibitor PD0325901. Treated and naive samples were assayed for expression of key members of the MAPK pathway. Global microRNA expression profiling of naive and resistant cells was performed via next generation sequencingand indicated pluripotency pathways in resistance. Parental cell lines were progressed to holoclones to confirm the miRNA stemness profileMembers of the MIR302/373/374/520 family of embryonic stem cell specific cell cycle regulating (ESCC) microRNAs were identified as differentially expressed between resistant BRAFV600E melanoma and thyroid cell lines. Upregulated expression of gene and protein stemness markers, upregulated expression of MAPK pathway genes and downregulation of the ESCC MIR302 cluster in BRAFV600E melanoma indicated an increased stem-like phenotype in resistant BRAFV600E melanoma. Conversely, downregulated expression of gene and protein stemness markers, downregulated expression of MAPK pathway genes, upregulation of the ESCC MIR520 cluster, reeexpression of cell surface receptors, and induced differentiation-associated morphology in resistant BRAFV600E indicate a differentiated phenotype associated with MEK inhibitor resistance in BRAFV600E thyroid cellsThe differential patterns of resistance observed between BRAFV600E melanoma and thyroid cell lines may reflect tissue type or de novo differentiation, but could have significant impact on the response of primary and metastatic cells to MEK inhibitor treatment. This study provides a basis for the investigation of the cellular differentiation/self-renewal access and its role in resistance to MEK inhibition.
Collapse
Affiliation(s)
- Emma R Dorris
- a Department of Histopathology , Sir Patrick Dun Research Lab, Trinity College Dublin , Dublin , Ireland
| | - Gordon Blackshields
- a Department of Histopathology , Sir Patrick Dun Research Lab, Trinity College Dublin , Dublin , Ireland
| | - Gary Sommerville
- a Department of Histopathology , Sir Patrick Dun Research Lab, Trinity College Dublin , Dublin , Ireland
| | - Mohsen Alhashemi
- a Department of Histopathology , Sir Patrick Dun Research Lab, Trinity College Dublin , Dublin , Ireland
| | - Andrew Dias
- a Department of Histopathology , Sir Patrick Dun Research Lab, Trinity College Dublin , Dublin , Ireland
| | - Victoria McEneaney
- a Department of Histopathology , Sir Patrick Dun Research Lab, Trinity College Dublin , Dublin , Ireland
| | - Paul Smyth
- a Department of Histopathology , Sir Patrick Dun Research Lab, Trinity College Dublin , Dublin , Ireland
| | - John J O'Leary
- a Department of Histopathology , Sir Patrick Dun Research Lab, Trinity College Dublin , Dublin , Ireland
| | - Orla Sheils
- a Department of Histopathology , Sir Patrick Dun Research Lab, Trinity College Dublin , Dublin , Ireland
| |
Collapse
|
39
|
Harrison A, Dubois LG, St John-Williams L, Moseley MA, Hardison RL, Heimlich DR, Stoddard A, Kerschner JE, Justice SS, Thompson JW, Mason KM. Comprehensive Proteomic and Metabolomic Signatures of Nontypeable Haemophilus influenzae-Induced Acute Otitis Media Reveal Bacterial Aerobic Respiration in an Immunosuppressed Environment. Mol Cell Proteomics 2015; 15:1117-38. [PMID: 26711468 DOI: 10.1074/mcp.m115.052498] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Indexed: 12/31/2022] Open
Abstract
A thorough understanding of the molecular details of the interactions between bacteria and host are critical to ultimately prevent disease. Recent technological advances allow simultaneous analysis of host and bacterial protein and metabolic profiles from a single small tissue sample to provide insight into pathogenesis. We used the chinchilla model of human otitis media to determine, for the first time, the most expansive delineation of global changes in protein and metabolite profiles during an experimentally induced disease. After 48 h of infection with nontypeable Haemophilus influenzae, middle ear tissue lysates were analyzed by high-resolution quantitative two-dimensional liquid chromatography-tandem mass spectrometry. Dynamic changes in 105 chinchilla proteins and 66 metabolites define the early proteomic and metabolomic signature of otitis media. Our studies indicate that establishment of disease coincides with actin morphogenesis, suppression of inflammatory mediators, and bacterial aerobic respiration. We validated the observed increase in the actin-remodeling complex, Arp2/3, and experimentally showed a role for Arp2/3 in nontypeable Haemophilus influenzae invasion. Direct inhibition of actin branch morphology altered bacterial invasion into host epithelial cells, and is supportive of our efforts to use the information gathered to modify outcomes of disease. The twenty-eight nontypeable Haemophilus influenzae proteins identified participate in carbohydrate and amino acid metabolism, redox homeostasis, and include cell wall-associated metabolic proteins. Quantitative characterization of the molecular signatures of infection will redefine our understanding of host response driven developmental changes during pathogenesis. These data represent the first comprehensive study of host protein and metabolite profiles in vivo in response to infection and show the feasibility of extensive characterization of host protein profiles during disease. Identification of novel protein targets and metabolic biomarkers will advance development of therapeutic and diagnostic options for treatment of disease.
Collapse
Affiliation(s)
- Alistair Harrison
- From the ‡The Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205
| | - Laura G Dubois
- ‡‡Duke Proteomics and Metabolomics Core Facility, Duke Center for Genomic and Computational Biology, Duke University, Medical Center, Durham, North Carolina 27710
| | - Lisa St John-Williams
- ‡‡Duke Proteomics and Metabolomics Core Facility, Duke Center for Genomic and Computational Biology, Duke University, Medical Center, Durham, North Carolina 27710
| | - M Arthur Moseley
- ‡‡Duke Proteomics and Metabolomics Core Facility, Duke Center for Genomic and Computational Biology, Duke University, Medical Center, Durham, North Carolina 27710
| | - Rachael L Hardison
- From the ‡The Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205
| | - Derek R Heimlich
- From the ‡The Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205
| | | | - Joseph E Kerschner
- ‖Department of Otolaryngology and Communication Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin 53226; **Division of Pediatric Otolaryngology, Children's Hospital of Wisconsin, Milwaukee, Wisconsin 53226
| | - Sheryl S Justice
- From the ‡The Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205; §The Center for Microbial Interface Biology and Department of Pediatrics, The Ohio State University, Columbus, Ohio 43210
| | - J Will Thompson
- ‡‡Duke Proteomics and Metabolomics Core Facility, Duke Center for Genomic and Computational Biology, Duke University, Medical Center, Durham, North Carolina 27710
| | - Kevin M Mason
- From the ‡The Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205; §The Center for Microbial Interface Biology and Department of Pediatrics, The Ohio State University, Columbus, Ohio 43210;
| |
Collapse
|
40
|
Lommel MJ, Trairatphisan P, Gäbler K, Laurini C, Muller A, Kaoma T, Vallar L, Sauter T, Schaffner-Reckinger E. L-plastin Ser5 phosphorylation in breast cancer cells and in vitro is mediated by RSK downstream of the ERK/MAPK pathway. FASEB J 2015; 30:1218-33. [PMID: 26631483 DOI: 10.1096/fj.15-276311] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 11/16/2015] [Indexed: 12/20/2022]
Abstract
Deregulated cell migration and invasion are hallmarks of metastatic cancer cells. Phosphorylation on residue Ser5 of the actin-bundling protein L-plastin activates L-plastin and has been reported to be crucial for invasion and metastasis. Here, we investigate signal transduction leading to L-plastin Ser5 phosphorylation using 4 human breast cancer cell lines. Whole-genome microarray analysis comparing cell lines with different invasive capacities and corresponding variations in L-plastin Ser5 phosphorylation level revealed that genes of the ERK/MAPK pathway are differentially expressed. It is noteworthy that in vitro kinase assays showed that ERK/MAPK pathway downstream ribosomal protein S6 kinases α-1 (RSK1) and α-3 (RSK2) are able to directly phosphorylate L-plastin on Ser5. Small interfering RNA- or short hairpin RNA-mediated knockdown and activation/inhibition studies followed by immunoblot analysis and computational modeling confirmed that ribosomal S6 kinase (RSK) is an essential activator of L-plastin. Migration and invasion assays showed that RSK knockdown led to a decrease of up to 30% of migration and invasion of MDA-MB-435S cells. Although the presence of L-plastin was not necessary for migration/invasion of these cells, immunofluorescence assays illustrated RSK-dependent recruitment of Ser5-phosphorylated L-plastin to migratory structures. Altogether, we provide evidence that the ERK/MAPK pathway is involved in L-plastin Ser5 phosphorylation in breast cancer cells with RSK1 and RSK2 kinases able to directly phosphorylate L-plastin residue Ser5.
Collapse
Affiliation(s)
- Maiti J Lommel
- *Laboratory of Cytoskeleton and Cell Plasticity and Systems Biology Group, Life Sciences Research Unit, University of Luxembourg, Luxembourg City, Luxembourg; and Genomics Research Unit, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Panuwat Trairatphisan
- *Laboratory of Cytoskeleton and Cell Plasticity and Systems Biology Group, Life Sciences Research Unit, University of Luxembourg, Luxembourg City, Luxembourg; and Genomics Research Unit, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Karoline Gäbler
- *Laboratory of Cytoskeleton and Cell Plasticity and Systems Biology Group, Life Sciences Research Unit, University of Luxembourg, Luxembourg City, Luxembourg; and Genomics Research Unit, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Christina Laurini
- *Laboratory of Cytoskeleton and Cell Plasticity and Systems Biology Group, Life Sciences Research Unit, University of Luxembourg, Luxembourg City, Luxembourg; and Genomics Research Unit, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Arnaud Muller
- *Laboratory of Cytoskeleton and Cell Plasticity and Systems Biology Group, Life Sciences Research Unit, University of Luxembourg, Luxembourg City, Luxembourg; and Genomics Research Unit, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Tony Kaoma
- *Laboratory of Cytoskeleton and Cell Plasticity and Systems Biology Group, Life Sciences Research Unit, University of Luxembourg, Luxembourg City, Luxembourg; and Genomics Research Unit, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Laurent Vallar
- *Laboratory of Cytoskeleton and Cell Plasticity and Systems Biology Group, Life Sciences Research Unit, University of Luxembourg, Luxembourg City, Luxembourg; and Genomics Research Unit, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Thomas Sauter
- *Laboratory of Cytoskeleton and Cell Plasticity and Systems Biology Group, Life Sciences Research Unit, University of Luxembourg, Luxembourg City, Luxembourg; and Genomics Research Unit, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Elisabeth Schaffner-Reckinger
- *Laboratory of Cytoskeleton and Cell Plasticity and Systems Biology Group, Life Sciences Research Unit, University of Luxembourg, Luxembourg City, Luxembourg; and Genomics Research Unit, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| |
Collapse
|
41
|
Dupré L, Houmadi R, Tang C, Rey-Barroso J. T Lymphocyte Migration: An Action Movie Starring the Actin and Associated Actors. Front Immunol 2015; 6:586. [PMID: 26635800 PMCID: PMC4649030 DOI: 10.3389/fimmu.2015.00586] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 11/02/2015] [Indexed: 12/14/2022] Open
Abstract
The actin cytoskeleton is composed of a dynamic filament meshwork that builds the architecture of the cell to sustain its fundamental properties. This physical structure is characterized by a continuous remodeling, which allows cells to accomplish complex motility steps such as directed migration, crossing of biological barriers, and interaction with other cells. T lymphocytes excel in these motility steps to ensure their immune surveillance duties. In particular, actin cytoskeleton remodeling is a key to facilitate the journey of T lymphocytes through distinct tissue environments and to tune their stop and go behavior during the scanning of antigen-presenting cells. The molecular mechanisms controlling actin cytoskeleton remodeling during T lymphocyte motility have been only partially unraveled, since the function of many actin regulators has not yet been assessed in these cells. Our review aims to integrate the current knowledge into a comprehensive picture of how the actin cytoskeleton drives T lymphocyte migration. We will present the molecular actors that control actin cytoskeleton remodeling, as well as their role in the different T lymphocyte motile steps. We will also highlight which challenges remain to be addressed experimentally and which approaches appear promising to tackle them.
Collapse
Affiliation(s)
- Loïc Dupré
- INSERM, UMR 1043, Centre de Physiopathologie de Toulouse Purpan , Toulouse , France ; Université Toulouse III Paul-Sabatier , Toulouse , France ; CNRS, UMR 5282 , Toulouse , France
| | - Raïssa Houmadi
- INSERM, UMR 1043, Centre de Physiopathologie de Toulouse Purpan , Toulouse , France ; Université Toulouse III Paul-Sabatier , Toulouse , France ; CNRS, UMR 5282 , Toulouse , France
| | - Catherine Tang
- INSERM, UMR 1043, Centre de Physiopathologie de Toulouse Purpan , Toulouse , France ; Université Toulouse III Paul-Sabatier , Toulouse , France ; CNRS, UMR 5282 , Toulouse , France ; Master BIOTIN, Université Montpellier I , Montpellier , France
| | - Javier Rey-Barroso
- INSERM, UMR 1043, Centre de Physiopathologie de Toulouse Purpan , Toulouse , France ; Université Toulouse III Paul-Sabatier , Toulouse , France ; CNRS, UMR 5282 , Toulouse , France
| |
Collapse
|
42
|
Dubey M, Singh AK, Awasthi D, Nagarkoti S, Kumar S, Ali W, Chandra T, Kumar V, Barthwal MK, Jagavelu K, Sánchez-Gómez FJ, Lamas S, Dikshit M. L-Plastin S-glutathionylation promotes reduced binding to β-actin and affects neutrophil functions. Free Radic Biol Med 2015; 86:1-15. [PMID: 25881549 DOI: 10.1016/j.freeradbiomed.2015.04.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 03/11/2015] [Accepted: 04/03/2015] [Indexed: 01/16/2023]
Abstract
Posttranslational modifications (PTMs) of cytoskeleton proteins due to oxidative stress associated with several pathological conditions often lead to alterations in cell function. The current study evaluates the effect of nitric oxide (DETA-NO)-induced oxidative stress-related S-glutathionylation of cytoskeleton proteins in human PMNs. By using in vitro and genetic approaches, we showed that S-glutathionylation of L-plastin (LPL) and β-actin promotes reduced chemotaxis, polarization, bactericidal activity, and phagocytosis. We identified Cys-206, Cys-283, and Cys-460as S-thiolated residues in the β-actin-binding domain of LPL, where cys-460 had the maximum score. Site-directed mutagenesis of LPL Cys-460 further confirmed the role in the redox regulation of LPL. S-Thiolation diminished binding as well as the bundling activity of LPL. The presence of S-thiolated LPL was detected in neutrophils from both diabetic patients and db/db mice with impaired PMN functions. Thus, enhanced nitroxidative stress may results in LPL S-glutathionylation leading to impaired chemotaxis, polarization, and bactericidal activity of human PMNs, providing a mechanistic basis for their impaired functions in diabetes mellitus.
Collapse
Affiliation(s)
- Megha Dubey
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Abhishek K Singh
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Deepika Awasthi
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sheela Nagarkoti
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sachin Kumar
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children׳s Research Foundation, Cincinnati, OH 45229, USA
| | - Wahid Ali
- King George׳s Medical University, Lucknow, India
| | | | - Vikas Kumar
- Centre for Cellular and Molecular Platforms, National Centre for Biological Sciences (NCBS-TIFR), Bangalore, India
| | - Manoj K Barthwal
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | | | - Francisco J Sánchez-Gómez
- Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Campus Universidad Autónoma, Nicolás, Cabrera 1, E-28049, Madrid, Spain
| | - Santiago Lamas
- Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Campus Universidad Autónoma, Nicolás, Cabrera 1, E-28049, Madrid, Spain
| | - Madhu Dikshit
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India.
| |
Collapse
|
43
|
Protein kinase C: a regulator of cytoskeleton remodelling and T-cell migration. Biochem Soc Trans 2015; 42:1490-7. [PMID: 25399559 DOI: 10.1042/bst20140204] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Protein kinase C (PKC) is a family of ten serine/threonine kinases that have diverse roles in the signalling pathways regulating cellular proliferation, differentiation, apoptosis and immune responses. Elucidating roles for individual PKC isoforms in the immune responses of T-cells have long been a challenging prospect, because these cells are known to express nine of these isoforms. A variety of approaches including the use of knockout mice, overexpression of kinase-inactive mutants, cell-permeable peptides, pharmacological inhibitors and siRNAs have shown that PKCs regulate the production of inflammatory cytokines and the cytotoxic responses of various T-cell subsets. Central to the T-cell immune response is a requirement to migrate to various organs and tissues in search of pathogens and micro-organisms. T-cell migration is guided by specific sets of chemokines and integrin ligands that activate their cognate chemokine receptors and integrins on T-cells, resulting in remodelling of the cytoskeleton and the dynamic protrusive/contractile forces necessary for cell adhesion and motility. In the present article, we review the role of PKC in T-cell migration, with an emphasis on studies that have defined their roles in cytoskeletal remodelling, cell polarity and intracellular trafficking downstream of chemokine receptors and integrins.
Collapse
|
44
|
Freeley M, Derrick E, Dempsey E, Hoff A, Davies A, Leake D, Vermeulen A, Kelleher D, Long A. RNAi Screening with Self-Delivering, Synthetic siRNAs for Identification of Genes That Regulate Primary Human T Cell Migration. ACTA ACUST UNITED AC 2015; 20:943-56. [DOI: 10.1177/1087057115588288] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 05/01/2015] [Indexed: 02/04/2023]
Abstract
Screening of RNA interference (RNAi) libraries in primary T cells is labor-intensive and technically challenging because these cells are hard to transfect. Chemically modified, self-delivering small interfering RNAs (siRNAs) offer a solution to this problem, because they enter hard-to-transfect cell types without needing a delivery reagent and are available in library format for RNAi screening. In this study, we have screened a library of chemically modified, self-delivering siRNAs targeting the expression of 72 distinct genes in conjunction with an image-based high-content-analysis platform as a proof-of-principle strategy to identify genes involved in lymphocyte function-associated antigen-1 (LFA-1)-mediated migration in primary human T cells. Our library-screening strategy identified the small GTPase RhoA as being crucial for T cell polarization and migration in response to LFA-1 stimulation and other migratory ligands. We also demonstrate that multiple downstream assays can be performed within an individual RNAi screen and have used the remainder of the cells for additional assays, including cell viability and adhesion to ICAM-1 (the physiological ligand for LFA-1) in the absence or presence of the chemokine SDF-1α. This study therefore demonstrates the ease and benefits of conducting siRNA library screens in primary human T cells using self-delivering, chemically modified siRNAs, and it emphasizes the feasibility and potential of this approach for elucidating the signaling pathways that regulate T cell function.
Collapse
Affiliation(s)
- Michael Freeley
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, Dublin, Ireland
| | - Emily Derrick
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, Dublin, Ireland
| | - Eugene Dempsey
- School of Biomolecular & Biomedical Science, Conway Institute, University College Dublin, Dublin, Ireland
| | - Antje Hoff
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, Dublin, Ireland
| | - Anthony Davies
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, Dublin, Ireland
| | - Devin Leake
- Dharmacon, GE Healthcare, Lafayette, Colorado, USA
| | | | | | - Aideen Long
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
45
|
Megrelis L, Delon J. Rapid and robust analysis of cellular and molecular polarization induced by chemokine signaling. J Vis Exp 2014:52140. [PMID: 25548867 PMCID: PMC4396962 DOI: 10.3791/52140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Cells respond to chemokine stimulation by losing their round shape in a process called polarization, and by altering the subcellular localization of many proteins. Classic imaging techniques have been used to study these phenomena. However, they required the manual acquisition of many cells followed by time consuming quantification of the morphology and the co-localization of the staining of tens of cells. Here, a rapid and powerful method is described to study these phenomena on samples consisting of several thousands of cells using an imaging flow cytometry technology that combines the advantages of a microscope with those of a cytometer. Using T lymphocytes stimulated with CCL19 and staining for MHC Class I molecules and filamentous actin, a gating strategy is presented to measure simultaneously the degree of shape alterations and the extent of co-localization of markers that are affected by CCL19 signaling. Moreover, this gating strategy allowed us to observe the segregation of filamentous actin (at the front) and phosphorylated Ezrin-Radixin-Moesin (phospho-ERM) proteins (at the rear) in polarized T cells after CXCL12 stimulation. This technique was also useful to observe the blocking effect on polarization of two different elements: inhibition of actin polymerization by a pharmacological inhibitor and expression of mutants of the Par6/atypical PKC signaling pathway. Thus, evidence is shown that this technique is useful to analyze both morphological alterations and protein redistributions.
Collapse
Affiliation(s)
- Laura Megrelis
- Inserm U1016, Institut Cochin; Cnrs, UMR8104; Université Paris Descartes, Sorbonne Paris Cité
| | - Jérôme Delon
- Inserm U1016, Institut Cochin; Cnrs, UMR8104; Université Paris Descartes, Sorbonne Paris Cité;
| |
Collapse
|
46
|
Verma NK, Kelleher D. Adaptor regulation of LFA-1 signaling in T lymphocyte migration: Potential druggable targets for immunotherapies? Eur J Immunol 2014; 44:3484-99. [PMID: 25251823 DOI: 10.1002/eji.201344428] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 09/16/2014] [Accepted: 09/22/2014] [Indexed: 01/24/2023]
Abstract
The integrin lymphocyte function associated antigen-1 (LFA-1) plays a key role in leukocyte trafficking and in adaptive immune responses through interactions with adhesive ligands, such as ICAM-1. Specific blockade of these interactions has validated LFA-1 as a therapeutic target in many chronic inflammatory diseases, however LFA-1 antagonists have not been clinically successful due to the development of a general immunosuppression, causing fatal side effects. Growing evidence has now established that LFA-1 mediates an array of intracellular signaling pathways by triggering a number of downstream molecules. In this context, a class of multimodular domain-containing proteins capable of recruiting two or more effector molecules, collectively known as "adaptor proteins," has emerged as important mediators in LFA-1 signal transduction. Here, we provide an overview of the adaptor proteins involved in the intracellular signaling cascades by which LFA-1 regulates T-cell motility and immune responses. The complexity of the LFA-1-associated signaling delineated in this review suggests that it may be an important and challenging focus for future research, enabling the identification of "tunable" targets for the development of immunotherapies.
Collapse
Affiliation(s)
- Navin K Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore; Singapore Eye Research Institute, Singapore, Singapore
| | | |
Collapse
|
47
|
Nishida Y, Aida K, Kihara M, Kobayashi T. Antibody-validated proteins in inflamed islets of fulminant type 1 diabetes profiled by laser-capture microdissection followed by mass spectrometry. PLoS One 2014; 9:e107664. [PMID: 25329145 PMCID: PMC4199548 DOI: 10.1371/journal.pone.0107664] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 08/13/2014] [Indexed: 01/05/2023] Open
Abstract
Background There are no reports of proteomic analyses of inflamed islets in type 1 diabetes. Procedures Proteins expressed in the islets of enterovirus-associated fulminant type 1 diabetes (FT1DM) with extensive insulitis were identified by laser-capture microdissection mass spectrometry using formalin-fixed paraffin-embedded pancreatic tissues. Results Thirty-eight proteins were identified solely in FT1DM islets, most of which have not been previously linked to type 1 diabetes. Five protein-protein interacting clusters were identified, and the cellular localization of selected proteins was validated immunohistochemically. Migratory activity-related proteins, including plastin-2 (LCP1), moesin (MSN), lamin-B1 (LMNB1), Ras GTPase-activating-like protein (IQGAP1) and others, were identified in CD8+ T cells and CD68+ macrophages infiltrated to inflamed FT1DM islets. Proteins involved in successive signaling in innate/adaptive immunity were identified, including SAM domain and HD domain-containing protein 1 (SAMHD1), Ras GTPase-activating-like protein (IQGAP1), proteasome activator complex subunit 1 (PSME1), HLA class I histocompatibility antigen (HLA-C), and signal transducer and activator of transcription 1-alpha/beta (STAT1). Angiogenic (thymidine phosphorylase (TYMP)) and anti-angiogenic (tryptophan-tRNA ligase (WARS)) factors were identified in migrating CD8+ T cells and CD68+ macrophages. Proteins related to virus replication and cell proliferation, including probable ATP-dependent RNA helicase DEAD box helicase 5 (DDX5) and heterogeneous nuclear ribonucleoprotein H (HNRNPH1), were identified. The anti-apoptotic protein T-complex protein 1 subunit epsilon (CCT5), the anti-oxidative enzyme 6-phosphogluconate dehydrogenase (PDG), and the anti-viral and anti-apoptotic proteins serpin B6 (SERPINB6) and heat shock 70 kDa protein1-like (HSPA1L), were identified in FT1DM-affected islet cells. Conclusion The identified FT1DM-characterizing proteins include those involved in aggressive beta cell destruction through massive immune cell migration and proteins involved in angiogenesis and islet vasculature bleeding, cell repair, and anti-inflammatory processes. Several target proteins for future type 1 diabetes interventions were identified.
Collapse
Affiliation(s)
- Yoriko Nishida
- Department of Nursing, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Kaoru Aida
- Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Makoto Kihara
- Medical ProteoScope Co., Ltd., Yokohama, Kanagawa, Japan
| | - Tetsuro Kobayashi
- Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
- Okinaka Memorial Institute for Medical Research, Tokyo, Japan
- * E-mail:
| |
Collapse
|
48
|
Mactier S, Kaufman KL, Wang P, Crossett B, Pupo GM, Kohnke PL, Thompson JF, Scolyer RA, Yang JY, Mann GJ, Christopherson RI. Protein signatures correspond to survival outcomes of AJCC stage III melanoma patients. Pigment Cell Melanoma Res 2014; 27:1106-16. [PMID: 24995518 PMCID: PMC4285183 DOI: 10.1111/pcmr.12290] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 07/02/2014] [Indexed: 11/28/2022]
Abstract
Summary Outcomes for melanoma patients with stage III disease differ widely even within the same subcategory. Molecular signatures that more accurately predict prognosis are needed to stratify patients according to risk. Proteomic analyses were used to identify differentially abundant proteins in extracts of surgically excised samples from patients with stage IIIc melanoma lymph node metastases. Analysis of samples from patients with poor (n = 14, <1 yr) and good (n = 19, >4 yr) survival outcomes identified 84 proteins that were differentially abundant between prognostic groups. Subsequent selected reaction monitoring analysis verified 21 proteins as potential biomarkers for survival. Poor prognosis patients are characterized by increased levels of proteins involved in protein metabolism, nucleic acid metabolism, angiogenesis, deregulation of cellular energetics and methylation processes, and decreased levels of proteins involved in apoptosis and immune response. These proteins are able to classify stage IIIc patients into prognostic subgroups (P < 0.02). This is the first report of potential prognostic markers from stage III melanoma using proteomic analyses. Validation of these protein markers in larger patient cohorts should define protein signatures that enable better stratification of stage III melanoma patients.
Collapse
Affiliation(s)
- Swetlana Mactier
- School of Molecular Bioscience, University of Sydney, Sydney, NSW, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Öztürk VÖ, Emingil G, Osterwalder V, Bostanci N. The actin-bundling protein L-plastin: a novel local inflammatory marker associated with periodontitis. J Periodontal Res 2014; 50:337-46. [DOI: 10.1111/jre.12212] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2014] [Indexed: 12/11/2022]
Affiliation(s)
- V. Ö. Öztürk
- Oral Translational Research; Institute of Oral Biology; Center of Dental Medicine; University of Zürich; Zürich Switzerland
- Department of Periodontology; School of Dentistry; Ege University; İzmir Turkey
| | - G. Emingil
- Department of Periodontology; School of Dentistry; Ege University; İzmir Turkey
| | - V. Osterwalder
- Oral Translational Research; Institute of Oral Biology; Center of Dental Medicine; University of Zürich; Zürich Switzerland
| | - N. Bostanci
- Oral Translational Research; Institute of Oral Biology; Center of Dental Medicine; University of Zürich; Zürich Switzerland
| |
Collapse
|
50
|
The landscape of protein biomarkers proposed for periodontal disease: markers with functional meaning. BIOMED RESEARCH INTERNATIONAL 2014; 2014:569632. [PMID: 25057495 PMCID: PMC4099050 DOI: 10.1155/2014/569632] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 04/07/2014] [Indexed: 12/12/2022]
Abstract
Periodontal disease (PD) is characterized by a deregulated inflammatory response which fails to resolve, activating bone resorption. The identification of the proteomes associated with PD has fuelled biomarker proposals; nevertheless, many questions remain. Biomarker selection should favour molecules representing an event which occurs throughout the disease progress. The analysis of proteome results and the information available for each protein, including its functional role, was accomplished using the OralOme database. The integrated analysis of this information ascertains if the suggested proteins reflect the cell and/or molecular mechanisms underlying the different forms of periodontal disease. The evaluation of the proteins present/absent or with very different concentrations in the proteome of each disease state was used for the identification of the mechanisms shared by different PD variants or specific to such state. The information presented is relevant for the adequate design of biomarker panels for PD. Furthermore, it will open new perspectives and help envisage future studies targeted to unveil the functional role of specific proteins and help clarify the deregulation process in the PD inflammatory response.
Collapse
|