1
|
Chan M, Kang Y, Osborne S, Zager M, Gujral TS. A kinase to cytokine explorer to identify molecular regulators and potential therapeutic opportunities. eLife 2024; 12:RP91472. [PMID: 38305363 PMCID: PMC10945549 DOI: 10.7554/elife.91472] [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] [Indexed: 02/03/2024] Open
Abstract
Cytokines and chemokines are secreted proteins that regulate various biological processes, such as inflammation, immune response, and cell differentiation. Therefore, disruption of signaling pathways involving these proteins has been linked to a range of diseases, including cancer. However, targeting individual cytokines, chemokines, or their receptors is challenging due to their regulatory redundancy and incomplete understanding of their signaling networks. To transform these difficult-to-drug targets into a pharmacologically manageable class, we developed a web-based platform called KinCytE. This platform was designed to link the effects of kinase inhibitors, a well-established class of drugs, with cytokine and chemokine release and signaling networks. The resulting KinCytE platform enables users to investigate protein kinases that regulate specific cytokines or chemokines, generate a ranked list of FDA-approved kinase inhibitors that affect cytokine/chemokine activity, and explore and visualize cytokine signaling network thus facilitating drugging this challenging target class. KinCytE is freely accessible via https://atlas.fredhutch.org/kincyte.
Collapse
Affiliation(s)
- Marina Chan
- Human Biology Division, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Yuqi Kang
- Human Biology Division, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Shannon Osborne
- Data Visualization Core, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Michael Zager
- Data Visualization Core, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Taranjit S Gujral
- Human Biology Division, Fred Hutchinson Cancer CenterSeattleUnited States
- Department of Pharmacology, University of WashingtonSeattleUnited States
| |
Collapse
|
2
|
Sun J, Ruiz Daniels R, Balic A, Andresen AMS, Bjørgen H, Dobie R, Henderson NC, Koppang EO, Martin SAM, Fosse JH, Taylor RS, Macqueen DJ. Cell atlas of the Atlantic salmon spleen reveals immune cell heterogeneity and cell-specific responses to bacterial infection. FISH & SHELLFISH IMMUNOLOGY 2024; 145:109358. [PMID: 38176627 DOI: 10.1016/j.fsi.2024.109358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/23/2023] [Accepted: 01/02/2024] [Indexed: 01/06/2024]
Abstract
The spleen is a conserved secondary lymphoid organ that emerged in parallel to adaptive immunity in early jawed vertebrates. Recent studies have applied single cell transcriptomics to reveal the cellular composition of spleen in several species, cataloguing diverse immune cell types and subpopulations. In this study, 51,119 spleen nuclei transcriptomes were comprehensively investigated in the commercially important teleost Atlantic salmon (Salmo salar L.), contrasting control animals with those challenged with the bacterial pathogen Aeromonas salmonicida. We identified clusters of nuclei representing the expected major cell types, namely T cells, B cells, natural killer-like cells, granulocytes, mononuclear phagocytes, endothelial cells, mesenchymal cells, erythrocytes and thrombocytes. We discovered heterogeneity within several immune lineages, providing evidence for resident macrophages and melanomacrophages, infiltrating monocytes, several candidate dendritic cell subpopulations, and B cells at distinct stages of differentiation, including plasma cells and an igt + subset. We provide evidence for twelve candidate T cell subsets, including cd4+ T helper and regulatory T cells, one cd8+ subset, three γδT subsets, and populations double negative for cd4 and cd8. The number of genes showing differential expression during the early stages of Aeromonas infection was highly variable across immune cell types, with the largest changes observed in macrophages and infiltrating monocytes, followed by resting mature B cells. Our analysis provides evidence for a local inflammatory response to infection alongside B cell maturation in the spleen, and upregulation of ccr9 genes in igt + B cells, T helper and cd8+ cells, and monocytes, consistent with the recruitment of immune cell populations to the gut to deal with Aeromonas infection. Overall, this study provides a new cell-resolved perspective of the immune actions of Atlantic salmon spleen, highlighting extensive heterogeneity hidden to bulk transcriptomics. We further provide a large catalogue of cell-specific marker genes that can be leveraged to further explore the function and structural organization of the salmonid immune system.
Collapse
Affiliation(s)
- Jianxuan Sun
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | - Rose Ruiz Daniels
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | - Adam Balic
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK; Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | | | - Håvard Bjørgen
- Unit of Anatomy, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Ross Dobie
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK; MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Erling Olaf Koppang
- Unit of Anatomy, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Samuel A M Martin
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | | | - Richard S Taylor
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | - Daniel J Macqueen
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK.
| |
Collapse
|
3
|
Laight BJ, Jawa NA, Tyryshkin K, Maslove DM, Boyd JG, Greer PA. Establishing the role of the FES tyrosine kinase in the pathogenesis, pathophysiology, and severity of sepsis and its outcomes. Front Immunol 2023; 14:1145826. [PMID: 37122758 PMCID: PMC10140553 DOI: 10.3389/fimmu.2023.1145826] [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: 01/16/2023] [Accepted: 04/03/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction Sepsis is a result of initial over-activation of the immune system in response to an infection or trauma that results in reduced blood flow and life-threatening end-organ damage, followed by suppression of the immune system that prevents proper clearance of the infection or trauma. Because of this, therapies that not only limit the activation of the immune system early on, but also improve blood flow to crucial organs and reactivate the immune system in late-stage sepsis, may be effective treatments. The tyrosine kinase FES may fulfill this role. FES is present in immune cells and serves to limit immune system activation. We hypothesize that by enhancing FES in early sepsis and inhibiting its effects in late sepsis, the severity and outcome of septic illness can be improved. Methods and analysis In vitro and in vivo modeling will be performed to determine the degree of inflammatory signaling, cytokine production, and neutrophil extracellular trap (NET) formation that occurs in wild-type (WT) and FES knockout (FES-/- ) mice. Clinically available treatments known to enhance or inhibit FES expression (lorlatinib and decitabine, respectively), will be used to explore the impact of early vs. late FES modulation on outcomes in WT mice. Bioinformatic analysis will be performed to examine FES expression levels in RNA transcriptomic data from sepsis patient cohorts, and correlate FES expression data with clinical outcomes (diagnosis of sepsis, illness severity, hospital length-of-stay). Ethics and dissemination Ethics approval pending from the Queen's University Health Sciences & Affiliated Teaching Hospitals Research Ethics Board. Results will be disseminated through scientific publications and through lay summaries to patients and families.
Collapse
Affiliation(s)
- Brian J. Laight
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, Queen’s University, Kingston, Ontario, ON, Canada
- School of Medicine, Faculty of Health Sciences, Queen’s University, Kingston, Ontario, ON, Canada
- Queen’s Cancer Research Institute, Queen’s University, Kingston, Ontario, ON, Canada
| | - Natasha A. Jawa
- School of Medicine, Faculty of Health Sciences, Queen’s University, Kingston, Ontario, ON, Canada
- Centre for Neuroscience Studies, Faculty of Health Sciences, Queen’s University, Kingston, Ontario, ON, Canada
| | - Kathrin Tyryshkin
- School of Computing, Queen’s University, Kingston, Ontario, ON, Canada
| | - David M. Maslove
- Division of Medicine and Critical Care Medicine, Department of Medicine, Faculty of Health Sciences, Queen’s University, Kingston, Ontario, ON, Canada
- Departments of Medicine and Critical Care Medicine, Kingston General Hospital, Kingston, Ontario, ON, Canada
| | - J. Gordon Boyd
- Centre for Neuroscience Studies, Faculty of Health Sciences, Queen’s University, Kingston, Ontario, ON, Canada
- Division of Medicine and Critical Care Medicine, Department of Medicine, Faculty of Health Sciences, Queen’s University, Kingston, Ontario, ON, Canada
- Departments of Medicine and Critical Care Medicine, Kingston General Hospital, Kingston, Ontario, ON, Canada
| | - Peter A. Greer
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, Queen’s University, Kingston, Ontario, ON, Canada
- Queen’s Cancer Research Institute, Queen’s University, Kingston, Ontario, ON, Canada
| |
Collapse
|
4
|
Karamanavi E, McVey DG, van der Laan SW, Stanczyk PJ, Morris GE, Wang Y, Yang W, Chan K, Poston RN, Luo J, Zhou X, Gong P, Jones PD, Cao J, Kostogrys RB, Webb TR, Pasterkamp G, Yu H, Xiao Q, Greer PA, Stringer EJ, Samani NJ, Ye S. The FES Gene at the 15q26 Coronary-Artery-Disease Locus Inhibits Atherosclerosis. Circ Res 2022; 131:1004-1017. [PMID: 36321446 PMCID: PMC9770135 DOI: 10.1161/circresaha.122.321146] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 10/16/2022] [Accepted: 10/17/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND Genome-wide association studies have discovered a link between genetic variants on human chromosome 15q26.1 and increased coronary artery disease (CAD) susceptibility; however, the underlying pathobiological mechanism is unclear. This genetic locus contains the FES (FES proto-oncogene, tyrosine kinase) gene encoding a cytoplasmic protein-tyrosine kinase involved in the regulation of cell behavior. We investigated the effect of the 15q26.1 variants on FES expression and whether FES plays a role in atherosclerosis. METHODS AND RESULTS Analyses of isogenic monocytic cell lines generated by CRISPR (clustered regularly interspaced short palindromic repeats)-mediated genome editing showed that monocytes with an engineered 15q26.1 CAD risk genotype had reduced FES expression. Small-interfering-RNA-mediated knockdown of FES promoted migration of monocytes and vascular smooth muscle cells. A phosphoproteomics analysis showed that FES knockdown altered phosphorylation of a number of proteins known to regulate cell migration. Single-cell RNA-sequencing revealed that in human atherosclerotic plaques, cells that expressed FES were predominately monocytes/macrophages, although several other cell types including smooth muscle cells also expressed FES. There was an association between the 15q26.1 CAD risk genotype and greater numbers of monocytes/macrophage in human atherosclerotic plaques. An animal model study demonstrated that Fes knockout increased atherosclerotic plaque size and within-plaque content of monocytes/macrophages and smooth muscle cells, in apolipoprotein E-deficient mice fed a high fat diet. CONCLUSIONS We provide substantial evidence that the CAD risk variants at the 15q26.1 locus reduce FES expression in monocytes and that FES depletion results in larger atherosclerotic plaques with more monocytes/macrophages and smooth muscle cells. This study is the first demonstration that FES plays a protective role against atherosclerosis and suggests that enhancing FES activity could be a potentially novel therapeutic approach for CAD intervention.
Collapse
Affiliation(s)
- Elisavet Karamanavi
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
| | - David G. McVey
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
| | - Sander W. van der Laan
- Central Diagnostic Laboratory, University of Utrecht, The Netherlands (S.W.v.d.L., G.P.)
| | - Paulina J. Stanczyk
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
| | - Gavin E. Morris
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
| | - Yifan Wang
- Cardiovascular Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (Y.W., H.Y., S.Y.)
| | - Wei Yang
- Shantou University Medical College, China (W.Y., J.C., S.Y.)
| | - Kenneth Chan
- William Harvey Research Institute, Queen Mary University of London, United Kingdom (K.C., R.N.P., J.L., X.Z., Q.X.)
| | - Robin N. Poston
- William Harvey Research Institute, Queen Mary University of London, United Kingdom (K.C., R.N.P., J.L., X.Z., Q.X.)
| | - Jun Luo
- William Harvey Research Institute, Queen Mary University of London, United Kingdom (K.C., R.N.P., J.L., X.Z., Q.X.)
| | - Xinmiao Zhou
- William Harvey Research Institute, Queen Mary University of London, United Kingdom (K.C., R.N.P., J.L., X.Z., Q.X.)
| | - Peng Gong
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
| | - Peter D. Jones
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
| | - Junjun Cao
- Shantou University Medical College, China (W.Y., J.C., S.Y.)
| | - Renata B. Kostogrys
- Department of Human Nutrition, University of Agriculture in Kraków, Poland (R.B.K.)
| | - Tom R. Webb
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
| | - Gerard Pasterkamp
- Central Diagnostic Laboratory, University of Utrecht, The Netherlands (S.W.v.d.L., G.P.)
| | - Haojie Yu
- Cardiovascular Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (Y.W., H.Y., S.Y.)
| | - Qingzhong Xiao
- William Harvey Research Institute, Queen Mary University of London, United Kingdom (K.C., R.N.P., J.L., X.Z., Q.X.)
| | - Peter A. Greer
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, Canada (P.A.G.)
| | - Emma J. Stringer
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
| | - Nilesh J. Samani
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
| | - Shu Ye
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
- Cardiovascular Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (Y.W., H.Y., S.Y.)
- Shantou University Medical College, China (W.Y., J.C., S.Y.)
| |
Collapse
|
5
|
Chemical genetics strategy to profile kinase target engagement reveals role of FES in neutrophil phagocytosis. Nat Commun 2020; 11:3216. [PMID: 32587248 PMCID: PMC7316778 DOI: 10.1038/s41467-020-17027-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 06/05/2020] [Indexed: 02/07/2023] Open
Abstract
Chemical tools to monitor drug-target engagement of endogenously expressed protein kinases are highly desirable for preclinical target validation in drug discovery. Here, we describe a chemical genetics strategy to selectively study target engagement of endogenous kinases. By substituting a serine residue into cysteine at the DFG-1 position in the ATP-binding pocket, we sensitize the non-receptor tyrosine kinase FES towards covalent labeling by a complementary fluorescent chemical probe. This mutation is introduced in the endogenous FES gene of HL-60 cells using CRISPR/Cas9 gene editing. Leveraging the temporal and acute control offered by our strategy, we show that FES activity is dispensable for differentiation of HL-60 cells towards macrophages. Instead, FES plays a key role in neutrophil phagocytosis via SYK kinase activation. This chemical genetics strategy holds promise as a target validation method for kinases.
Collapse
|
6
|
Haque N, Ouda R, Chen C, Ozato K, Hogg JR. ZFR coordinates crosstalk between RNA decay and transcription in innate immunity. Nat Commun 2018; 9:1145. [PMID: 29559679 PMCID: PMC5861047 DOI: 10.1038/s41467-018-03326-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 02/05/2018] [Indexed: 12/29/2022] Open
Abstract
Control of type I interferon production is crucial to combat infection while preventing deleterious inflammatory responses, but the extent of the contribution of post-transcriptional mechanisms to innate immune regulation is unclear. Here, we show that human zinc finger RNA-binding protein (ZFR) represses the interferon response by regulating alternative pre-mRNA splicing. ZFR expression is tightly controlled during macrophage development; monocytes express truncated ZFR isoforms, while macrophages induce full-length ZFR to modulate macrophage-specific alternative splicing. Interferon-stimulated genes are constitutively activated by ZFR depletion, and immunostimulation results in hyper-induction of interferon β (IFNβ/IFNB1). Through whole-genome analyses, we show that ZFR controls interferon signaling by preventing aberrant splicing and nonsense-mediated decay of histone variant macroH2A1/H2AFY mRNAs. Together, our data suggest that regulation of ZFR in macrophage differentiation guards against aberrant interferon responses and reveal a network of mRNA processing and decay that shapes the transcriptional response to infection. Type I interferon signaling is critical for the control of infection. Here the authors show that zinc finger RNA-binding protein (ZFR) can control type I interferon responses, and that this control is itself regulated by distinct ZFR truncation patterns that differ between monocytes and macrophages.
Collapse
Affiliation(s)
- Nazmul Haque
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Drive, Room 2341, Bethesda, MD, 20892, USA.
| | - Ryota Ouda
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, 6 Center Drive, Room 2A01, Bethesda, MD, 20892, USA
| | - Chao Chen
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, 6 Center Drive, Room 2A01, Bethesda, MD, 20892, USA
| | - Keiko Ozato
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, 6 Center Drive, Room 2A01, Bethesda, MD, 20892, USA
| | - J Robert Hogg
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Drive, Room 2341, Bethesda, MD, 20892, USA.
| |
Collapse
|
7
|
Asai A, Miyata Y, Takehara K, Kanda S, Watanabe SI, Greer PA, Sakai H. Pathological significance and prognostic significance of FES expression in bladder cancer vary according to tumor grade. J Cancer Res Clin Oncol 2017; 144:21-31. [PMID: 28952025 PMCID: PMC5756570 DOI: 10.1007/s00432-017-2524-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/17/2017] [Indexed: 12/13/2022]
Abstract
Purpose The feline sarcoma oncogene protein (FES) is a non-receptor tyrosine kinase implicated in both oncogenesis and tumor suppression. Here, cancer cell lines and human tissues were employed to clarify the pathological and prognostic significance of FES in bladder cancer. Methods The relationship between FES expression and cancer aggressiveness was investigated using 3 cell lines (T24: corresponding to grade 3, 5637: corresponding to grade 2, and RT4: corresponding to grade 1) and 203 tissues derived from human bladder malignancies. Proliferation, invasion, and migration of cancer cells were assessed following the knockdown (KD) of FES expression by the siRNA method. Relationships between FES expression and pathological features, aggressiveness, and outcome were investigated. Results FES-KD inhibited the proliferation, migration, and invasion of T24 cells but not of RT4 cells and 5637 cells. Considering all patients, FES expression demonstrated a negative relationship with grade but no association with muscle invasion or cancer cell proliferation. However, it was positively correlated with pT stage and cell proliferation in high-grade tumors (p = 0.002); no such association was found for low-grade tumors. In addition, elevated FES expression was a negative prognostic indicator of metastasis after radical surgery for patients with high-grade tumors (p = 0.021) but not for those with low-grade malignancies. Conclusions FES appeared to act as a suppressor of carcinogenesis, being associated with low tumor grade in the overall patient group. However, its expression correlated with cancer aggressiveness and poor outcome in high-grade bladder cancer. FES, therefore, represents a potential therapeutic target and useful prognostic factor for such patients.
Collapse
Affiliation(s)
- Akihiro Asai
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Yasuyoshi Miyata
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan.
| | - Kosuke Takehara
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Shigeru Kanda
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Shin-Ichi Watanabe
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Peter A Greer
- Division of Cancer Biology and Genetics, Department of Pathology and Molecular Medicine, Queen's Cancer Research Institute, Queens University, Kingston, ON, K7L 3N6, Canada
| | - Hideki Sakai
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| |
Collapse
|
8
|
Sodium restriction modulates innate immunity and prevents cardiac remodeling in a rat model of metabolic syndrome. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1568-1574. [DOI: 10.1016/j.bbadis.2017.02.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/16/2017] [Accepted: 02/24/2017] [Indexed: 12/19/2022]
|
9
|
Pesce M, Felaco P, Franceschelli S, Speranza L, Grilli A, De Lutiis MA, Ferrone A, Sirolli V, Bonomini M, Felaco M, Patruno A. Effect of erythropoietin on primed leucocyte expression profile. Open Biol 2014; 4:140026. [PMID: 24920275 PMCID: PMC4077059 DOI: 10.1098/rsob.140026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 05/20/2014] [Indexed: 02/04/2023] Open
Abstract
Resistance to erythropoietin (EPO) affects a significant number of anaemic patients with end-stage renal disease. Previous reports suggest that inflammation is one of the major independent predictors of EPO resistance, and the effects of EPO treatment on inflammatory mediators are not well established. The aim of this study was to investigate EPO-induced modification to gene expression in primary cultured leucocytes. Microarray experiments were performed on primed ex vivo peripheral blood mononuclear cells (PBMCs) and treated with human EPO-α. Data suggested that EPO-α modulated genes involved in cell movement and interaction in primed PBMCs. Of note, EPO-α exerts anti-inflammatory effects inhibiting the expression of pro-inflammatory cytokine IL-8 and its receptor CXCR2; by contrast, EPO-α increases expression of genes relating to promotion of inflammation encoding for IL-1β and CCL8, and induces de novo synthesis of IL-1α, CXCL1 and CXCL5 in primed cells. The reduction in MAPK p38-α activity is involved in modulating both IL-1β and IL-8 expression. Unlike the induction of MAPK, Erk1/2 activity leads to upregulation of IL-1β, but does not affect IL-8 expression and release. Furthermore, EPO-α treatment of primed cells induces the activation of caspase-1 upstream higher secretion of IL-1β, and this process is not dependent on caspase-8 activation. In conclusion, our findings highlight new potential molecules involved in EPO resistance and confirm the anti-inflammatory role for EPO, but also suggest a plausible in vivo scenario in which the positive correlation found between EPO resistance and elevated levels of some pro-inflammatory mediators is due to treatment with EPO itself.
Collapse
Affiliation(s)
- Mirko Pesce
- Department of Psychological, Humanistic and Territorial Sciences, University 'G. D'Annunzio', Chieti, Italy
| | - Paolo Felaco
- Department of Medicine and Science of Aging, University 'G. D'Annunzio', Chieti, Italy
| | - Sara Franceschelli
- Department of Medicine and Science of Aging, University 'G. D'Annunzio', Chieti, Italy
| | - Lorenza Speranza
- Department of Medicine and Science of Aging, University 'G. D'Annunzio', Chieti, Italy
| | - Alfredo Grilli
- Department of Medicine and Science of Aging, University 'G. D'Annunzio', Chieti, Italy
| | - Maria Anna De Lutiis
- Department of Medicine and Science of Aging, University 'G. D'Annunzio', Chieti, Italy
| | - Alessio Ferrone
- Department of Medicine and Science of Aging, University 'G. D'Annunzio', Chieti, Italy
| | - Vittorio Sirolli
- Department of Medicine and Science of Aging, University 'G. D'Annunzio', Chieti, Italy
| | - Mario Bonomini
- Department of Medicine and Science of Aging, University 'G. D'Annunzio', Chieti, Italy
| | - Mario Felaco
- Department of Medicine and Science of Aging, University 'G. D'Annunzio', Chieti, Italy
| | - Antonia Patruno
- Department of Medicine and Science of Aging, University 'G. D'Annunzio', Chieti, Italy
| |
Collapse
|
10
|
Kumar V, Everingham S, Hall C, Greer PA, Craig AWB. Calpains promote neutrophil recruitment and bacterial clearance in an acute bacterial peritonitis model. Eur J Immunol 2013; 44:831-41. [PMID: 24375267 DOI: 10.1002/eji.201343757] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 10/15/2013] [Accepted: 11/29/2013] [Indexed: 12/20/2022]
Abstract
Activation of the innate immune system is critical for clearance of bacterial pathogens to limit systemic infections and host tissue damage. Here, we report a key role for calpain proteases in bacterial clearance in mice with acute peritonitis. Using transgenic mice expressing Cre recombinase primarily in innate immune cells (fes-Cre), we generated conditional capns1 knockout mice. Consistent with capns1 being essential for stability and function of the ubiquitous calpains (calpain-1, calpain-2), peritoneal cells from these mice had reduced levels of calpain-2/capns1, and reduced proteolysis of their substrate selenoprotein K. Using an acute bacterial peritonitis model, we observed impaired bacterial killing within the peritoneum and development of bacteremia in calpain knockout mice. These defects correlated with significant reductions in IL-1α release, neutrophil recruitment, and generation of reactive oxygen species in calpain knockout mice with acute bacterial peritonitis. Peritoneal macrophages from calpain knockout mice infected with enterobacteria ex vivo, were competent in phagocytosis of bacteria, but showed impaired clearance of intracellular bacteria compared with control macrophages. Together, these results implicate calpains as key mediators of effective innate immune responses to acute bacterial infections, to prevent systemic dissemination of bacteria that can lead to sepsis.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | | | | | | | | |
Collapse
|
11
|
Hu J, Wang G, Liu X, Zhou L, Jiang M, Yang L. Polo-like kinase 1 (PLK1) is involved in toll-like receptor (TLR)-mediated TNF-α production in monocytic THP-1 cells. PLoS One 2013; 8:e78832. [PMID: 24205328 PMCID: PMC3799749 DOI: 10.1371/journal.pone.0078832] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 09/16/2013] [Indexed: 12/30/2022] Open
Abstract
Polo-like kinases (PLKs) have been reported to be essential components of anti-viral pathways. However, the role of PLKs in the production of pro-inflammatory cytokines induced by TLR activation is uncertain. We report here that monocytic THP-1 cells expressed PLK1, PLK2, PLK3 and PLK4. When THP-1 cells were treated with GW843682X, an inhibitor of PLK1 and PLK3, the results showed that GW843682X down-regulated Pam3CSK4- and LPS-induced TNF-α at both the gene and protein levels. GW843682X did not impact Pam3CSK4-induced IL-1β and IL-8 or LPS-induced IL-1β, but it down-regulated LPS-induced IL-8 significantly. Moreover, western blot results showed that TLRs activated PLK1, and PLK1 inhibition by RNA interference down-regulated Pam3CSK4-induced TNF-α production, suggesting the involvement of PLK1 in TNF-α up-regulation. In addition, GW843682X treatment for 12 to 24 h induced cell death and down-regulated MyD88, but neither of these roles contributed to the down-regulation of TNF-α, as TNF-α gene expression was up-regulated at 1 h. Furthermore, GW843682X inhibited Pam3CSK4-induced activation of ERK and NF-κB, which contributed to Pam3CSK4-induced up-regulation of TNF-α. GW843682X also inhibited LPS-induced activation of ERK, p38 and NF-κB, which contributed to LPS-induced up-regulation of TNF-α. Taken together, these results suggested that PLK1 is involved in TLR2- and TLR4-induced inflammation, and GW843682X may be valuable for the regulation of the inflammatory response.
Collapse
Affiliation(s)
- Jinyue Hu
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan, China
| | - Guihua Wang
- Cancer Center, Changsha Central Hospital, Changsha, Hunan, China
| | - Xueting Liu
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan, China
| | - Lina Zhou
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Manli Jiang
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan, China
| | - Li Yang
- Tuberculosis Research Center, Changsha Central Hospital, Changsha, Hunan, China
| |
Collapse
|
12
|
Khajah M, Andonegui G, Chan R, Craig AW, Greer PA, McCafferty DM. Fer kinase limits neutrophil chemotaxis toward end target chemoattractants. THE JOURNAL OF IMMUNOLOGY 2013; 190:2208-16. [PMID: 23355730 DOI: 10.4049/jimmunol.1200322] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Neutrophil recruitment and directional movement toward chemotactic stimuli are important processes in innate immune responses. This study examines the role of Fer kinase in neutrophil recruitment and chemotaxis to various chemoattractants in vitro and in vivo. Mice targeted with a kinase-inactivating mutation (Fer(DR/DR)) or wild type (WT) were studied using time-lapse intravital microscopy to examine leukocyte recruitment and chemotaxis in vivo. In response to keratinocyte-derived cytokine, no difference in leukocyte chemotaxis was observed between WT and Fer(DR/DR) mice. However, in response to the chemotactic peptide WKYMVm, a selective agonist of the formyl peptide receptor, a 2-fold increase in leukocyte emigration was noted in Fer(DR/DR) mice (p < 0.05). To determine whether these defects were due to Fer signaling in the endothelium or other nonhematopoietic cells, bone marrow chimeras were generated. WKYMVm-induced leukocyte recruitment in chimeric mice (WT bone marrow to Fer(DR/DR) recipients or vice versa) was similar to WT mice, suggesting that Fer kinase signaling in both leukocytes and endothelial cells serves to limit chemotaxis. Purified Fer(DR/DR) neutrophils demonstrated enhanced chemotaxis toward end target chemoattractants (WKYMVm and C5a) compared with WT using an under-agarose gel chemotaxis assay. These defects were not observed in response to intermediate chemoattractants (keratinocyte-derived cytokine, MIP-2, or LTB(4)). Increased WKYMVm-induced chemotaxis of Fer(DR/DR) neutrophils correlated with sustained PI3K activity and reduced reliance on the p38 MAPK pathway compared with WT neutrophils. Together, these data identify Fer as a novel inhibitory kinase for neutrophil chemotaxis toward end target chemoattractants through modulation of PI3K activity.
Collapse
Affiliation(s)
- Maitham Khajah
- Department of Physiology and Pharmacology, Gastrointestinal Research Group, Institute of Inflammation, Immunity, and Infection, University of Calgary, Calgary, Alberta, Canada T2N 4N1
| | | | | | | | | | | |
Collapse
|
13
|
Kwok E, Everingham S, Zhang S, Greer PA, Allingham JS, Craig AW. FES Kinase Promotes Mast Cell Recruitment to Mammary Tumors via the Stem Cell Factor/KIT Receptor Signaling Axis. Mol Cancer Res 2012; 10:881-91. [DOI: 10.1158/1541-7786.mcr-12-0115] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
14
|
Constitutive MHC class I molecules negatively regulate TLR-triggered inflammatory responses via the Fps-SHP-2 pathway. Nat Immunol 2012; 13:551-9. [PMID: 22522491 DOI: 10.1038/ni.2283] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 03/06/2012] [Indexed: 02/06/2023]
Abstract
The molecular mechanisms that fine-tune Toll-like receptor (TLR)-triggered innate inflammatory responses remain to be fully elucidated. Major histocompatibility complex (MHC) molecules can mediate reverse signaling and have nonclassical functions. Here we found that constitutively expressed membrane MHC class I molecules attenuated TLR-triggered innate inflammatory responses via reverse signaling, which protected mice from sepsis. The intracellular domain of MHC class I molecules was phosphorylated by the kinase Src after TLR activation, then the tyrosine kinase Fps was recruited via its Src homology 2 domain to phosphorylated MHC class I molecules. This led to enhanced Fps activity and recruitment of the phosphatase SHP-2, which interfered with TLR signaling mediated by the signaling molecule TRAF6. Thus, constitutive MHC class I molecules engage in crosstalk with TLR signaling via the Fps-SHP-2 pathway and control TLR-triggered innate inflammatory responses.
Collapse
|
15
|
Miyata Y, Watanabe SI, Matsuo T, Hayashi T, Sakai H, Xuan JW, Greer PA, Kanda S. Pathological significance and predictive value for biochemical recurrence of c-Fes expression in prostate cancer. Prostate 2012; 72:201-8. [PMID: 21563194 DOI: 10.1002/pros.21422] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Accepted: 04/19/2011] [Indexed: 02/01/2023]
Abstract
BACKGROUND c-Fes is a proto-oncogene encoded non-receptor protein-tyrosine kinase (PTK). However, genetic studies have indicated that it has anti-tumorigenic effects in certain cancers. The pathological and clinical significance of c-Fes in prostate cancer are unknown. METHODS Expression of c-Fes was evaluated in normal glands, prostatic intraepithelial neoplasia (PIN), cancer cells in tissues of knock-in mouse adenocarcinoma prostate (KIMAP) model, and prostate cancer patients free of metastasis. Expression of c-Fes was analyzed by immunohistochemistry, and quantified by using the immunoreactivity score (IRS) (staining intensity × percentage of positive cells). Relationships between c-Fes expression and pT stage, Gleason's score (GS), and biochemical recurrence in patients who underwent radical surgery were also investigated. RESULTS In KIMAP, the percentage in normal glands, PIN and cancer cells positive for c-Fes expression were 0 (0/7), 25.0 (2/8), and 100% (7/7), respectively. In human tissues, c-Fes expression was also significantly higher in cancer cells than in normal cells and PIN, and it correlated with pT stage (P < 0.001) and GS (P = 0.047). Multivariate analysis showed that c-Fes expression was an independent predictor of poor outcome poor prognosis (hazard ratio = 3.21, 95% confidence interval = 1.11-9.37, P = 0.032). CONCLUSION The results suggested that c-Fes expression is a useful predictor of biochemical recurrence after radical surgery. The results also suggested that c-Fes is a potentially useful therapeutic target in prostate cancer and a predictor of biochemical recurrence after radical prostatectomy.
Collapse
Affiliation(s)
- Yasuyoshi Miyata
- Department of Nephro-Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Sampson HW, Chaput CD, Brannen J, Probe RA, Guleria RS, Pan J, Baker KM, VanBuren V. Alcohol induced epigenetic perturbations during the inflammatory stage of fracture healing. Exp Biol Med (Maywood) 2011; 236:1389-401. [PMID: 22087020 DOI: 10.1258/ebm.2011.011207] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
It is well recognized by orthopedic surgeons that fractures of alcoholics are more difficult to heal successfully and have a higher incidence of non-union, but the mechanism of alcohol's effect on fracture healing is unknown. In order to give direction for the study of the effects of alcohol on fracture healing, we propose to identify gene expression and microRNA changes during the early stages of fracture healing that might be attributable to alcohol consumption. As the inflammatory stage appears to be the most critical for successful fracture healing, this paper focuses on the events at day three following fracture or the stage of inflammation. Sprague-Dawley rats were placed on an ethanol-containing or pair-fed Lieber and DeCarli diet for four weeks prior to surgical fracture. Following insertion of a medullary pin, a closed mid-diaphyseal fracture was induced using a Bonnarens and Einhorn fracture device. At three days' post-fracture, the region of the fracture calluses was harvested from the right hind-limb. RNA was extracted and microarray analysis was conducted against the entire rat genome. There were 35 genes that demonstrated significant increased expression due to alcohol consumption and 20 that decreased due to alcohol. In addition, the expression of 20 microRNAs was increased and six decreased. In summary, while it is recognized that mRNA levels may or may not represent protein levels successfully produced by the cell, these studies reveal changes in gene expression that support the hypothesis that alcohol consumption affects events involved with inflammation. MicroRNAs are known to modulate mRNA and these findings were consistent with much of what was seen with mRNA microarray analysis, especially the involvement of smad4 which was demonstrated by mRNA microarray, microRNA and polymerase chain reaction.
Collapse
Affiliation(s)
- H Wayne Sampson
- Department of Systems Biology and Translational Medicine, Texas A&M Health Science Center, College of Medicine, USA.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Matijevic N, Wu KK, Howard AG, Wasserman B, Wang WYW, Folsom AR, Sharrett AR. Association of blood monocyte and platelet markers with carotid artery characteristics: the atherosclerosis risk in communities carotid MRI study. Cerebrovasc Dis 2011; 31:552-8. [PMID: 21487219 DOI: 10.1159/000324389] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 01/11/2011] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Atherosclerosis is characterized by infiltration of inflammatory cells from circulating blood. Blood cell activation could play an important role in plaque formation. METHODS We analyzed the relationship between blood cellular markers and quantitative measures of carotid wall components in 1,546 participants from the ARIC (Atherosclerosis Risk in Communities) Carotid MRI Study. Carotid imaging was performed using a gadolinium contrast-enhanced MRI and cellular phenotyping by flow cytometry. RESULTS Monocyte Toll-like receptor (TLR)-2 is associated with larger plaques, while CD14, myeloperoxidase, and TLR-4 associate with smaller. Platelet CD40L is associated with smaller plaques and thinner caps, while P-selectin is associated with smaller core size. CONCLUSIONS Blood cell activation is significantly associated with atherosclerotic changes of the carotid wall.
Collapse
Affiliation(s)
- N Matijevic
- Center for Translational Injury Research, Department of Surgery, University of Texas Health Science Center at Houston, 77030, USA.
| | | | | | | | | | | | | |
Collapse
|
18
|
Zhang S, Chitu V, Stanley ER, Elliott BE, Greer PA. Fes tyrosine kinase expression in the tumor niche correlates with enhanced tumor growth, angiogenesis, circulating tumor cells, metastasis, and infiltrating macrophages. Cancer Res 2010; 71:1465-73. [PMID: 21159660 DOI: 10.1158/0008-5472.can-10-3757] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Fes is a protein tyrosine kinase with cell autonomous oncogenic activities that are well established in cell culture and animal models, but its involvement in human cancer has been unclear. Abundant expression of Fes in vascular endothelial cells and myeloid cell lineages prompted us to explore roles for Fes in the tumor microenvironment. In an orthotopic mouse model of breast cancer, we found that loss of Fes in the host correlated with reductions in engrafted tumor growth rates, metastasis, and circulating tumor cells. The tumor microenvironment in Fes-deficient mice also showed reduced vascularity and fewer macrophages. In co-culture with tumor cells, Fes-deficient macrophages also poorly promoted tumor cell invasive behavior. Taken together, our observations argue that Fes inhibition might provide therapeutic benefits in breast cancer, in part by attenuating tumor-associated angiogenesis and the metastasis-promoting functions of tumor-associated macrophages.
Collapse
Affiliation(s)
- Shengnan Zhang
- Division of Cancer Biology and Genetics, Department of Pathology and Molecular Medicine, Queen's Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
| | | | | | | | | |
Collapse
|
19
|
Gundacker NC, Haudek VJ, Wimmer H, Slany A, Griss J, Bochkov V, Zielinski C, Wagner O, Stöckl J, Gerner C. Cytoplasmic proteome and secretome profiles of differently stimulated human dendritic cells. J Proteome Res 2009; 8:2799-811. [PMID: 19351150 DOI: 10.1021/pr8011039] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Dendritic cells (DCs), the most potent and specialized antigen-presenting cells, play a key role in the regulation of the adaptive immunity. Immature DCs were generated by in vitro culturing of peripheral blood monocytes and functionally activated with the classical pathogen-associated molecular pattern lipopolysaccharide (LPS). Alternative activation resulting in Th-2 polarization was induced with lipid oxidation products derived from 1-palmitoyl-2-arachidoyl-sn-glycerol-3-phosphorylcholin (OxPAPC). Tolerogenic cells were obtained by treating DCs with human rhinovirus (HRV). The aim of this study was the identification of proteome profiles related to the functionally different dendritic cell phenotypes. Cytoplasmic proteins were analyzed by shotgun proteomics resulting in the identification of 1690 proteins. While mature and alternatively activated DCs displayed highly distinct protein expression profiles, HRV-treated DCs showed minor proteome alterations. As DCs exert many specific functions via secretion, we investigated the secretomes by a combination of 2D-PAGE and shotgun proteomics. We successfully identified a broad variety of cytokines (e.g., GM-CSF, TNF-alpha, interleukin-1beta, 6, 12 beta, 28B and 29), chemokines (e.g., CCL3, 5, 8, 17, 18, 19, 24, CXCL1, 2, 9 and 10) and growth factors (growth/differentiation factor 8, C-type lectin domain family 11 member A). The relative composition of secretome profiles, although comprising much less proteins, was found to be much more affected by functional alteration of cells than the cytoplasmic protein composition. In conclusion, we demonstrate that functional distinct subsets of DCs display distinct proteome profiles which comprise biomarker candidates. These proteins may prove useful for the interpretation of complex clinical proteomics data.
Collapse
Affiliation(s)
- Nina C Gundacker
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
fps/fes knockout mice display a lactation defect and the fps/fes tyrosine kinase is a component of E-cadherin-based adherens junctions in breast epithelial cells during lactation. Exp Cell Res 2009; 315:2929-40. [PMID: 19732771 DOI: 10.1016/j.yexcr.2009.08.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2009] [Revised: 08/21/2009] [Accepted: 08/25/2009] [Indexed: 12/18/2022]
Abstract
The fps/fes proto-oncogene encodes a cytoplasmic protein-tyrosine kinase implicated in vesicular trafficking and cytokine and growth factor signaling in hematopoietic, neuronal, vascular endothelial and epithelial lineages. Genetic evidence has suggested a tumor suppressor role for Fps/Fes in breast and colon. Here we used fps/fes knockout mice to investigate potential roles for this kinase in development and function of the mammary gland. Fps/Fes expression was induced during pregnancy and lactation, and its kinase activity was dramatically enhanced. Milk protein and fat composition from nursing fps/fes-null mothers was normal; however, pups reared by them gained weight more slowly than pups reared by wild-type mothers. Fps/Fes displayed a predominantly dispersed punctate intracellular distribution which was consistent with vesicles within the luminal epithelial cells of lactating breast, while a small fraction co-localized with beta-catenin and E-cadherin on their basolateral surfaces. Fps/Fes was found to be a component of the E-cadherin adherens junction (AJ) complex; however, the phosphotyrosine status of beta-catenin and core AJ components in fps/fes-null breast tissue was unaltered, and epithelial cell AJs and gland morphology were intact. We conclude that Fps/Fes is not essential for the maintenance of epithelial cell AJs in the lactating breast but may instead play important roles in vesicular trafficking and milk secretion.
Collapse
|
21
|
Contributions of F-BAR and SH2 domains of Fes protein tyrosine kinase for coupling to the FcepsilonRI pathway in mast cells. Mol Cell Biol 2008; 29:389-401. [PMID: 19001085 DOI: 10.1128/mcb.00904-08] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
This study investigates the roles of Fer-CIP4 homology (FCH)-Bin/amphiphysin/Rvs (F-BAR) and SH2 domains of Fes protein tyrosine kinase in regulating its activation and signaling downstream of the high-affinity immunoglobulin G (IgE) receptor (FcepsilonRI) in mast cells. Homology modeling of the Fes F-BAR domain revealed conservation of some basic residues implicated in phosphoinositide binding (R113/K114). The Fes F-BAR can bind phosphoinositides and induce tubulation of liposomes in vitro. Mutation of R113/K114 to uncharged residues (RK/QQ) caused a significant reduction in phosphoinositide binding in vitro and a more diffuse cytoplasmic localization in transfected COS-7 cells. RBL-2H3 mast cells expressing full-length Fes carrying the RK/QQ mutation show defects in FcepsilonRI-induced Fes tyrosine phosphorylation and degranulation compared to cells expressing wild-type Fes. This correlated with reduced localization to Lyn kinase-containing membrane fractions for the RK/QQ mutant compared to wild-type Fes in mast cells. The Fes SH2 domain also contributes to Fes signaling in mast cells, via interactions with the phosphorylated FcepsilonRI beta chain and the actin regulatory protein HS1. We show that Fes phosphorylates C-terminal tyrosine residues in HS1 implicated in actin stabilization. Thus, coordinated actions of the F-BAR and SH2 domains of Fes allow for coupling to FcepsilonRI signaling and potential regulation the actin reorganization in mast cells.
Collapse
|
22
|
Filippakopoulos P, Kofler M, Hantschel O, Gish GD, Grebien F, Salah E, Neudecker P, Kay LE, Turk BE, Superti-Furga G, Pawson T, Knapp S. Structural coupling of SH2-kinase domains links Fes and Abl substrate recognition and kinase activation. Cell 2008; 134:793-803. [PMID: 18775312 PMCID: PMC2572732 DOI: 10.1016/j.cell.2008.07.047] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2008] [Revised: 06/23/2008] [Accepted: 07/29/2008] [Indexed: 11/05/2022]
Abstract
The SH2 domain of cytoplasmic tyrosine kinases can enhance catalytic activity and substrate recognition, but the molecular mechanisms by which this is achieved are poorly understood. We have solved the structure of the prototypic SH2-kinase unit of the human Fes tyrosine kinase, which appears specialized for positive signaling. In its active conformation, the SH2 domain tightly interacts with the kinase N-terminal lobe and positions the kinase αC helix in an active configuration through essential packing and electrostatic interactions. This interaction is stabilized by ligand binding to the SH2 domain. Our data indicate that Fes kinase activation is closely coupled to substrate recognition through cooperative SH2-kinase-substrate interactions. Similarly, we find that the SH2 domain of the active Abl kinase stimulates catalytic activity and substrate phosphorylation through a distinct SH2-kinase interface. Thus, the SH2 and catalytic domains of active Fes and Abl pro-oncogenic kinases form integrated structures essential for effective tyrosine kinase signaling.
Collapse
Affiliation(s)
- Panagis Filippakopoulos
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Aspenström P. Roles of F-BAR/PCH proteins in the regulation of membrane dynamics and actin reorganization. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2008; 272:1-31. [PMID: 19121815 DOI: 10.1016/s1937-6448(08)01601-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The Pombe Cdc15 Homology (PCH) proteins have emerged in many species as important coordinators of signaling pathways that regulate actomyosin assembly and membrane dynamics. The hallmark of the PCH proteins is the presence of a Fes/CIP4 homology-Bin/Amphiphysin/Rvsp (F-BAR) domain; therefore they are commonly referred to as F-BAR proteins. The prototype F-BAR protein, Cdc15p of Schizosaccharomyces pombe, has a role in the formation of the contractile actomyosin ring during cytokinesis. Vertebrate F-BAR proteins have an established role in binding phospholipids and they participate in membrane deformations, for instance, during the internalization of transmembrane receptors. This way the F-BAR proteins will function as linkers between the actin polymerization apparatus and the machinery regulating membrane dynamics. Interestingly, some members of the F-BAR proteins are implicated in inflammatory or neurodegenerative disorders and the observations can be expected to have clinical implications for the treatment of the diseases.
Collapse
Affiliation(s)
- Pontus Aspenström
- Ludwig Institute for Cancer Research, Uppsala University, SE-751 24 Uppsala, Sweden
| |
Collapse
|
24
|
Voisset E, Lopez S, Dubreuil P, De Sepulveda P. The tyrosine kinase FES is an essential effector of KITD816V proliferation signal. Blood 2007; 110:2593-9. [PMID: 17595334 DOI: 10.1182/blood-2007-02-076471] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
KIT is a tyrosine kinase receptor that is aberrantly activated in several neoplasms. In human pathologies, the most frequent mutation of KIT occurs at codon 816. The resulting KIT mutant protein is activated in the absence of ligand and is resistant to the clinically available inhibitors of KIT. In this report, we provide evidence for an essential function of the cytoplasmic tyrosine kinase FES downstream of KITD816V. FES is phosphorylated on tyrosine residues in cells that carry KITD816V mutation, and this phosphorylation is KIT dependent. Reduction of FES expression using RNA interference results in decreased cell proliferation in human or murine cells harboring KITD816V or the homologous mouse mutation KITD814Y. The reduced cell growth can be rescued using another cytokine (granulocyte-macrophage colony-stimulating factor [GM-CSF]) and is not observed when the closely related fer gene is targeted. Finally, signaling downstream of KITD816V is altered in cells lacking FES expression. This study shows a major function of FES downstream of activated KIT receptor and thereby points to FES as a novel target in KIT-related pathologies.
Collapse
Affiliation(s)
- Edwige Voisset
- Institut National de la Santé et de la Rècherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 599, Centre de Recherche en Cancérologie de Marseille, Laboratoire d'Hématopoïèse Moléculaire et Fonctionnelle, Marseille, France
| | | | | | | |
Collapse
|
25
|
Parsons SA, Mewburn JD, Truesdell P, Greer PA. The Fps/Fes kinase regulates leucocyte recruitment and extravasation during inflammation. Immunology 2007; 122:542-50. [PMID: 17627769 PMCID: PMC2228385 DOI: 10.1111/j.1365-2567.2007.02670.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Fps/Fes and Fer comprise a distinct subfamily of cytoplasmic protein-tyrosine kinases, and have both been implicated in the regulation of innate immunity. Previous studies showed that Fps/Fes-knockout mice were hypersensitive to systemic lipopolysaccharide (LPS) challenge, and Fer-deficient mice displayed enhanced recruitment of leucocytes in response to localized LPS challenge. We show here for the first time, a role for Fps in the regulation of leucocyte recruitment to areas of inflammation. Using the cremaster muscle intravital microscopy model, we observed increased leucocyte adherence to venules, and increased rates and degrees of transendothelial migration in Fps/Fes-knockout mice relative to wild-type animals subsequent to localized LPS challenge. There was also a decreased vessel wall shear rate in the post-capillary venules of LPS-challenged Fps/Fes-knockout mice, and an increase in neutrophil migration into the peritoneal cavity subsequent to thioglycollate challenge. Using flow cytometry to quantify the expression of surface molecules, we observed prolonged expression of the selectin ligand PSGL-1 on peripheral blood neutrophils from Fps/Fes-knockout mice stimulated ex vivo with LPS. These observations provide important insights into the observed in vivo behaviour of leucocytes in LPS-challenged Fps/Fes-knockout mice and provide evidence that the Fps/Fes kinase plays an important role in the innate immune response.
Collapse
Affiliation(s)
- Sean A Parsons
- Division of Cancer Biology and Genetics, Queen's University Cancer Research Institute, Queens University, Kingston, Ontario, Canada
| | | | | | | |
Collapse
|
26
|
Chitu V, Stanley ER. Pombe Cdc15 homology (PCH) proteins: coordinators of membrane-cytoskeletal interactions. Trends Cell Biol 2007; 17:145-56. [PMID: 17296299 DOI: 10.1016/j.tcb.2007.01.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2006] [Revised: 12/20/2006] [Accepted: 01/25/2007] [Indexed: 12/27/2022]
Abstract
Cellular adhesion, motility, endocytosis, exocytosis and cytokinesis involve the coordinated reorganization of the cytoskeleton and of the plasma membrane. The 'Pombe Cdc15 homology' (PCH) family of adaptor proteins has recently been shown to coordinate the membrane and cytoskeletal dynamics involved in these processes by curving membranes, recruiting dynamin and controlling the architecture of the actin cytoskeleton. Mutations in PCH family members or proteins that interact with them are associated with autoinflammatory, neurological or neoplastic diseases. Here, we review the nature, actions and disease associations of the vertebrate PCH family members, highlighting their fundamental roles in the regulation of processes involving membrane-cytoskeletal interactions.
Collapse
Affiliation(s)
- Violeta Chitu
- Department of Developmental Biology and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York, NY 10461, USA
| | | |
Collapse
|