1
|
Chen X, Kang Y, Tang C, Zhang L, Guo L. TLR4 promotes smooth muscle cell-derived foam cells formation by inducing receptor-independent macropinocytosis. Biosci Biotechnol Biochem 2024; 89:22-32. [PMID: 39455413 DOI: 10.1093/bbb/zbae153] [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: 06/04/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
Foam cells are primarily formed through scavenger receptors that mediate the uptake of various modified low-density lipoproteins (LDL) into cells. In addition to the receptor-dependent pathway, macropinocytosis is an essential nonreceptor endocytic pathway for vascular smooth muscle cells (VSMCs) to take up lipids. However, the molecular mechanisms underlying this process remain unclear. Primary cultured VSMCs were stimulated with 200 ng/mL lipopolysaccharide (LPS) and 200 µg/mL native LDL (nLDL). We observed a significant increase in Toll-like receptor 4 (TLR4) protein expression and a significant activation of macropinocytosis, which correlated with the highest uptake of nLDL and intracellular lipid deposition in WT VSMCs. However, macropinocytosis was inhibited and lipid accumulation decreased after treatment with macropinocytosis inhibitors and Syk inhibitors in WT VSMCs. Consistently, TLR4 knockout significantly suppressed macropinocytosis and lipid droplets accumulation in VSMCs. Taken together, our findings suggest a critical role of TLR4/Syk signaling in promoting receptor-independent macropinocytosis leading to VSMC-derived foam cells formation.
Collapse
MESH Headings
- Toll-Like Receptor 4/metabolism
- Pinocytosis/drug effects
- Animals
- Foam Cells/metabolism
- Foam Cells/cytology
- Foam Cells/drug effects
- Syk Kinase/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Signal Transduction
- Lipopolysaccharides/pharmacology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/cytology
- Mice
- Lipoproteins, LDL/metabolism
- Lipoproteins, LDL/pharmacology
- Cells, Cultured
- Mice, Knockout
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Xue Chen
- Department of Rheumatology and Clinical Immunology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Yulai Kang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Chunhua Tang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Lili Zhang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Lu Guo
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| |
Collapse
|
2
|
Skubitz KM. The role of CEACAMs in neutrophil function. Eur J Clin Invest 2024; 54 Suppl 2:e14349. [PMID: 39674879 DOI: 10.1111/eci.14349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/22/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND In addition to the long-known antibacterial actions of neutrophils, neutrophils are recognized to have a variety of other effects and are functionally diverse. Neutrophils can either stimulate or inhibit B cells and T cells, regulate NK development and activity, augment or direct the resolution of inflammation, act as myeloid-derived suppressor cells, modulate tumour growth and metastasis and trigger autoimmune diseases. CEACAMs 1, 3, 6 and 8 are expressed on human neutrophils. METHODS A literature review was performed on the role of CEACAMs in neutrophil function. RESULTS CEACAMs 1, 6 and 8 can be upregulated from intracellular stores, while CEACAM3, an opsonin-independent phagocytic receptor, is constitutively expressed. CEACAM1 has an intracellular ITIM motif and an ITSM motif, and CEACAM3 has an ITAM-like motif; CEACAMs 6 and 8 are glycosylphosphatidylinositol-linked. CEACAM8 can also be released in a soluble form. These CEACAMs can interact with multiple other host CEACAMs as well as other molecules on bacteria, fungi and host cells, both transmitting and receiving signals. Known CEACAM-binding pathogens bind the CFG face of the N domain which is also important in CEACAM-CEACAM binding, although the ABDE face also appears to be involved in higher-order oligomers. CONCLUSIONS Understanding the exact role of each individual CEACAM in human neutrophils is complicated by the fact that the neutrophil CEACAMs can interact with multiple ligands. The data demonstrates some of the many roles of CEACAMs in neutrophil function and the extensive role of the neutrophil in human biology beyond its classical role as a short-lived phagocyte.
Collapse
Affiliation(s)
- Keith M Skubitz
- Department of Medicine, Masonic Cancer Center, University of Minnesota Medical Center, Minneapolis, Minnesota, USA
| |
Collapse
|
3
|
Najjar SM, Shively JE. Regulation of lipid storage and inflammation in the liver by CEACAM1. Eur J Clin Invest 2024; 54 Suppl 2:e14338. [PMID: 39674882 DOI: 10.1111/eci.14338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 10/11/2024] [Indexed: 12/17/2024]
Abstract
This review focuses on a special aspect of hepatic lipid storage and inflammation that occurs during nutritional excess in obesity. Mounting evidence supports that prolonged excess fatty acid (FA) uptake in the liver is strongly associated with hepatic lipid storage and inflammation and that the two processes are closely linked by a homeostatic mechanism. There is also strong evidence that bacterial lipids may enter the gut by a common mechanism with lipid absorption and that there is a set point to determine when their uptake triggers an inflammatory response in the liver. In fact, the progression from high uptake of FAs in the liver resulting in Metabolic dysfunction-associated steatotic liver disease (MASLD) to the development of the more serious Metabolic dysfunction-associated steatohepatitis (MASH) depends on the degree of inflammation and its progression from an acute to a chronic state. Thus, MASLD/MASH implicates both excess fatty acids and progressive inflammation in the aetiology of liver disease. We start the discussion by introduction of CD36, a major player in FA and lipopolysaccharide (LPS) uptake in the duodenum, liver and adipose tissue. We will then introduce CEACAM1, a major player in the regulation of hepatic de novo lipogenesis and the inflammatory response in the liver, and its dual association with CD36 in enterocytes and hepatocytes. We conclude that CEACAM1 and CD36 together regulate lipid droplet formation and inflammation in the liver.
Collapse
Affiliation(s)
- Sonia M Najjar
- Department of Biomedical Sciences and the Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| | - John E Shively
- Department of Immunology and Theranostics, Arthur D. Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| |
Collapse
|
4
|
Dery KJ, Najjar SM, Beauchemin N, Shively JE, Kupiec-Weglinski JW. Mechanism and function of CEACAM1 splice isoforms. Eur J Clin Invest 2024; 54 Suppl 2:e14350. [PMID: 39674874 DOI: 10.1111/eci.14350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 10/29/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Alternative splicing is a fundamental mechanism in the post-transcriptional regulation of genes. The multifunctional transmembrane glycoprotein receptor carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) undergoes extensive alternative splicing to allow for tunable functions in cell signalling, adhesion and modulation of immune and metabolic responses. Splice isoforms that differ in their ectodomain and short or long cytoplasmic tail (CEACAM1-S/CEACAM1-L) have distinct functional roles. The mechanisms that regulate CEACAM1 RNA splicing remain elusive. METHODS This narrative review summarizes the current knowledge of the mechanism and function of CEACAM1 splice isoforms. Historical perspectives address the biological significance of the glycosylated Ig domains, the variable exon 7, and phosphorylation events that dictate its signal transduction pathways. The use of small antisense molecules to target mis-spliced variable exon 7 is discussed. RESULTS The Ig variable-like N domain mediates cell adhesion and immune checkpoint inhibitory functions. Gly and Tyr residues in the transmembrane (TM) domain are essential for dimerization. Calmodulin, Calcium/Calmodulin-dependent protein kinase II delta (CamK2D), Actin and Annexin A2 are binding partners of CEACAM1-S. Homology studies of the muCEACAM1-S and huCEACAM1-S TM predict differences in their signal transduction pathways. Hypoxia-inducible factor 1-α (HIF-1-α) induces alternative splicing to produce CEACAM1-S under limited oxygen conditions. Antisense small molecules directed to exon 7 may correct faulty expression of the short and long cytoplasmic tail splicing isoforms. CONCLUSION More pre-clinical and clinical studies are needed to elucidate the precise mechanisms by which CEACAM1 RNA splicing may be exploited to develop targeted interventions towards novel therapeutic strategies.
Collapse
Affiliation(s)
- Kenneth J Dery
- Department of Surgery, University of California Los Angeles, Los Angeles, California, USA
| | - Sonia M Najjar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| | - Nicole Beauchemin
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| | - John E Shively
- Department of Theranostics and Immunology, Arthur D. Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | | |
Collapse
|
5
|
Sarker S, Köster M, Desai O, Rahim MI, Herrmann S, Behme S, Stiesch M, Hauser H, Wirth D. A generic cell-based biosensor converts bacterial infection signals into chemoattractants for immune cells. Biofabrication 2024; 17:015020. [PMID: 39467389 DOI: 10.1088/1758-5090/ad8bf4] [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: 07/05/2024] [Accepted: 10/28/2024] [Indexed: 10/30/2024]
Abstract
Bacterial infection is a major challenge to human health. Although various potent antibiotics have emerged in recent decades, current challenges arise from the increasing number of multi-drug-resistant species. Infections associated with implants represent a particular challenge because they are usually diagnosed at an advanced stage and are difficult to treat with antibiotics owing to the formation of protective biofilms. In this study, we designed and explored a synthetic biology-inspired cell-based biosensor/actor for the detection and counteraction of bacterial infections. The system is generic, as it senses diverse types of infections and acts by enhancing the endogenous immune system. This strategy is based on genetically engineered sensor/actor cells that can sense type I interferons (IFNs), which are released by immune cells at the early stages of infection. IFN signalling activates a synthetic circuit to induce reporter genes with a sensitivity of only 5 pg ml-1of IFN and leads to a therapeutic protein output of 100 ng ml-1, resulting in theranostic cells that can visualize and fight infections. Robustness and resilience were achieved by implementing a positive feedback loop. We showed that diverse gram-positive and gram-negative implant-associated pathogenic bacteria activate the cascade in co-culture systems in a dose-dependent manner. Finally, we showed that this system can be used to secrete chemoattractants that facilitate the infiltration of immune cells in response to bacterial triggers. Together, the system is not only universal to bacterial infections, but also hypersensitive, allowing the sensing of infections at initial stages.
Collapse
Affiliation(s)
- Sushobhan Sarker
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Inhoffenstr. 7, Braunschweig 38124, Germany
| | - Mario Köster
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Inhoffenstr. 7, Braunschweig 38124, Germany
| | - Omkar Desai
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Inhoffenstr. 7, Braunschweig 38124, Germany
| | - Muhammad Imran Rahim
- Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Stadtfelddamm 34, 30625 Hannover, Germany
| | - Sabrina Herrmann
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Inhoffenstr. 7, Braunschweig 38124, Germany
| | - Sara Behme
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Inhoffenstr. 7, Braunschweig 38124, Germany
| | - Meike Stiesch
- Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Stadtfelddamm 34, 30625 Hannover, Germany
| | - Hansjörg Hauser
- Scientific Strategy, Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124 Braunschweig, Germany
- iBET-Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
| | - Dagmar Wirth
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Inhoffenstr. 7, Braunschweig 38124, Germany
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
6
|
Moussavi-Harami SF, Cleary SJ, Magnen M, Seo Y, Conrad C, English BC, Qiu L, Wang KM, Abram CL, Lowell CA, Looney MR. Neutrophil-specific Shp1 loss results in lethal pulmonary hemorrhage in mouse models of acute lung injury. J Clin Invest 2024; 134:e183161. [PMID: 39352872 PMCID: PMC11645157 DOI: 10.1172/jci183161] [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: 05/23/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is associated with significant morbidity and mortality, and neutrophils are critical to its pathogenesis. Neutrophil activation is closely regulated by inhibitory tyrosine phosphatases including Src homology region 2 domain-containing phosphatase-1 (Shp1). Here, we report that loss of neutrophil Shp1 in mice produced hyperinflammation and lethal pulmonary hemorrhage in sterile inflammation and pathogen-induced models of acute lung injury (ALI) through a Syk kinase-dependent mechanism. We observed large intravascular neutrophil clusters, perivascular inflammation, and excessive neutrophil extracellular traps in neutrophil-specific Shp1-KO mice, suggesting an underlying mechanism for the observed pulmonary hemorrhage. Targeted immunomodulation through the administration of a Shp1 activator (SC43) reduced agonist-induced reactive oxygen species in vitro and ameliorated ALI-induced alveolar neutrophilia and NETs in vivo. We propose that the pharmacologic activation of Shp1 has the potential to fine tune neutrophil hyperinflammation that is central to the pathogenesis of ARDS.
Collapse
Affiliation(s)
- S. Farshid Moussavi-Harami
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
- Division of Pediatric Critical Care Medicine, Department of Pediatrics
| | - Simon J Cleary
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Mélia Magnen
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Yurim Seo
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Catharina Conrad
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | | | - Longhui Qiu
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Kristin M. Wang
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Clare L. Abram
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | | | - Mark R. Looney
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| |
Collapse
|
7
|
Warner S, Teague HL, Ramos-Benitez MJ, Panicker S, Allen K, Gairhe S, Moyer T, Parachalil Gopalan B, Douagi I, Shet A, Kanthi Y, Suffredini AF, Chertow DS, Strich JR. R406 reduces lipopolysaccharide-induced neutrophil activation. Cell Immunol 2024; 403-404:104860. [PMID: 39084187 PMCID: PMC11387147 DOI: 10.1016/j.cellimm.2024.104860] [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: 05/15/2024] [Revised: 07/05/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024]
Abstract
Modulating SYK has been demonstrated to have impacts on pathogenic neutrophil responses in COVID-19. During sepsis, neutrophils are vital in early bacterial clearance but also contribute to the dysregulated immune response and organ injury when hyperactivated. Here, we evaluated the impact of R406, the active metabolite of fostamatinib, on neutrophils stimulated by LPS. We demonstrate that R406 was able to effectively inhibit NETosis, degranulation, ROS generation, neutrophil adhesion, and the formation of CD16low neutrophils that have been linked to detrimental outcomes in severe sepsis. Further, the neutrophils remain metabolically active, capable of releasing cytokines, perform phagocytosis, and migrate in response to IL-8. Taken together, this data provides evidence of the potential efficacy of utilizing fostamatinib in bacterial sepsis.
Collapse
Affiliation(s)
- Seth Warner
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Heather L Teague
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Marcos J Ramos-Benitez
- Basic Science Department, Microbiology Division, School of Medicine, Ponce Health Sciences University, Ponce, PR, USA
| | - Sumith Panicker
- Laboratory of Vascular Thrombosis and Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kiana Allen
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Salina Gairhe
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Tom Moyer
- Flow Cytometry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bindu Parachalil Gopalan
- Laboratory of Sickle Thrombosis and Vascular Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Iyadh Douagi
- Flow Cytometry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA; NIH Center for Human Immunology, Inflammation, and Autoimmunity, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Arun Shet
- Laboratory of Sickle Thrombosis and Vascular Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yogendra Kanthi
- Laboratory of Vascular Thrombosis and Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anthony F Suffredini
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Daniel S Chertow
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA; Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey R Strich
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
8
|
L’Estrange-Stranieri E, Gottschalk TA, Wright MD, Hibbs ML. The dualistic role of Lyn tyrosine kinase in immune cell signaling: implications for systemic lupus erythematosus. Front Immunol 2024; 15:1395427. [PMID: 39007135 PMCID: PMC11239442 DOI: 10.3389/fimmu.2024.1395427] [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: 03/03/2024] [Accepted: 06/17/2024] [Indexed: 07/16/2024] Open
Abstract
Systemic lupus erythematosus (SLE, lupus) is a debilitating, multisystem autoimmune disease that can affect any organ in the body. The disease is characterized by circulating autoantibodies that accumulate in organs and tissues, which triggers an inflammatory response that can cause permanent damage leading to significant morbidity and mortality. Lyn, a member of the Src family of non-receptor protein tyrosine kinases, is highly implicated in SLE as remarkably both mice lacking Lyn or expressing a gain-of-function mutation in Lyn develop spontaneous lupus-like disease due to altered signaling in B lymphocytes and myeloid cells, suggesting its expression or activation state plays a critical role in maintaining tolerance. The past 30 years of research has begun to elucidate the role of Lyn in a duplicitous signaling network of activating and inhibitory immunoreceptors and related targets, including interactions with the interferon regulatory factor family in the toll-like receptor pathway. Gain-of-function mutations in Lyn have now been identified in human cases and like mouse models, cause severe systemic autoinflammation. Studies of Lyn in SLE patients have presented mixed findings, which may reflect the heterogeneity of disease processes in SLE, with impairment or enhancement in Lyn function affecting subsets of SLE patients that may be a means of stratification. In this review, we present an overview of the phosphorylation and protein-binding targets of Lyn in B lymphocytes and myeloid cells, highlighting the structural domains of the protein that are involved in its function, and provide an update on studies of Lyn in SLE patients.
Collapse
Affiliation(s)
- Elan L’Estrange-Stranieri
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Timothy A. Gottschalk
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Mark D. Wright
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Margaret L. Hibbs
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
9
|
Kuiper JWP, Gregg HL, Schüber M, Klein J, Hauck CR. Controling the cytoskeleton during CEACAM3-mediated phagocytosis. Eur J Cell Biol 2024; 103:151384. [PMID: 38215579 DOI: 10.1016/j.ejcb.2024.151384] [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: 09/01/2023] [Revised: 11/20/2023] [Accepted: 01/03/2024] [Indexed: 01/14/2024] Open
Abstract
Phagocytosis, an innate defense mechanism of multicellular animals, is initiated by specialized surface receptors. A phagocytic receptor expressed by human polymorphonuclear granulocytes, the major professional phagocytes in our body, is one of the fastest evolving human proteins implying a special role in human biology. This receptor, CEACAM3, is a member of the CarcinoEmbryonic Antigen-related Cell Adhesion Molecule (CEACAM) family and dedicated to the immediate recognition and rapid internalization of human-restricted pathogens. In this focused contribution, we will review the special adaptations of this protein, which co-evolves with different species of mucosa-colonizing bacteria. While the extracellular Immunoglobulin-variable (IgV)-like domain recognizes various bacterial adhesins, an Immunoreceptor Tyrosine-based Activation Motif (ITAM)-like sequence in the cytoplasmic tail of CEACAM3 constitutes the central signaling hub to trigger actin rearrangements needed for efficient phagocytosis. A major emphasis of this review will be placed on recent findings, which have revealed the multi-level control of this powerful phagocytic device. As tyrosine phosphorylation and small GTPase activity are central for CEACAM3-mediated phagocytosis, the counterregulation of CEACAM3 activity involves the receptor-type protein tyrosine phosphatase J (PTPRJ) as well as the Rac-GTP scavenging protein Cyri-B. Interference with such negative regulatory circuits has revealed that CEACAM3-mediated phagocytosis can be strongly enhanced. In principle, the knowledge gained by studying CEACAM3 can be applied to other phagocytic systems and opens the door to treatments, which boost the phagocytic capacity of professional phagocytes.
Collapse
Affiliation(s)
| | - Helena L Gregg
- Lehrstuhl Zellbiologie, Fachbereich Biologie, Universität Konstanz, Germany
| | - Meike Schüber
- Lehrstuhl Zellbiologie, Fachbereich Biologie, Universität Konstanz, Germany
| | - Jule Klein
- Lehrstuhl Zellbiologie, Fachbereich Biologie, Universität Konstanz, Germany
| | - Christof R Hauck
- Lehrstuhl Zellbiologie, Fachbereich Biologie, Universität Konstanz, Germany; Konstanz Research School Chemical Biology, Universität Konstanz, Germany.
| |
Collapse
|
10
|
Curran CS, Cui X, Li Y, Jeakle M, Sun J, Demirkale CY, Minkove S, Hoffmann V, Dhamapurkar R, Chumbris S, Bolyard C, Iheanacho A, Eichacker PQ, Torabi-Parizi P. Anti-PD-L1 therapy altered inflammation but not survival in a lethal murine hepatitis virus-1 pneumonia model. Front Immunol 2024; 14:1308358. [PMID: 38259435 PMCID: PMC10801642 DOI: 10.3389/fimmu.2023.1308358] [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: 10/06/2023] [Accepted: 12/05/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction Because prior immune checkpoint inhibitor (ICI) therapy in cancer patients presenting with COVID-19 may affect outcomes, we investigated the beta-coronavirus, murine hepatitis virus (MHV)-1, in a lethal pneumonia model in the absence (Study 1) or presence of prior programmed cell death ligand-1 (PD-L1) antibody (PD-L1mAb) treatment (Study 2). Methods In Study 1, animals were inoculated intratracheally with MHV-1 or vehicle and evaluated at day 2, 5, and 10 after infection. In Study 2, uninfected or MHV-1-infected animals were pretreated intraperitoneally with control or PD-L1-blocking antibodies (PD-L1mAb) and evaluated at day 2 and 5 after infection. Each study examined survival, physiologic and histologic parameters, viral titers, lung immunophenotypes, and mediator production. Results Study 1 results recapitulated the pathogenesis of COVID-19 and revealed increased cell surface expression of checkpoint molecules (PD-L1, PD-1), higher expression of the immune activation marker angiotensin converting enzyme (ACE), but reduced detection of the MHV-1 receptor CD66a on immune cells in the lung, liver, and spleen. In addition to reduced detection of PD-L1 on all immune cells assayed, PD-L1 blockade was associated with increased cell surface expression of PD-1 and ACE, decreased cell surface detection of CD66a, and improved oxygen saturation despite reduced blood glucose levels and increased signs of tissue hypoxia. In the lung, PD-L1mAb promoted S100A9 but inhibited ACE2 production concomitantly with pAKT activation and reduced FOXO1 levels. PD-L1mAb promoted interferon-γ but inhibited IL-5 and granulocyte-macrophage colony-stimulating factor (GM-CSF) production, contributing to reduced bronchoalveolar lavage levels of eosinophils and neutrophils. In the liver, PD-L1mAb increased viral clearance in association with increased macrophage and lymphocyte recruitment and liver injury. PD-L1mAb increased the production of virally induced mediators of injury, angiogenesis, and neuronal activity that may play role in COVID-19 and ICI-related neurotoxicity. PD-L1mAb did not affect survival in this murine model. Discussion In Study 1 and Study 2, ACE was upregulated and CD66a and ACE2 were downregulated by either MHV-1 or PD-L1mAb. CD66a is not only the MHV-1 receptor but also an identified immune checkpoint and a negative regulator of ACE. Crosstalk between CD66a and PD-L1 or ACE/ACE2 may provide insight into ICI therapies. These networks may also play role in the increased production of S100A9 and neurological mediators in response to MHV-1 and/or PD-L1mAb, which warrant further study. Overall, these findings support observational data suggesting that prior ICI treatment does not alter survival in patients presenting with COVID-19.
Collapse
Affiliation(s)
- Colleen S. Curran
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Xizhong Cui
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Yan Li
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Mark Jeakle
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Cumhur Y. Demirkale
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Samuel Minkove
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Victoria Hoffmann
- Division of Veterinary Resources, National Institutes of Health, Bethesda, MD, United States
| | - Rhea Dhamapurkar
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Symya Chumbris
- Texcell North-America, Inc., Frederick, MD, United States
| | | | | | - Peter Q. Eichacker
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Parizad Torabi-Parizi
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
11
|
Curson JE, Liu L, Luo L, Muusse TW, Lucas RM, Gunther KS, Vajjhala PR, Abrol R, Jones A, Kapetanovic R, Stacey KJ, Stow JL, Sweet MJ. TLR4 phosphorylation at tyrosine 672 activates the ERK/c-FOS signaling module for LPS-induced cytokine responses in macrophages. Eur J Immunol 2023; 53:e2250056. [PMID: 37058370 PMCID: PMC10947571 DOI: 10.1002/eji.202250056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 03/20/2023] [Accepted: 04/11/2023] [Indexed: 04/15/2023]
Abstract
TLRs engage numerous adaptor proteins and signaling molecules, enabling a complex series of post-translational modifications (PTMs) to mount inflammatory responses. TLRs themselves are post-translationally modified following ligand-induced activation, with this being required to relay the full spectrum of proinflammatory signaling responses. Here, we reveal indispensable roles for TLR4 Y672 and Y749 phosphorylation in mounting optimal LPS-inducible inflammatory responses in primary mouse macrophages. LPS promotes phosphorylation at both tyrosine residues, with Y749 phosphorylation being required for maintenance of total TLR4 protein levels and Y672 phosphorylation exerting its pro-inflammatory effects more selectively by initiating ERK1/2 and c-FOS phosphorylation. Our data also support a role for the TLR4-interacting membrane proteins SCIMP and the SYK kinase axis in mediating TLR4 Y672 phosphorylation to permit downstream inflammatory responses in murine macrophages. The corresponding residue in human TLR4 (Y674) is also required for optimal LPS signaling responses. Our study, thus, reveals how a single PTM on one of the most widely studied innate immune receptors orchestrates downstream inflammatory responses.
Collapse
Affiliation(s)
- James E.B. Curson
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Liping Liu
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Lin Luo
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Timothy W. Muusse
- School of Chemistry and Molecular Biosciences (SCMB) and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Richard M. Lucas
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Kimberley S. Gunther
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Parimala R. Vajjhala
- School of Chemistry and Molecular Biosciences (SCMB) and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Rishika Abrol
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Alun Jones
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Ronan Kapetanovic
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | - Katryn J. Stacey
- School of Chemistry and Molecular Biosciences (SCMB) and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Jennifer L. Stow
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Matthew J. Sweet
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| |
Collapse
|
12
|
Catton EA, Bonsor DA, Herrera C, Stålhammar-Carlemalm M, Lyndin M, Turner CE, Soden J, van Strijp JAG, Singer BB, van Sorge NM, Lindahl G, McCarthy AJ. Human CEACAM1 is targeted by a Streptococcus pyogenes adhesin implicated in puerperal sepsis pathogenesis. Nat Commun 2023; 14:2275. [PMID: 37080973 PMCID: PMC10119177 DOI: 10.1038/s41467-023-37732-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 03/27/2023] [Indexed: 04/22/2023] Open
Abstract
Life-threatening bacterial infections in women after childbirth, known as puerperal sepsis, resulted in classical epidemics and remain a global health problem. While outbreaks of puerperal sepsis have been ascribed to Streptococcus pyogenes, little is known about disease mechanisms. Here, we show that the bacterial R28 protein, which is epidemiologically associated with outbreaks of puerperal sepsis, specifically targets the human receptor CEACAM1. This interaction triggers events that would favor the development of puerperal sepsis, including adhesion to cervical cells, suppression of epithelial wound repair and subversion of innate immune responses. High-resolution structural analysis showed that an R28 domain with IgI3-like fold binds to the N-terminal domain of CEACAM1. Together, these findings demonstrate that a single adhesin-receptor interaction can drive the pathogenesis of bacterial sepsis and provide molecular insights into the pathogenesis of one of the most important infectious diseases in medical history.
Collapse
Affiliation(s)
- Erin A Catton
- Centre for Bacterial Resistance Biology, Section of Molecular Microbiology, Department of Infectious Diseases, Imperial College London, London, SW7 2AZ, UK
| | - Daniel A Bonsor
- University of Maryland, Baltimore, MD, 21201, USA
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Carolina Herrera
- Section of Immunology of Infection, Department of Infectious Disease, Imperial College London, London, W2 1NY, UK
| | | | - Mykola Lyndin
- Sumy State University, Sumy, 40000, Ukraine
- Institute of Anatomy, Medical Faculty, University of Duisburg-Essen, Essen, 45147, Germany
| | - Claire E Turner
- The School of Biosciences, The Florey Institute, The University of Sheffield, Sheffield, S10 2TN, UK
| | - Jo Soden
- Retrogenix, Chinley, High Peak, SK23 6FJ, Chinley, UK
| | - Jos A G van Strijp
- Department of Medical Microbiology, UMC Utrecht, Utrecht, 3584 CX, The Netherlands
| | - Bernhard B Singer
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC location University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, 1105 AZ, The Netherlands
| | - Nina M van Sorge
- Department of Medical Microbiology, UMC Utrecht, Utrecht, 3584 CX, The Netherlands.
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC location University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, 1105 AZ, The Netherlands.
- Netherlands Reference Laboratory for Bacterial Meningitis, Amsterdam UMC, location AMC, Amsterdam, 1105 AZ, The Netherlands.
| | - Gunnar Lindahl
- Department of Laboratory Medicine, Division of Medical Microbiology, Lund University, Lund, 223 62, Sweden.
- Department of Chemistry, Division of Applied Microbiology, Lund University, Lund, 221 00, Sweden.
| | - Alex J McCarthy
- Centre for Bacterial Resistance Biology, Section of Molecular Microbiology, Department of Infectious Diseases, Imperial College London, London, SW7 2AZ, UK.
- Department of Medical Microbiology, UMC Utrecht, Utrecht, 3584 CX, The Netherlands.
| |
Collapse
|
13
|
Hirao H, Kojima H, Dery KJ, Nakamura K, Kadono K, Zhai Y, Farmer DG, Kaldas FM, Kupiec-Weglinski JW. Neutrophil CEACAM1 determines susceptibility to NETosis by regulating the S1PR2/S1PR3 axis in liver transplantation. J Clin Invest 2023; 133:e162940. [PMID: 36719377 PMCID: PMC9888387 DOI: 10.1172/jci162940] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 11/17/2022] [Indexed: 02/01/2023] Open
Abstract
Neutrophils, the largest innate immune cell population in humans, are the primary proinflammatory sentinel in the ischemia-reperfusion injury (IRI) mechanism in orthotopic liver transplantation (OLT). Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1, CC1, or CD66a) is essential in neutrophil activation and serves as a checkpoint regulator of innate immune-driven IRI cascade in OLT. Although CC1 alternative splicing generates two functionally distinct short and long cytoplasmic isoforms, their role in neutrophil activation remains unknown. Here, we undertook molecular and functional studies to interrogate the significance of neutrophil CC1 signaling in mouse and human OLT recipients. In the experimental arm, we employed a mouse OLT model to document that ablation of recipient-derived neutrophil CC1-long (CC1-L) isotype aggravated hepatic IRI by promoting neutrophil extracellular traps (NETs). Notably, by regulating the S1P-S1PR2/S1PR3 axis, neutrophil CC1-L determined susceptibility to NET formation via autophagy signaling. In the clinical arm, liver grafts from 55 transplant patients selectively enriched for neutrophil CC1-L showed relative resistance to ischemia-reperfusion (IR) stress/tissue damage, improved hepatocellular function, and clinical outcomes. In conclusion, despite neutrophils being considered a principal villain in peritransplant tissue injury, their CC1-L isoform may serve as a regulator of IR stress resistance/NETosis in human and mouse OLT recipients.
Collapse
Affiliation(s)
- Hirofumi Hirao
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Hidenobu Kojima
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Kenneth J. Dery
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Kojiro Nakamura
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kentaro Kadono
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Yuan Zhai
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Douglas G. Farmer
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Fady M. Kaldas
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jerzy W. Kupiec-Weglinski
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
14
|
Scozzi D, Gelman AE. Avoid being trapped by your liver: ischemia-reperfusion injury in liver transplant triggers S1P-mediated NETosis. J Clin Invest 2023; 133:e167012. [PMID: 36719374 PMCID: PMC9888375 DOI: 10.1172/jci167012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Liver transplantation can be a life-saving treatment for end-stage hepatic disease. Unfortunately, some recipients develop ischemia-reperfusion injury (IRI) that leads to poor short- and long-term outcomes. Recent work has shown neutrophils contribute to IRI by undergoing NETosis, a form of death characterized by DNA ejection resulting in inflammatory extracellular traps. In this issue of the JCI, Hirao and Kojima et al. report that sphingosine-1-phosphate (S1P) expression induced by liver transplant-mediated IRI triggers NETosis. They also provide evidence that neutrophil expression of the carcinoembryonic antigen-related cell adhesion molecule-1 (CC1) long isoform inhibited NETosis by controlling S1P receptor-mediated autophagic flux. These findings suggest stimulating regulatory mechanisms that suppress NETosis could be used to prevent IRI.
Collapse
Affiliation(s)
- Davide Scozzi
- Norton Thoracic Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - Andrew E. Gelman
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
15
|
CEACAM1 Is a Prognostic Biomarker and Correlated with Immune Cell Infiltration in Clear Cell Renal Cell Carcinoma. DISEASE MARKERS 2023; 2023:3606362. [PMID: 36712923 PMCID: PMC9876685 DOI: 10.1155/2023/3606362] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/24/2022] [Accepted: 01/05/2023] [Indexed: 01/19/2023]
Abstract
Background CEACAM1 has been shown to be aberrantly expressed in a variety of tumors, and modulation of CEACAM1-related signaling pathways has been suggested as a novel approach for cancer immunotherapy in recent years. However, its role in clear cell renal cell carcinoma (ccRCC) is unclear. Methods The relationship between CEACAM1 and ccRCC was demonstrated based on data from TCGA, GEO, and HPA databases. And the relationship between clinicopathological features and CEACAM1 expression was also assessed. Survival curve analysis was performed to analyze the prognostic relationship between CEACAM1 expression and ccRCC. Protein interaction network analysis was used to analyze the relationship between CEACAM1 and microenvironment-related proteins. In addition, the immunomodulatory role of CEACAM1 in ccRCC was assessed by analyzing CEACAM1 and immune cell infiltration. Results The expression of CEACAM1 was lower in ccRCC tissues than in adjacent normal tissues, and its expression level was negatively correlated with tumor size status (P < 0.001), metastasis status (P = 0.009), pathological stage (P = 0.002), gender (P < 0.001), histological grade (P < 0.001), and primary therapy outcome (P = 0.045) of ccRCC. Survival curve analysis showed that ccRCC patients with lower CEACAM1 expression exhibited shorter overall survival (P < 0.001), and CEACAM1 interacted with microenvironmental molecules such as fibronectin and integrins. Furthermore, immune infiltration analysis showed that CEACAM1 expression correlated with CD8+ and CD4+ T cells, macrophage, neutrophil, and dendritic cell infiltration in ccRCC. Conclusions CEACAM1 expression correlates with progression, prognosis, and immune cell infiltration in ccRCC patients, and it may be a promising prognostic biomarker and therapeutic target for ccRCC.
Collapse
|
16
|
Matsumoto H, Fujita Y, Onizawa M, Saito K, Sumichika Y, Yoshida S, Temmoku J, Matsuoka N, Yashiro-Furuya M, Asano T, Sato S, Suzuki E, Machida T, Watanabe H, Migita K. Increased CEACAM1 expression on peripheral blood neutrophils in patients with rheumatoid arthritis. Front Immunol 2022; 13:978435. [PMID: 36591283 PMCID: PMC9794574 DOI: 10.3389/fimmu.2022.978435] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Altered expression of adhesion molecules in immune cells has been demonstrated in rheumatoid arthritis (RA). Carcinoembryonic-antigen-related cell-adhesion molecule 1 (CEACAM1) is an adhesion molecule that acts as a coinhibitory receptor in the immune system. We investigated the role of CEACAM1 in immune cell subsets of patients with RA. Peripheral blood was obtained from 37 patients with RA and 20 healthy controls (HC). The expression of CEACAM1 and T-cell immunoglobulin mucin domain molecule (TIM) -3 on peripheral blood mononuclear cells and neutrophils was analyzed by flow cytometry. Intracellular TIM-3 expression was analyzed using cellular lysates by Western blot analysis. Serum levels of soluble CEACAM1 (sCEACAM1) were estimated by an enzyme-linked immunosorbent assay. CEACAM1 expression was not detected in peripheral blood mononuclear cells, including in CD14(+) monocytes and CD3(+) lymphocytes isolated from patients with RA or HC. However, substantial cell-surface expression of CEACAM1 was detected in peripheral blood neutrophils, and it was significantly elevated in samples from patients with RA without remission compared to those in remission. There was no significant difference in serum levels of sCEACAM1 between patients with RA and HC. Cell-surface expression of TIM-3 was not detected in peripheral blood neutrophils from patients with RA or HC but was seen in CD14(+) monocytes. However, there was no significant difference in TIM-3 expression on monocytes between patients with RA and HC. Our data indicate that cell-surface expression of CEACAM1 on peripheral blood neutrophils are higher in patients with RA and that it is associated with rheumatoid inflammation. Further studies are needed to explore the potential role of CEACAM1 in rheumatoid inflammatory pathways.
Collapse
Affiliation(s)
- Haruki Matsumoto
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Yuya Fujita
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Michio Onizawa
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Kenji Saito
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Yuya Sumichika
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Shuhei Yoshida
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Jumpei Temmoku
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Naoki Matsuoka
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Makiko Yashiro-Furuya
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Tomoyuki Asano
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Shuzo Sato
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Eiji Suzuki
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Takeshi Machida
- Department of Immunology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Hiroshi Watanabe
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Kiyoshi Migita
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan,*Correspondence: Kiyoshi Migita,
| |
Collapse
|
17
|
Cao M, Ma L, Yan C, Wang H, Ran M, Chen Y, Wang X, Liang X, Chai L, Li X. Mouse Ocilrp2/Clec2i negatively regulates LPS-mediated IL-6 production by blocking Dap12-Syk interaction in macrophage. Front Immunol 2022; 13:984520. [PMID: 36300111 PMCID: PMC9589251 DOI: 10.3389/fimmu.2022.984520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 09/20/2022] [Indexed: 11/24/2022] Open
Abstract
C-type lectin Ocilrp2/Clec2i is widely expressed in dendritic cells, lymphokine-activated killer cells and activated T cells. Previous studies have shown that Ocilrp2 is an important regulator in the activation of T cells and NK cells. However, the role of Ocilrp2 in the inflammatory responses by activated macrophages is currently unknown. This study investigated the expression of inflammatory cytokines in LPS-induced macrophages from primary peritoneal macrophages silenced by specific siRNA target Ocilrp2. Ocilrp2 was significantly downregulated in macrophages via NF-κB and pathways upon LPS stimuli or VSV infection. Silencing Ocilrp2 resulted in the increased expression of IL-6 in LPS-stimulated peritoneal macrophages and mice. Moreover, IL-6 expression was reduced in LPS-induced Ocilrp2 over-expressing iBMDM cells. Furthermore, we found that Ocilrp2-related Syk activation is responsible for expressing inflammatory cytokines in LPS-stimulated macrophages. Silencing Ocilrp2 significantly promotes the binding of Syk to Dap12. Altogether, we identified the Ocilrp2 as a critical role in the TLR4 signaling pathway and inflammatory macrophages’ immune regulation, and added mechanistic insights into the crosstalk between TLR and Syk signaling.
Collapse
Affiliation(s)
- Mingya Cao
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
- Institute of Translational Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Lina Ma
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Chenyang Yan
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Han Wang
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Mengzhe Ran
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Ying Chen
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Xiao Wang
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Xiaonan Liang
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Lihui Chai
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
- Institute of Translational Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
- *Correspondence: Lihui Chai, ; Xia Li,
| | - Xia Li
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
- Institute of Translational Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
- *Correspondence: Lihui Chai, ; Xia Li,
| |
Collapse
|
18
|
Zhang Y, Wang Y, Wu W, Liu P, Sun S, Hong M, Yuan Y, Xia Q, Chen Z. Elevation of neutrophil carcinoembryonic antigen-related cell adhesion molecule 1 associated with multiple inflammatory mediators was related to different clinical stages in ischemic stroke patients. J Clin Lab Anal 2022; 36:e24526. [PMID: 35657334 PMCID: PMC9279952 DOI: 10.1002/jcla.24526] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND We aimed to analyze the level of carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) in neutrophils of ischemic stroke (IS) patients at different stages, together with its roles in neutrophils. PATIENTS AND METHODS Sixty-seven patients were classified into acute phase group (n = 19), subacute phase group (n = 28), and stable phase group (n = 20), and 20 healthy individuals who had received physical examination at the same time period as healthy control. We then analyzed the expression level of CEACAM1 and cell viability in CEACAM1 positive and CEACAM1 negative neutrophils by flow cytometry and the content of plasma CEACAM1, neutrophil gelatinase-associated lipocalin (NGAL), matrix metalloproteinases-9 (MMP-9) was measured using enzyme-linked immunosorbent assay (ELISA), while that of interleukin-10 (IL-10) and tumor necrosis factor (TNF) was determined using a Human Enhanced Sensitivity Flex set. RESULTS Compared with healthy control, the percentage of CEACAM1 positive neutrophils in IS patients showed a significant increase, and a significant increase was also noticed in the content of plasma CEACAM1 at the subacute stage. Reduction in cell viability was observed in CEACAM1 positive neutrophils compared with CEACAM1 negative counterparts. There was a positive correlation between CEACAM1 expression rate in neutrophils and plasma CEACAM1 and IL-10 content in the subacute group. Compared with acute group and healthy control group, there was an instinct increase in the level of plasma MMP-9 and NGAL in subacute group. CONCLUSIONS Our data showed that there was a rapid increase of CEACAM1 in neutrophils at the acute stage of IS. We speculated that CEACAM1 may serve as an inhibitory regulator involving in the progression of IS.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Laboratory MedicineThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang ProvinceHangzhouChina
| | - Yijie Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Wei Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Ping Liu
- Department of NeurologyThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Shanshan Sun
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Meng Hong
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Yuan Yuan
- Department of NeurologyThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Qi Xia
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Zhi Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| |
Collapse
|
19
|
Takagane K, Umakoshi M, Itoh G, Kuriyama S, Goto A, Tanaka M. SKAP2 suppresses inflammation-mediated tumorigenesis by regulating SHP-1 and SHP-2. Oncogene 2022; 41:1087-1099. [PMID: 35034964 DOI: 10.1038/s41388-021-02153-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 12/01/2021] [Accepted: 12/09/2021] [Indexed: 12/29/2022]
Abstract
Inflammatory bowel diseases, like ulcerative colitis and Crohn's disease are frequently accompanied by colorectal cancers. However, the mechanisms underlying colitis-associated cancers are not fully understood. Src Kinase Associated Phosphoprotein 2 (SKAP2), a substrate of Src family kinases, is highly expressed in macrophages. Here, we examined the effects of SKAP2 on inflammatory responses in a mouse model of tumorigenesis with colitis induced by azoxymethane/dextran sulfate sodium. SKAP2 knockout increased the severity of colitis and tumorigenesis, as well as lipopolysaccharide (LPS) induced acute inflammation. SKAP2 attenuated inflammatory signaling in macrophages induced by uptake of cancer cell-derived exosomes. SKAP2-/- mice were characterized by the activation of NF-κB signaling and the upregulation and release of cytokines including TNFα, IL-1β, IL-6, CXCL-9/-10/-13, and sICAM1; SKAP2 overexpression attenuated NF-κB activation. Mechanistically, SKAP2 formed a complex with the SHP-1 tyrosine phosphatase via association with the Sirpα transmembrane receptor. SKAP2 also physically associated with the TIR domain of MyD88, TIRAP, and TRAM, adaptors of toll-like receptor 4 (TLR4). SKAP2-mediated recruitment of the Sirpα/SHP-1 complex to TLR4 attenuated inflammatory responses, whereas direct interaction of SKAP2 with SHP-2 decreased SHP-2 activation. SHP-2 is required for efficient NF-κB activation and suppresses the TRAM/TRIF-INFβ pathway; therefore, SKAP2-mediated SHP-2 inhibition affected two signaling axes from TLR4. The present findings indicate that SKAP2 prevents excess inflammation by inhibiting the TLR4-NF-κB pathway, and it activates the TLR4-IFNβ pathway through SHP-1 and SHP-2, thereby suppressing inflammation-mediated tumorigenesis.
Collapse
Affiliation(s)
- Kurara Takagane
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
- Technical Division, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Michinobu Umakoshi
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Go Itoh
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Sei Kuriyama
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Akiteru Goto
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Masamitsu Tanaka
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan.
| |
Collapse
|
20
|
Klaile E, Prada Salcedo JP, Klassert TE, Besemer M, Bothe AK, Durotin A, Müller MM, Schmitt V, Luther CH, Dittrich M, Singer BB, Dandekar T, Slevogt H. Antibody ligation of CEACAM1, CEACAM3, and CEACAM6, differentially enhance the cytokine release of human neutrophils in responses to Candida albicans. Cell Immunol 2021; 371:104459. [PMID: 34847408 DOI: 10.1016/j.cellimm.2021.104459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/27/2021] [Accepted: 11/15/2021] [Indexed: 11/30/2022]
Abstract
Invasive candidiasis is a healthcare-associated fungal infection with a high mortality rate. Neutrophils, the first line of defense during fungal infections, express the immunoregulatory Candida albicans receptors CEACAM1, CEACAM3, and CEACAM6. We analyzed the effects of specific antibodies on C. albicans-induced neutrophil responses. CEACAM6 ligation by 1H7-4B and to some extent CEACAM1 ligation by B3-17, but not CEACAM3 ligation by 308/3-3, resulted in the immediate release of stored CXCL8 and altered transcriptional responses of the C. albicans-stimulated neutrophils. Integrated network analyses and dynamic simulations of signaling cascades predicted alterations in apoptosis and cytokine secretion. We verified that CEACAM6 ligation enhanced Candida-induced neutrophil apoptosis and increased long-term IL-1β/IL-6 release in responses to C. albicans. CEACAM3 ligation, but not CEACAM1 ligation, increased the long-term release of pro-inflammatory IL-1β/IL-6. Taken together, we demonstrated for the first time that ligation of CEACAM receptors differentially affects the regulation of C. albicans-induced immune functions in human neutrophils.
Collapse
Affiliation(s)
- Esther Klaile
- ZIK Septomics, University Hospital Jena, Albert-Einstein-Straße 10, 07749 Jena, Germany.
| | - Juan P Prada Salcedo
- Dept. of Bioinformatics, University of Würzburg, Biocenter/Am Hubland, 97074 Würzburg, Germany.
| | - Tilman E Klassert
- ZIK Septomics, University Hospital Jena, Albert-Einstein-Straße 10, 07749 Jena, Germany.
| | - Matthias Besemer
- ZIK Septomics, University Hospital Jena, Albert-Einstein-Straße 10, 07749 Jena, Germany.
| | - Anne-Katrin Bothe
- ZIK Septomics, University Hospital Jena, Albert-Einstein-Straße 10, 07749 Jena, Germany.
| | - Adrian Durotin
- ZIK Septomics, University Hospital Jena, Albert-Einstein-Straße 10, 07749 Jena, Germany.
| | - Mario M Müller
- ZIK Septomics, University Hospital Jena, Albert-Einstein-Straße 10, 07749 Jena, Germany.
| | - Verena Schmitt
- Institute of Anatomy, University Hospital, University Duisburg-Essen, Hufelandstraße 55, 45122 Essen, Germany.
| | - Christian H Luther
- Dept. of Bioinformatics, University of Würzburg, Biocenter/Am Hubland, 97074 Würzburg, Germany.
| | - Marcus Dittrich
- Dept. of Bioinformatics, University of Würzburg, Biocenter/Am Hubland, 97074 Würzburg, Germany; Dept. of Human Genetics, University of Würzburg, Biocenter/Am Hubland, 97074 Würzburg, Germany.
| | - Bernhard B Singer
- Institute of Anatomy, University Hospital, University Duisburg-Essen, Hufelandstraße 55, 45122 Essen, Germany.
| | - Thomas Dandekar
- Dept. of Bioinformatics, University of Würzburg, Biocenter/Am Hubland, 97074 Würzburg, Germany.
| | - Hortense Slevogt
- ZIK Septomics, University Hospital Jena, Albert-Einstein-Straße 10, 07749 Jena, Germany.
| |
Collapse
|
21
|
Fan Z, Pathak JL, Ge L. The Potential Role of RP105 in Regulation of Inflammation and Osteoclastogenesis During Inflammatory Diseases. Front Cell Dev Biol 2021; 9:713254. [PMID: 34414191 PMCID: PMC8369417 DOI: 10.3389/fcell.2021.713254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/09/2021] [Indexed: 11/16/2022] Open
Abstract
Inflammatory diseases have a negative impact on bone homeostasis via exacerbated local and systemic inflammation. Bone resorbing osteoclasts are mainly derived from hematopoietic precursors and bone marrow monocytes. Induced osteoclastogenesis during inflammation, autoimmunity, metabolic diseases, and cancers is associated with bone loss and osteoporosis. Proinflammatory cytokines, pathogen-associated molecular patterns, or endogenous pathogenic factors induce osteoclastogenic differentiation by binding to the Toll-like receptor (TLR) family expressed on surface of osteoclast precursors. As a non-canonical member of the TLRs, radioprotective 105 kDa (RP105 or CD180) and its ligand, myeloid differentiation protein 1 (MD1), are involved in several bone metabolic disorders. Reports from literature had demonstrated RP105 as an important activator of B cells, bone marrow monocytes, and macrophages, which regulates inflammatory cytokines release from immune cells. Reports from literature had shown the association between RP105 and other TLRs, and the downstream signaling mechanisms of RP105 with different “signaling-competent” partners in immune cells during different disease conditions. This review is focused to summarize: (1) the role of RP105 on immune cells’ function and inflammation regulation (2) the potential regulatory roles of RP105 in different disease-mediated osteoclast activation and the underlying mechanisms, and (3) the different “signaling-competent” partners of RP105 that regulates osteoclastogenesis.
Collapse
Affiliation(s)
- Zhou Fan
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Janak L Pathak
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Linhu Ge
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China.,Institute of Oral Disease, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
22
|
Seumen CHT, Grimm TM, Hauck CR. Protein phosphatases in TLR signaling. Cell Commun Signal 2021; 19:45. [PMID: 33882943 PMCID: PMC8058998 DOI: 10.1186/s12964-021-00722-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptors (TLRs) are critical sensors for the detection of potentially harmful microbes. They are instrumental in initiating innate and adaptive immune responses against pathogenic organisms. However, exaggerated activation of TLR receptor signaling can also be responsible for the onset of autoimmune and inflammatory diseases. While positive regulators of TLR signaling, such as protein serine/threonine kinases, have been studied intensively, only little is known about phosphatases, which counterbalance and limit TLR signaling. In this review, we summarize protein phosphorylation events and their roles in the TLR pathway and highlight the involvement of protein phosphatases as negative regulators at specific steps along the TLR-initiated signaling cascade. Then, we focus on individual phosphatase families, specify the function of individual enzymes in TLR signaling in more detail and give perspectives for future research. A better understanding of phosphatase-mediated regulation of TLR signaling could provide novel access points to mitigate excessive immune activation and to modulate innate immune signaling.![]() Video Abstract
Collapse
Affiliation(s)
- Clovis H T Seumen
- Lehrstuhl Zellbiologie, Universität Konstanz, Universitätsstraße 10, Postablage 621, 78457, Konstanz, Germany
| | - Tanja M Grimm
- Lehrstuhl Zellbiologie, Universität Konstanz, Universitätsstraße 10, Postablage 621, 78457, Konstanz, Germany.,Konstanz Research School Chemical Biology, Universität Konstanz, 78457, Konstanz, Germany
| | - Christof R Hauck
- Lehrstuhl Zellbiologie, Universität Konstanz, Universitätsstraße 10, Postablage 621, 78457, Konstanz, Germany. .,Konstanz Research School Chemical Biology, Universität Konstanz, 78457, Konstanz, Germany.
| |
Collapse
|
23
|
Chen HY, Gao LT, Yuan JQ, Zhang YJ, Liu P, Wang G, Ni X, Liu WN, Gao L. Decrease in SHP-1 enhances myometrium remodeling via FAK activation leading to labor. Am J Physiol Endocrinol Metab 2020; 318:E930-E942. [PMID: 32343611 DOI: 10.1152/ajpendo.00068.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Preterm birth is one of the most common complications during human pregnancy and is associated with a dramatic switch within the uterus from quiescence to contractility. However, the mechanisms underlying uterine remodeling are largely unknown. Protein kinases and phosphatases play critical roles in regulating the phosphorylation of proteins involved in the smooth muscle cell functions. In the present study, we found that Src-homology phosphatase type-1 (SHP-1, PTPN6) was significantly decreased in human myometrium in labor compared with that not in labor. Timed-pregnant mice injected intraperitoneally with the specific SHP-1 inhibitor protein tyrosine phosphatase inhibitor I (PTPI-1) manifested significantly preterm labor, with enriched plasmalemmal dense plaques between myometrial cells and increased phosphorylation at Tyr397 and Tyr576/577 sites of focal adhesion kinase (FAK) in myometrial cells, which remained to the time of labor, whereas the phosphorylation levels of ERK1/2 and phosphatidylinositol 3 kinase (PI3K) showed a rapid increase upon PTPI-1 injection but fell back to normal at the time of labor. The Tyr576/577 in FAK played an important role in the interaction between FAK and SHP-1. Knockdown of SHP-1 dramatically increased the spontaneous contraction of human uterine smooth muscle cells (HUSMCs), which was reversed by coinfection of a FAK-knockdown lentivirus. PGF2α downregulated SHP-1 via PLCβ-PKC-NF-κB or PI3K-NF-κB pathways, suggesting the regenerative downregulation of SHP-1 enhances the uterine remodeling and plasticity by activating FAK and subsequent focal adhesion pathway, which eventually facilitates myometrium contraction and leads to labor. The study sheds new light on understanding of mechanisms that underlie the initiation of labor, and interventions for modulation of SHP-1 may provide a potential strategy for preventing preterm birth.
Collapse
Affiliation(s)
- Huai-Yan Chen
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Ling-Tong Gao
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Jian-Qiang Yuan
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Yu-Ji Zhang
- Department of Physiology, Second Military Medical University, Shanghai, China
- Department of Cardiovascular Surgery, Shenyang Northern Hospital, Shenyang, China
| | - Pei Liu
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Gang Wang
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Xin Ni
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Wei-Na Liu
- Department of Physiology, Second Military Medical University, Shanghai, China
- Department of Obstetrics and Gynecology, Chinese Eastern Theatre Naval Hospital, Ningbo, China
| | - Lu Gao
- Department of Physiology, Second Military Medical University, Shanghai, China
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| |
Collapse
|
24
|
Klaile E, Müller MM, Zubiría-Barrera C, Brehme S, Klassert TE, Stock M, Durotin A, Nguyen TD, Feer S, Singer BB, Zipfel PF, Rudolphi S, Jacobsen ID, Slevogt H. Unaltered Fungal Burden and Lethality in Human CEACAM1-Transgenic Mice During Candida albicans Dissemination and Systemic Infection. Front Microbiol 2019; 10:2703. [PMID: 31849868 PMCID: PMC6889641 DOI: 10.3389/fmicb.2019.02703] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/07/2019] [Indexed: 12/29/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1, CD66a) is a receptor for Candida albicans. It is crucial for the immune response of intestinal epithelial cells to this opportunistic pathogen. Moreover, CEACAM1 is of importance for the mucosal colonization by different bacterial pathogens. We therefore studied the influence of the human CEACAM1 receptor in human CEACAM1-transgenic mice on the C. albicans colonization and infection utilizing a colonization/dissemination and a systemic infection mouse model. Our results showed no alterations in the host response between the transgenic mice and the wild-type littermates to the C. albicans infections. Both mouse strains showed comparable C. albicans colonization and mycobiota, similar fungal burdens in various organs, and a similar survival in the systemic infection model. Interestingly, some of the mice treated with anti-bacterial antibiotics (to prepare them for C. albicans colonization via oral infection) also showed a strong reduction in endogenous fungi instead of the normally observed increase in fungal numbers. This was independent of the expression of human CEACAM1. In the systemic infection model, the human CEACAM1 expression was differentially regulated in the kidneys and livers of Candida-infected transgenic mice. Notably, in the kidneys, a total loss of the largest human CEACAM1 isoform was observed. However, the overwhelming immune response induced in the systemic infection model likely covered any CEACAM1-specific effects in the transgenic animals. In vitro studies using bone marrow-derived neutrophils from both mouse strains also revealed no differences in their reaction to C. albicans. In conclusion, in contrast to bacterial pathogens interacting with CEACAM1 on different mucosal surfaces, the human CEACAM1-transgenic mice did not reveal a role of human CEACAM1 in the in vivo candidiasis models used here. Further studies and different approaches will be needed to reveal a putative role of CEACAM1 in the host response to C. albicans.
Collapse
Affiliation(s)
- Esther Klaile
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Mario M Müller
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Cristina Zubiría-Barrera
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Saskia Brehme
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Tilman E Klassert
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Magdalena Stock
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Adrian Durotin
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Tien D Nguyen
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Sabina Feer
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Bernhard B Singer
- Medical Faculty, Institute of Anatomy, University Duisburg-Essen, Essen, Germany
| | - Peter F Zipfel
- Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany.,Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany
| | - Sven Rudolphi
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany.,Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany.,Center for Sepsis Control and Care (CSCC), University Hospital Jena, Jena, Germany
| | - Ilse D Jacobsen
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany.,Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany.,Center for Sepsis Control and Care (CSCC), University Hospital Jena, Jena, Germany
| | - Hortense Slevogt
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| |
Collapse
|
25
|
Leukocyte immunoglobulin-like receptor B4 deficiency exacerbates acute lung injury via NF-κB signaling in bone marrow-derived macrophages. Biosci Rep 2019; 39:BSR20181888. [PMID: 31138763 PMCID: PMC6566464 DOI: 10.1042/bsr20181888] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 05/17/2019] [Accepted: 05/22/2019] [Indexed: 02/08/2023] Open
Abstract
Acute lung injury (ALI) is an acute inflammatory disease. Leukocyte immunoglobulin-like receptor B4 (LILRB4) is an immunoreceptor tyrosine-based inhibitory motif (ITIM)-bearing inhibitory receptor that is implicated in various pathological processes. However, the function of LILRB4 in ALI remains largely unknown. The aim of the present study was to explore the role of LILRB4 in ALI. LILRB4 knockout mice (LILRB4 KO) were used to construct a model of ALI. Bone marrow cell transplantation was used to identify the cell source of the LILRB4 deficiency-aggravated inflammatory response in ALI. The effect on ALI was analyzed by pathological and molecular analyses. Our results indicated that LILRB4 KO exacerbated ALI triggered by LPS. Additionally, LILRB4 deficiency can enhance lung inflammation. According to the results of our bone marrow transplant model, LILRB4 regulates the occurrence and development of ALI by bone marrow-derived macrophages (BMDMs) rather than by stromal cells in the lung. The observed inflammation was mainly due to BMDM-induced NF-κB signaling. In conclusion, our study demonstrates that LILRB4 deficiency plays a detrimental role in ALI-associated BMDM activation by prompting the NF-κB signal pathway.
Collapse
|
26
|
Identification of Potential Transcriptional Biomarkers Differently Expressed in Both S. aureus- and E. coli-Induced Sepsis via Integrated Analysis. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2487921. [PMID: 31093495 PMCID: PMC6481126 DOI: 10.1155/2019/2487921] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/13/2019] [Accepted: 03/25/2019] [Indexed: 01/13/2023]
Abstract
Sepsis is a critical, complex medical condition, and the major causative pathogens of sepsis are both Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli). Genome-wide studies identify differentially expressed genes for sepsis. However, the results for the identification of DEGs are inconsistent or discrepant among different studies because of heterogeneity of specimen sources, various data processing methods, or different backgrounds of the samples. To identify potential transcriptional biomarkers that are differently expressed in S. aureus- and E. coli-induced sepsis, we have analyzed four microarray datasets from GEO database and integrated results with bioinformatics tools. 42 and 54 DEGs were identified in both S. aureus and E. coli samples from any three different arrays, respectively. Hierarchical clustering revealed dramatic differences between control and sepsis samples. GO functional annotations suggested that DEGs in the S. aureus group were mainly involved in the responses of both defense and immune regulation, but DEGs in the E. coli group were mainly related to the regulation of endopeptidase activity involved in the apoptotic signaling pathway. Although KEGG showed inflammatory bowel disease in the E. coli group, the KEGG pathway analysis showed that these DEGs were mainly involved in the tumor necrosis factor signaling pathway, fructose metabolism, and mannose metabolism in both S. aureus- and E. coli-induced sepsis. Eight common genes were identified between sepsis patients with either S. aureus or E. coli infection and controls in this study. All the candidate genes were further validated to be differentially expressed by an ex-vivo human blood model, and the relative expression of these genes was performed by qPCR. The qPCR results suggest that GK and PFKFB3 might contribute to the progression of S. aureus-induced sepsis, and CEACAM1, TNFAIP6, PSTPIP2, SOCS3, and IL18RAP might be closely linked with E. coli-induced sepsis. These results provide new viewpoints for the pathogenesis of both sepsis and pathogen identification.
Collapse
|
27
|
Size Matters: The Functional Role of the CEACAM1 Isoform Signature and Its Impact for NK Cell-Mediated Killing in Melanoma. Cancers (Basel) 2019; 11:cancers11030356. [PMID: 30871206 PMCID: PMC6468645 DOI: 10.3390/cancers11030356] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/21/2019] [Accepted: 03/09/2019] [Indexed: 12/22/2022] Open
Abstract
Malignant melanoma is the most aggressive and treatment resistant type of skin cancer. It is characterized by continuously rising incidence and high mortality rate due to its high metastatic potential. Various types of cell adhesion molecules have been implicated in tumor progression in melanoma. One of these, the carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), is a multi-functional receptor protein potentially expressed in epithelia, endothelia, and leukocytes. CEACAM1 often appears in four isoforms differing in the length of their extracellular and intracellular domains. Both the CEACAM1 expression in general, and the ratio of the expressed CEACAM1 splice variants appear very dynamic. They depend on both the cell activation stage and the cell growth phase. Interestingly, normal melanocytes are negative for CEACAM1, while melanomas often show high expression. As a cell–cell communication molecule, CEACAM1 mediates the direct interaction between tumor and immune cells. In the tumor cell this interaction leads to functional inhibitions, and indirectly to decreased cancer cell immunogenicity by down-regulation of ligands of the NKG2D receptor. On natural killer (NK) cells it inhibits NKG2D-mediated cytolysis and signaling. This review focuses on novel mechanistic insights into CEACAM1 isoforms for NK cell-mediated immune escape mechanisms in melanoma, and their clinical relevance in patients suffering from malignant melanoma.
Collapse
|
28
|
Al-Harbi NO, Nadeem A, Ahmad SF, Alanazi MM, Aldossari AA, Alasmari F. Amelioration of sepsis-induced acute kidney injury through inhibition of inflammatory cytokines and oxidative stress in dendritic cells and neutrophils respectively in mice: Role of spleen tyrosine kinase signaling. Biochimie 2019; 158:102-110. [DOI: 10.1016/j.biochi.2018.12.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 12/21/2018] [Indexed: 02/06/2023]
|
29
|
Zhang Z, La Placa D, Nguyen T, Kujawski M, Le K, Li L, Shively JE. CEACAM1 regulates the IL-6 mediated fever response to LPS through the RP105 receptor in murine monocytes. BMC Immunol 2019; 20:7. [PMID: 30674283 PMCID: PMC6345024 DOI: 10.1186/s12865-019-0287-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 01/11/2019] [Indexed: 12/12/2022] Open
Abstract
Background Systemic inflammation and the fever response to pathogens are coordinately regulated by IL-6 and IL-1β. We previously showed that CEACAM1 regulates the LPS driven expression of IL-1β in murine neutrophils through its ITIM receptor. Results We now show that the prompt secretion of IL-6 in response to LPS is regulated by CEACAM1 expression on bone marrow monocytes. Ceacam1−/− mice over-produce IL-6 in response to an i.p. LPS challenge, resulting in prolonged surface temperature depression and overt diarrhea compared to their wild type counterparts. Intraperitoneal injection of a 64Cu-labeled LPS, PET imaging agent shows confined localization to the peritoneal cavity, and fluorescent labeled LPS is taken up by myeloid splenocytes and muscle endothelial cells. While bone marrow monocytes and their progenitors (CD11b+Ly6G−) express IL-6 in the early response (< 2 h) to LPS in vitro, these cells are not detected in the bone marrow after in vivo LPS treatment perhaps due to their rapid and complete mobilization to the periphery. Notably, tissue macrophages are not involved in the early IL-6 response to LPS. In contrast to human monocytes, TLR4 is not expressed on murine bone marrow monocytes. Instead, the alternative LPS receptor RP105 is expressed and recruits MD1, CD14, Src, VAV1 and β-actin in response to LPS. CEACAM1 negatively regulates RP105 signaling in monocytes by recruitment of SHP-1, resulting in the sequestration of pVAV1 and β-actin from RP105. Conclusion This novel pathway and regulation of IL-6 signaling by CEACAM1 defines a novel role for monocytes in the fever response of mice to LPS. Electronic supplementary material The online version of this article (10.1186/s12865-019-0287-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhifang Zhang
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, 1500E Duarte Road, Duarte, CA91010, USA.
| | - Deirdre La Placa
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, 1500E Duarte Road, Duarte, CA91010, USA
| | - Tung Nguyen
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, 1500E Duarte Road, Duarte, CA91010, USA
| | - Maciej Kujawski
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, 1500E Duarte Road, Duarte, CA91010, USA
| | - Keith Le
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, 1500E Duarte Road, Duarte, CA91010, USA
| | - Lin Li
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, 1500E Duarte Road, Duarte, CA91010, USA
| | - John E Shively
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, 1500E Duarte Road, Duarte, CA91010, USA.
| |
Collapse
|
30
|
Schirbel A, Rebert N, Sadler T, West G, Rieder F, Wagener C, Horst A, Sturm A, de la Motte C, Fiocchi C. Mutual Regulation of TLR/NLR and CEACAM1 in the Intestinal Microvasculature: Implications for IBD Pathogenesis and Therapy. Inflamm Bowel Dis 2019; 25:294-305. [PMID: 30295747 PMCID: PMC6327233 DOI: 10.1093/ibd/izy316] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Indexed: 12/16/2022]
Abstract
Background Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) displays multiple activities, among which pathogen binding and angiogenesis are particularly prominent. These same functions are also exerted by Toll- and NOD-like receptors (TLRs and NLRs), which are critical mediators of innate immune responses. We investigated whether a functional inter-relationship exists between CEACAM1 and TLRs and NLRs and its potential impact on induction of intestinal angiogenesis. Methods This hypothesis was tested using human intestinal microvascular endothelial cells, a unique cell population exposed to microbial products under physiological and pathological conditions. Results The results show that activation of TLR2/4, TLR4, NOD1, and NOD2 by specific bacterial ligands selectively and differentially upregulates the levels of cellular and soluble CEACAM1 produced by intestinal microvascular endothelial cells. The results also show that CEACAM1 regulates the migration, transmigration, and tube formation of these endothelial cells and mediates vessel sprouting induced by specific TLR and NLR bacterial ligands. Combined, these results demonstrate a close and reciprocal regulatory interaction between CEACAM1 and bacterial products in mediating multiple functions essential to new vessel formation in the gut mucosa. Conclusions A coordinated and reciprocal interaction of CEACAM1 and microbiota-derived factors is necessary to optimize angiogenesis in the gut mucosa. This suggests that a coordination of endogenous and exogenous innate immune responses is necessary to promote intestinal angiogenesis under physiological and inflammatory conditions such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Anja Schirbel
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
- Department of Inflammation and Immunity, Lerner Research Institute, Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio
| | - Nancy Rebert
- Department of Inflammation and Immunity, Lerner Research Institute, Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio
| | - Tammy Sadler
- Department of Inflammation and Immunity, Lerner Research Institute, Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio
| | - Gail West
- Department of Inflammation and Immunity, Lerner Research Institute, Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio
| | | | - Andrea Horst
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas Sturm
- DRK Kliniken Berlin, Schwerpunkt Gastroenterologie, Berlin, Germany
| | - Carol de la Motte
- Department of Inflammation and Immunity, Lerner Research Institute, Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio
| | - Claudio Fiocchi
- Department of Inflammation and Immunity, Lerner Research Institute, Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
31
|
Horst AK, Najjar SM, Wagener C, Tiegs G. CEACAM1 in Liver Injury, Metabolic and Immune Regulation. Int J Mol Sci 2018; 19:ijms19103110. [PMID: 30314283 PMCID: PMC6213298 DOI: 10.3390/ijms19103110] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/02/2018] [Accepted: 10/04/2018] [Indexed: 02/06/2023] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is a transmembrane glycoprotein that is expressed on epithelial, endothelial and immune cells. CEACAM1 is a differentiation antigen involved in the maintenance of epithelial polarity that is induced during hepatocyte differentiation and liver regeneration. CEACAM1 regulates insulin sensitivity by promoting hepatic insulin clearance, and controls liver tolerance and mucosal immunity. Obese insulin-resistant humans with non-alcoholic fatty liver disease manifest loss of hepatic CEACAM1. In mice, deletion or functional inactivation of CEACAM1 impairs insulin clearance and compromises metabolic homeostasis which initiates the development of obesity and hepatic steatosis and fibrosis with other features of non-alcoholic steatohepatitis, and adipogenesis in white adipose depot. This is followed by inflammation and endothelial and cardiovascular dysfunctions. In obstructive and inflammatory liver diseases, soluble CEACAM1 is shed into human bile where it can serve as an indicator of liver disease. On immune cells, CEACAM1 acts as an immune checkpoint regulator, and deletion of Ceacam1 gene in mice causes exacerbation of inflammation and hyperactivation of myeloid cells and lymphocytes. Hence, hepatic CEACAM1 resides at the central hub of immune and metabolic homeostasis in both humans and mice. This review focuses on the regulatory role of CEACAM1 in liver and biliary tract architecture in health and disease, and on its metabolic role and function as an immune checkpoint regulator of hepatic inflammation.
Collapse
Affiliation(s)
- Andrea Kristina Horst
- Institute of Experimental Immunology and Hepatology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany.
| | - Sonia M Najjar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Irvine Hall, 1 Ohio University, Athens, OH 45701-2979, USA.
- The Diabetes Institute, Heritage College of Osteopathic Medicine, Irvine Hall, 1 Ohio University, Athens, OH 45701-2979, USA.
| | - Christoph Wagener
- University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany.
| | - Gisa Tiegs
- Institute of Experimental Immunology and Hepatology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany.
| |
Collapse
|
32
|
Fong JJ, Tsai CM, Saha S, Nizet V, Varki A, Bui JD. Siglec-7 engagement by GBS β-protein suppresses pyroptotic cell death of natural killer cells. Proc Natl Acad Sci U S A 2018; 115:10410-10415. [PMID: 30254166 PMCID: PMC6187154 DOI: 10.1073/pnas.1804108115] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Natural killer (NK) cells are innate immune lymphocytes that recognize and destroy abnormal host cells, such as tumor cells or those infected by viral pathogens. To safely accomplish these functions, NK cells display activating receptors that detect stress molecules or viral ligands displayed at the cell surface, balanced by inhibitory receptors that bind to self-molecules. To date, such activating and inhibitory receptors on NK cells are not known to recognize bacterial determinants. Moreover, NK cell responses to direct interactions with extracellular bacteria are poorly explored. In this study, we observed the human neonatal pathogen group B Streptococcus (GBS) can directly engage human NK cells. The interaction was mediated through the B6N segment of streptococcal β-protein, binding to the inhibitory receptor Siglec-7 via its amino-terminal V-set domain. Unlike classical Siglec binding, the interaction is also independent of its sialic acid recognition property. In contrast to WT GBS, mutants lacking β-protein induced efficient pyroptosis of NK cells through the NLRP3 inflammasome, with production and secretion of the proinflammatory cytokine IL-1β and dissemination of the cytotoxic molecule granzyme B. We postulate that GBS evolved β-protein engagement of inhibitory human Siglec-7 to suppress the pyroptotic response of NK cells and thereby block recruitment of a broader innate immune response, i.e., by "silencing the sentinel."
Collapse
Affiliation(s)
- Jerry J Fong
- Glycobiology Research and Training Center, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Chih-Ming Tsai
- Glycobiology Research and Training Center, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Sudeshna Saha
- Glycobiology Research and Training Center, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Victor Nizet
- Glycobiology Research and Training Center, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Skaggs School of Pharmacy and Pharmaceutical Sciences, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Ajit Varki
- Glycobiology Research and Training Center, School of Medicine, University of California, San Diego, La Jolla, CA 92093;
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Jack D Bui
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
33
|
The Mycobacterial Adjuvant Analogue TDB Attenuates Neuroinflammation via Mincle-Independent PLC-γ1/PKC/ERK Signaling and Microglial Polarization. Mol Neurobiol 2018; 56:1167-1187. [PMID: 29876879 DOI: 10.1007/s12035-018-1135-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 05/18/2018] [Indexed: 02/06/2023]
Abstract
Microglial activation has long been recognized as a hallmark of neuroinflammation. Recently, the bacillus Calmette-Guerin (BCG) vaccine has been reported to exert neuroprotective effects against several neurodegenerative disorders. Trehalose-6,6'-dibehenate (TDB) is a synthetic analogue of trehalose-6,6'-dimycolate (TDM, also known as the mycobacterial cord factor) and is a new adjuvant of tuberculosis subunit vaccine currently in clinical trials. Both TDM and TDB can activate macrophages and dendritic cells through binding to C-type lectin receptor Mincle; however, its action mechanism in microglia and their relationship with neuroinflammation are still unknown. In this article, we found that TDB inhibited LPS-induced M1 microglial polarization in primary microglia and BV-2 cells. However, TDB itself had no effects on IKK, p38, and JNK activities or cytokine expression. In contrast, TDB activated ERK1/2 through PLC-γ1/PKC signaling and in turn decreased LPS-induced NF-κB nuclear translocation. Furthermore, TDB-induced AMPK activation via PLC-γ1/calcium/CaMKKβ-dependent pathway and thereby enhanced M2 gene expressions. Interestingly, knocking out Mincle did not alter the anti-inflammatory and M2 polarization effects of TDB in microglia. Conditional media from LPS-stimulated microglial cells can induce in vitro neurotoxicity, and this action was attenuated by TDB. Using a mouse neuroinflammation model, we found that TDB suppressed LPS-induced M1 microglial activation and sickness behavior, but promoted M2 microglial polarization in both WT and Mincle-/- mice. Taken together, our results suggest that TDB can act independently of Mincle to inhibit LPS-induced inflammatory response through PLC-γ1/PKC/ERK signaling and promote microglial polarization towards M2 phenotype via PLC-γ1/calcium/CaMKKβ/AMPK pathway. Thus, TDB may be a promising therapeutic agent for the treatment of neuroinflammatory diseases.
Collapse
|
34
|
Dery KJ, Silver C, Yang L, Shively JE. Interferon regulatory factor 1 and a variant of heterogeneous nuclear ribonucleoprotein L coordinately silence the gene for adhesion protein CEACAM1. J Biol Chem 2018; 293:9277-9291. [PMID: 29720400 DOI: 10.1074/jbc.ra117.001507] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/27/2018] [Indexed: 12/14/2022] Open
Abstract
The adhesion protein carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is widely expressed in epithelial cells as a short cytoplasmic isoform (S-iso) and in leukocytes as a long cytoplasmic isoform (L-iso) and is frequently silenced in cancer by unknown mechanisms. Previously, we reported that interferon response factor 1 (IRF1) biases alternative splicing (AS) to include the variable exon 7 (E7) in CEACAM1, generating long cytoplasmic isoforms. We now show that IRF1 and a variant of heterogeneous nuclear ribonucleoprotein L (Lv1) coordinately silence the CEACAM1 gene. RNAi-mediated Lv1 depletion in IRF1-treated HeLa and melanoma cells induced significant CEACAM1 protein expression, reversed by ectopic Lv1 expression. The Lv1-mediated CEACAM1 repression resided in residues Gly71-Gly89 and Ala38-Gly89 in Lv1's N-terminal extension. ChIP analysis of IRF1- and FLAG-tagged Lv1-treated HeLa cells and global treatment with the global epigenetic modifiers 5-aza-2'-deoxycytidine and trichostatin A indicated that IRF1 and Lv1 together induce chromatin remodeling, restricting IRF1 access to the CEACAM1 promoter. In interferon γ-treated HeLa cells, the transcription factor SP1 did not associate with the CEACAM1 promoter, but binding by upstream transcription factor 1 (USF1), a known CEACAM1 regulator, was greatly enhanced. ChIP-sequencing revealed that Lv1 overexpression in IRF1-treated cells induces transcriptional silencing across many genes, including DCC (deleted in colorectal carcinoma), associated with CEACAM5 in colon cancer. Notably, IRF1, but not IRF3 and IRF7, affected CEACAM1 expression via translational repression. We conclude that IRF1 and Lv1 coordinately regulate CEACAM1 transcription, alternative splicing, and translation and may significantly contribute to CEACAM1 silencing in cancer.
Collapse
Affiliation(s)
- Kenneth J Dery
- From the Department of Molecular Immunology, Beckman Research Institute of the City of Hope, Duarte, California 91010
| | - Craig Silver
- Department of Biological Sciences, California State Polytechnic University, Pomona, California 91768, and
| | - Lu Yang
- The Integrative Genomics and Bioinformatics Core, Beckman Research Institute of the City of Hope, Duarte, California 91010
| | - John E Shively
- From the Department of Molecular Immunology, Beckman Research Institute of the City of Hope, Duarte, California 91010,
| |
Collapse
|
35
|
Abstract
Neutrophils are a major player in host immunity to infection; however, the mechanisms by which human neutrophils respond to the intracellular protozoan parasite Toxoplasma gondii are still poorly understood. In the current study, we found that, whereas primary human monocytes produced interleukin-1beta (IL-1β) in response to T. gondii infection, human neutrophils from the same blood donors did not. Moreover, T. gondii inhibited lipopolysaccharide (LPS)-induced IL-1β synthesis in human peripheral blood neutrophils. IL-1β suppression required active parasite invasion, since heat-killed or mycalolide B-treated parasites did not inhibit IL-1β release. By investigating the mechanisms involved in this process, we found that T. gondii infection of neutrophils treated with LPS resulted in reduced transcript levels of IL-1β and NLRP3 and reduced protein levels of pro-IL-1β, mature IL-1β, and the inflammasome sensor NLRP3. In T. gondii-infected neutrophils stimulated with LPS, the levels of MyD88, TRAF6, IKKα, IKKβ, and phosphorylated IKKα/β were not affected. However, LPS-induced IκBα degradation and p65 phosphorylation were reduced in T. gondii-infected neutrophils, and degradation of IκBα was reversed by treatment with the proteasome inhibitor MG-132. Finally, we observed that T. gondii inhibited the cleavage and activity of caspase-1 in human neutrophils. These results indicate that T. gondii suppression of IL-1β involves a two-pronged strategy whereby T. gondii inhibits both NF-κB signaling and activation of the NLRP3 inflammasome. These findings represent a novel mechanism of T. gondii evasion of human neutrophil-mediated host defense by targeting the production of IL-1β. Toxoplasma gondii is an obligate intracellular parasite that infects approximately one-third of humans worldwide and can invade virtually any nucleated cell in the human body. Although it is well documented that neutrophils infiltrate the site of acute T. gondii infection, there is limited understanding of how human neutrophils respond to T. gondii. Neutrophils control infectious pathogens by a variety of mechanisms, including the release of the cytokine IL-1β, a major driver of inflammation during infection. This study reveals that T. gondii is able to inhibit IL-1β production in human neutrophils by impairing the activation of the NF-κB signaling pathway and by inhibiting the inflammasome, the protein complex responsible for IL-1β maturation. This two-pronged strategy of targeting the IL-1β pathway may facilitate the survival and spread of T. gondii during acute infection.
Collapse
|
36
|
Azcutia V, Parkos CA, Brazil JC. Role of negative regulation of immune signaling pathways in neutrophil function. J Leukoc Biol 2017; 103:10.1002/JLB.3MIR0917-374R. [PMID: 29345376 PMCID: PMC6203665 DOI: 10.1002/jlb.3mir0917-374r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/03/2017] [Accepted: 11/05/2017] [Indexed: 12/26/2022] Open
Abstract
Polymorphonuclear neutrophils (PMNs) play a critical role in host defense against infection and in the resolution of inflammation. However, immune responses mediated by PMN must be tightly regulated to facilitate elimination of invading pathogens without inducing detrimental inflammation and host tissue damage. Specific engagement of cell surface immunoreceptors by a diverse range of extracellular signals regulates PMN effector functions through differential activation of intracellular signaling cascades. Although mechanisms of PMN activation mediated via cell signaling pathways have been well described, less is known about negative regulation of PMN function by immune signaling cascades. Here, we provide an overview of immunoreceptor-mediated negative regulation of key PMN effector functions including maturation, migration, phagocytosis, reactive oxygen species release, degranulation, apoptosis, and NET formation. Increased understanding of mechanisms of suppression of PMN effector functions may point to possible future therapeutic targets for the amelioration of PMN-mediated autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Veronica Azcutia
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109 USA
| | - Charles A. Parkos
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109 USA
| | - Jennifer C. Brazil
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109 USA
| |
Collapse
|
37
|
Abram CL, Lowell CA. Shp1 function in myeloid cells. J Leukoc Biol 2017; 102:657-675. [PMID: 28606940 DOI: 10.1189/jlb.2mr0317-105r] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 01/28/2023] Open
Abstract
The motheaten mouse was first described in 1975 as a model of systemic inflammation and autoimmunity, as a result of immune system dysregulation. The phenotype was later ascribed to mutations in the cytoplasmic tyrosine phosphatase Shp1. This phosphatase is expressed widely throughout the hematopoietic system and has been shown to impact a multitude of cell signaling pathways. The determination of which cell types contribute to the different aspects of the phenotype caused by global Shp1 loss or mutation and which pathways within these cell types are regulated by Shp1 is important to further our understanding of immune system regulation. In this review, we focus on the role of Shp1 in myeloid cells and how its dysregulation affects immune function, which can impact human disease.
Collapse
Affiliation(s)
- Clare L Abram
- Department of Laboratory Medicine and Immunology Program, University of California, San Francisco, California, USA
| | - Clifford A Lowell
- Department of Laboratory Medicine and Immunology Program, University of California, San Francisco, California, USA
| |
Collapse
|
38
|
Zimmermann W, Kammerer R. Coevolution of paired receptors in Xenopus carcinoembryonic antigen-related cell adhesion molecule families suggests appropriation as pathogen receptors. BMC Genomics 2016; 17:928. [PMID: 27852220 PMCID: PMC5112662 DOI: 10.1186/s12864-016-3279-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 11/09/2016] [Indexed: 02/08/2023] Open
Abstract
Background In mammals, CEACAM1 and closely related members represent paired receptors with similar extracellular ligand-binding regions and cytoplasmic domains with opposing functions. Human CEACAM1 and CEACAM3 which have inhibitory ITIM/ITSM and activating ITAM-like motifs, respectively, in their cytoplasmic regions are such paired receptors. Various bacterial pathogens bind to CEACAM1 on epithelial and immune cells facilitating both entry into the host and down-regulation of the immune response whereas interaction with granulocyte-specific CEACAM3 leads to their uptake and destruction. It is unclear whether paired CEACAM receptors also exist in other vertebrate clades. Results We identified more than 80 ceacam genes in Xenopus tropicalis and X. laevis. They consist of two subgroups containing one or two putative paired receptor pairs each. Analysis of genomic sequences of paired receptors provide evidence that their highly similar ligand binding domains were adjusted by recent gene conversion events. In contrast, selection for diversification is observed among inhibitory receptor orthologs of the two frogs which split some 60 million years ago. The allotetraploid X. laevis arose later by hybridization of two closely related species. Interestingly, despite the conservation of the genomic landscape surrounding the homeologous ceacam loci only one locus resembles the one found in X. tropicalis. From the second X. laevis locus more than 80 % of the ceacam genes were lost including 5 of the 6 paired receptor genes. This suggests that once the gene for one of the paired receptors is lost the remaining gene cluster degrades rapidly probably due to lack of selection pressure exerted by pathogens. Conclusions The presence of paired receptors and selection for diversification suggests that also in amphibians CEACAM1-related inhibitory proteins are or were used as pathogen receptors. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3279-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wolfgang Zimmermann
- Tumor Immunology Laboratory, LIFE Center, University Clinic, Ludwig-Maximilians-University, Feodor-Lynen-Str. 19, 81377, Munich, Germany. .,Department of Urology, University Clinic, Ludwig-Maximilians-University, Marchioninistr. 15, 81377, Munich, Germany.
| | - Robert Kammerer
- Institute of Immunology, Friedrich-Loeffler Institut, 17493, Greifswald-Insel Riems, Germany
| |
Collapse
|
39
|
Florian W, Lenfert E, Gerstel D, von Ehrenstein L, Einhoff J, Schmidt G, Logsdon M, Brandner J, Tiegs G, Beauchemin N, Wagener C, Deppert W, Horst AK. CEACAM1 controls the EMT switch in murine mammary carcinoma in vitro and in vivo. Oncotarget 2016; 7:63730-63746. [PMID: 27572314 PMCID: PMC5325399 DOI: 10.18632/oncotarget.11650] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 08/08/2016] [Indexed: 12/29/2022] Open
Abstract
We analyzed the molecular basis for carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1)-controlled inhibition of epithelial-mesenchymal transition (EMT) in a mouse model for mammary adenocarcinoma (WAP-T mice). We demonstrate that silencing of CEACAM1 in WAP-T tumor-derived G-2 cells induces epithelial-mesenchymal plasticity (EMP), as evidenced by typical changes of gene expression, morphology and increased invasion. In contrast, reintroduction of CEACAM1 into G-2 cells reversed up-regulation of genes imposing mesenchymal transition, as well as cellular invasion. We identified the Wnt-pathway as target for CEACAM1-mediated repression of EMT. Importantly, β-catenin phosphorylation status and transcriptional activity strongly depend on CEACAM1 expression: CEACAM1high G-2 cells displayed enhanced phosphorylation of β-catenin at S33/S37/T41 and decreased phosphorylation at Y86, thereby inhibiting canonical Wnt/β-catenin signaling. We identified Src-homology 2 domain-containing phosphatase 2 (SHP-2) as a critical binding partner of CEACAM1 that could modulate β-catenin Y86 phosphorylation. Hence, CEACAM1 serves as a scaffold that controls membrane proximal β-catenin signaling. In vivo, mammary tumors of WAP-T/CEACAM1null mice displayed increased nuclear translocation of β-catenin and a dramatically enhanced metastasis rate compared to WAP-T mice. Hence, CEACAM1 controls EMT in vitro and in vivo by site-specific regulation of β-catenin phosphorylation. Survival analyses of human mammary carcinoma patients corroborated these data, indicating that CEACAM1 is a prognostic marker for breast cancer survival.
Collapse
Affiliation(s)
- Wegwitz Florian
- Clinic for General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Georg-August-University of Göttingen, D-37077 Göttingen, Germany
- Institute for Tumor Biology, University Medical Center-Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Eva Lenfert
- Institute for Tumor Biology, University Medical Center-Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Daniela Gerstel
- Center for Diagnostics, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Lena von Ehrenstein
- Institute for Tumor Biology, University Medical Center-Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Julia Einhoff
- Institute for Tumor Biology, University Medical Center-Hamburg-Eppendorf, D-20251 Hamburg, Germany
- Pharmaceutical Institute, Christian-Albrechts-University Kiel, D-24118 Kiel, Germany
| | - Geske Schmidt
- Clinic for General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Georg-August-University of Göttingen, D-37077 Göttingen, Germany
| | - Matthew Logsdon
- Clinic for General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Georg-August-University of Göttingen, D-37077 Göttingen, Germany
| | - Johanna Brandner
- Dermatology and Venerology Department and Clinic, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Gisa Tiegs
- Institute for Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Nicole Beauchemin
- Goodman Cancer Research Centre and Departments of Biochemistry, Medicine and Oncology, McGill University, Montreal, H3G1Y6, Canada
| | - Christoph Wagener
- Center for Diagnostics, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Wolfgang Deppert
- Institute for Tumor Biology, University Medical Center-Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Andrea Kristina Horst
- Institute for Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| |
Collapse
|
40
|
van Rees DJ, Szilagyi K, Kuijpers TW, Matlung HL, van den Berg TK. Immunoreceptors on neutrophils. Semin Immunol 2016; 28:94-108. [PMID: 26976825 PMCID: PMC7129252 DOI: 10.1016/j.smim.2016.02.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 02/24/2016] [Accepted: 02/26/2016] [Indexed: 12/12/2022]
Abstract
Neutrophil activities must be tightly controlled to maintain immune homeostasis. Activating and inhibitory receptors balance the outcome of immune cell activation. Immunoreceptors contain Ig-like extracellular domains and signal via ITAMs or ITIMs. Syk or SHP/SHIP mediate downstream signaling after immunoreceptor activation. Targeting immunoreceptors provides opportunities for therapeutic interventions.
Neutrophils play a critical role in the host defense against infection, and they are able to perform a variety of effector mechanisms for this purpose. However, there are also a number of pathological conditions, including autoimmunity and cancer, in which the activities of neutrophils can be harmful to the host. Thus the activities of neutrophils need to be tightly controlled. As in the case of other immune cells, many of the neutrophil effector functions are regulated by a series of immunoreceptors on the plasma membrane. Here, we review what is currently known about the functions of the various individual immunoreceptors and their signaling in neutrophils. While these immunoreceptors allow for the recognition of a diverse range of extracellular ligands, such as cell surface structures (like proteins, glycans and lipids) and extracellular matrix components, they commonly signal via conserved ITAM or ITIM motifs and their associated downstream pathways that depend on the phosphorylation of tyrosine residues in proteins and/or inositol lipids. This allows for a balanced homeostatic regulation of neutrophil effector functions. Given the number of available immunoreceptors and their fundamental importance for neutrophil behavior, it is perhaps not surprising that pathogens have evolved means to evade immune responses through some of these pathways. Inversely, some of these receptors evolved to specifically recognize these pathogens. Finally, some interactions mediated by immunoreceptors in neutrophils have been identified as promising targets for therapeutic intervention.
Collapse
Affiliation(s)
- Dieke J van Rees
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Katka Szilagyi
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Taco W Kuijpers
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Hanke L Matlung
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Timo K van den Berg
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
41
|
Pathogenesis of human diffusely adhering Escherichia coli expressing Afa/Dr adhesins (Afa/Dr DAEC): current insights and future challenges. Clin Microbiol Rev 2015; 27:823-69. [PMID: 25278576 DOI: 10.1128/cmr.00036-14] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The pathogenicity and clinical pertinence of diffusely adhering Escherichia coli expressing the Afa/Dr adhesins (Afa/Dr DAEC) in urinary tract infections (UTIs) and pregnancy complications are well established. In contrast, the implication of intestinal Afa/Dr DAEC in diarrhea is still under debate. These strains are age dependently involved in diarrhea in children, are apparently not involved in diarrhea in adults, and can also be asymptomatic intestinal microbiota strains in children and adult. This comprehensive review analyzes the epidemiology and diagnosis and highlights recent progress which has improved the understanding of Afa/Dr DAEC pathogenesis. Here, I summarize the roles of Afa/Dr DAEC virulence factors, including Afa/Dr adhesins, flagella, Sat toxin, and pks island products, in the development of specific mechanisms of pathogenicity. In intestinal epithelial polarized cells, the Afa/Dr adhesins trigger cell membrane receptor clustering and activation of the linked cell signaling pathways, promote structural and functional cell lesions and injuries in intestinal barrier, induce proinflammatory responses, create angiogenesis, instigate epithelial-mesenchymal transition-like events, and lead to pks-dependent DNA damage. UTI-associated Afa/Dr DAEC strains, following adhesin-membrane receptor cell interactions and activation of associated lipid raft-dependent cell signaling pathways, internalize in a microtubule-dependent manner within urinary tract epithelial cells, develop a particular intracellular lifestyle, and trigger a toxin-dependent cell detachment. In response to Afa/Dr DAEC infection, the host epithelial cells generate antibacterial defense responses. Finally, I discuss a hypothetical role of intestinal Afa/Dr DAEC strains that can act as "silent pathogens" with the capacity to emerge as "pathobionts" for the development of inflammatory bowel disease and intestinal carcinogenesis.
Collapse
|
42
|
Van Avondt K, van Sorge NM, Meyaard L. Bacterial immune evasion through manipulation of host inhibitory immune signaling. PLoS Pathog 2015; 11:e1004644. [PMID: 25742647 PMCID: PMC4351076 DOI: 10.1371/journal.ppat.1004644] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Kristof Van Avondt
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nina M. van Sorge
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Linde Meyaard
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
43
|
CEA a thrombus CAM: CEACAM2, a twin of CEACAM1? Blood 2015; 124:2323-4. [PMID: 25301335 DOI: 10.1182/blood-2014-08-594101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this issue of Blood, Alshahrani et al demonstrate that carcinoembryonic antigen-related cell adhesion molecule 2 (CEACAM2) is expressed on platelets and negatively regulates the collagen receptor glycoprotein (GP)VI-FcRγ chain and C-type lectin-like receptor 2 (CLEC-2)-mediated platelet activation.
Collapse
|
44
|
Khairnar V, Duhan V, Maney SK, Honke N, Shaabani N, Pandyra AA, Seifert M, Pozdeev V, Xu HC, Sharma P, Baldin F, Marquardsen F, Merches K, Lang E, Kirschning C, Westendorf AM, Häussinger D, Lang F, Dittmer U, Küppers R, Recher M, Hardt C, Scheffrahn I, Beauchemin N, Göthert JR, Singer BB, Lang PA, Lang KS. CEACAM1 induces B-cell survival and is essential for protective antiviral antibody production. Nat Commun 2015; 6:6217. [PMID: 25692415 PMCID: PMC4346637 DOI: 10.1038/ncomms7217] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 01/07/2015] [Indexed: 01/03/2023] Open
Abstract
B cells are essential for antiviral immune defence because they produce neutralizing antibodies, present antigen and maintain the lymphoid architecture. Here we show that intrinsic signalling of CEACAM1 is essential for generating efficient B-cell responses. Although CEACAM1 exerts limited influence on the proliferation of B cells, expression of CEACAM1 induces survival of proliferating B cells via the BTK/Syk/NF-κB-axis. The absence of this signalling cascade in naive Ceacam1−/− mice limits the survival of B cells. During systemic infection with cytopathic vesicular stomatitis virus, Ceacam1−/− mice can barely induce neutralizing antibody responses and die early after infection. We find, therefore, that CEACAM1 is a crucial regulator of B-cell survival, influencing B-cell numbers and protective antiviral antibody responses. Antibody responses are regulated by selective survival of B cells with proper antigen specificity. Here the authors show that CEACAM1 is critical for B-cell survival during homeostasis and antiviral responses.
Collapse
Affiliation(s)
- Vishal Khairnar
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Vikas Duhan
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Sathish Kumar Maney
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Nadine Honke
- 1] Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany [2] Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Namir Shaabani
- 1] Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany [2] Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Aleksandra A Pandyra
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Marc Seifert
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Virchowstrasse 173, Essen 45122, Germany
| | - Vitaly Pozdeev
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Haifeng C Xu
- 1] Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany [2] Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Piyush Sharma
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Fabian Baldin
- Clinic for Primary Immunodeficiency, Medical Outpatient Unit and Immunodeficiency Laboratory, Department of Biomedicine, University Hospital, Basel 4031, Switzerland
| | - Florian Marquardsen
- Clinic for Primary Immunodeficiency, Medical Outpatient Unit and Immunodeficiency Laboratory, Department of Biomedicine, University Hospital, Basel 4031, Switzerland
| | - Katja Merches
- 1] Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany [2] Department of Physiology I, University of Tuebingen, Gmelinstrasse 5, Tuebingen 72076, Germany
| | - Elisabeth Lang
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Carsten Kirschning
- Institute of Medical Microbiology, Faculty of Medicine, University Hospital Essen, Hufelandstrasse 55, Essen 45122, Germany
| | - Astrid M Westendorf
- Institute of Medical Microbiology, Faculty of Medicine, University Hospital Essen, Hufelandstrasse 55, Essen 45122, Germany
| | - Dieter Häussinger
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Florian Lang
- Department of Physiology I, University of Tuebingen, Gmelinstrasse 5, Tuebingen 72076, Germany
| | - Ulf Dittmer
- Institute of Virology, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Ralf Küppers
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Virchowstrasse 173, Essen 45122, Germany
| | - Mike Recher
- Clinic for Primary Immunodeficiency, Medical Outpatient Unit and Immunodeficiency Laboratory, Department of Biomedicine, University Hospital, Basel 4031, Switzerland
| | - Cornelia Hardt
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Inka Scheffrahn
- Clinic of Gastroenterology and Hepatology, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Nicole Beauchemin
- Rosalind and Morris Goodman Cancer Centre, Departments of Biochemistry, Medicine and Oncology, McIntyre Medical Science Building, Montreal, Quebec, Canada H3G 1Y6
| | - Joachim R Göthert
- Department of Hematology, West German Cancer Center (WTZ), University Hospital Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Bernhard B Singer
- Institute of Anatomy, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Philipp A Lang
- 1] Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany [2] Department of Molecular Medicine II, Heinrich Heine University Düsseldorf, Universitätsstrasse 1, Düsseldorf 40225, Germany
| | - Karl S Lang
- 1] Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany [2] Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| |
Collapse
|
45
|
Brungs S, Kolanus W, Hemmersbach R. Syk phosphorylation - a gravisensitive step in macrophage signalling. Cell Commun Signal 2015; 13:9. [PMID: 25644261 PMCID: PMC4326470 DOI: 10.1186/s12964-015-0088-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 01/20/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The recognition of pathogen patterns followed by the production of reactive oxygen species (ROS) during the oxidative burst is one of the major functions of macrophages. This process is the first line of defence and is crucial for the prevention of pathogen-associated diseases. There are indications that the immune system of astronauts is impaired during spaceflight, which could result in an increased susceptibility to infections. Several studies have indicated that the oxidative burst of macrophages is highly impaired after spaceflight, but the underlying mechanism remained to be elucidated. Here, we investigated the characteristics of reactive oxygen species production during the oxidative burst after pathogen pattern recognition in simulated microgravity by using a fast-rotating Clinostat to mimic the condition of microgravity. Furthermore, spleen tyrosine kinase (Syk) phosphorylation, which is required for ROS production, and the translocation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) to the nucleus were monitored to elucidate the influence of altered gravity on macrophage signalling. RESULTS Simulated microgravity leads to significantly diminished ROS production in macrophages upon zymosan, curdlan and lipopolysaccharide stimulation. To address the signalling mechanisms involved, Syk phosphorylation was examined, revealing significantly reduced phosphorylation in simulated microgravity compared to normal gravity (1 g) conditions. In contrast, a later signalling step, the translocation of NF-κB to the nucleus, demonstrated no gravity-dependent alterations. CONCLUSIONS The results obtained in simulated microgravity show that ROS production in macrophages is a highly gravisensitive process, caused by a diminished Syk phosphorylation. In contrast, NF-κB signalling remains consistent in simulated microgravity. This difference reveals that early signalling steps, such as Syk phosphorylation, are affected by microgravity, whereas the lack of effects in later steps might indicate adaptation processes. Taken together, this study clearly demonstrates that macrophages display impaired signalling upon pattern recognition when exposed to simulated microgravity conditions, which if verified in real microgravity this may be one reason why astronauts display higher susceptibility to infections.
Collapse
Affiliation(s)
- Sonja Brungs
- Biomedical Research Institute of Aerospace Medicine, German Aerospace Center (DLR), Linder Hoehe, 51147, Koeln, Germany.
| | - Waldemar Kolanus
- Molecular Immunology, LIMES Institute, University of Bonn, Carl-Troll Str. 31, 53115, Bonn, Germany.
| | - Ruth Hemmersbach
- Biomedical Research Institute of Aerospace Medicine, German Aerospace Center (DLR), Linder Hoehe, 51147, Koeln, Germany.
| |
Collapse
|
46
|
Wu NL, Huang DY, Tsou HN, Lin YC, Lin WW. Syk Mediates IL−17-Induced CCL20 Expression by Targeting Act1-Dependent K63-Linked Ubiquitination of TRAF6. J Invest Dermatol 2015; 135:490-498. [DOI: 10.1038/jid.2014.383] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 08/19/2014] [Accepted: 08/22/2014] [Indexed: 12/21/2022]
|
47
|
Kigawa Y, Miyazaki T, Lei XF, Nakamachi T, Oguchi T, Kim-Kaneyama JR, Taniyama M, Tsunawaki S, Shioda S, Miyazaki A. NADPH oxidase deficiency exacerbates angiotensin II-induced abdominal aortic aneurysms in mice. Arterioscler Thromb Vasc Biol 2014; 34:2413-20. [PMID: 25189573 DOI: 10.1161/atvbaha.114.303086] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Although nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2) is reportedly essential for phagocyte host defenses, it has been found to aggravate atherosclerosis in apolipoprotein E (Apoe)-null mice through excess production of superoxide. We therefore assessed the role of NOX2 in an experimental model of abdominal aortic aneurysm (AAA) and assessed the mechanism of NOX2 action in AAA. APPROACH AND RESULTS AAA was induced in low-density lipoprotein receptor-null (Ldlr(-/-)) mice by infusing angiotensin II. Nox2 expression was elevated in the abdominal aortae of these mice during infusion of angiotensin II, with enhanced Nox2 expression mainly because of the recruitment of NOX2-enriched macrophages into AAA lesions. Unexpectedly, systemic Nox2 deficiency promoted AAA development but reduced the level of reactive oxygen species in AAA lesions. Nox2 deficiency stimulated macrophage conversion toward the M1 subset, enhancing expression of interleukin (IL)-1β and matrix metalloproteinase-9/12 mRNA. Administration of neutralizing antibody against IL-1β abolished AAA development in Nox2-deficient mice. Bone marrow transplantation experiments revealed that AAA aggravation by Nox2 deficiency is because of bone marrow-derived cells. Isolated bone marrow-derived macrophages from Nox2-null mice could not generate reactive oxygen species. In contrast, IL-1β expression in peritoneal and bone marrow-derived macrophages, but not in peritoneal neutrophils, was substantially enhanced by Nox2 deficiency. Pharmacological inhibition of Janus kinase/signal transducers and activators of transcription signaling inhibited excess IL-1β expression in Nox2-deficient macrophages, whereas matrix metalloproteinase-9 secretion was constitutively stimulated via nuclear factor-κB signals. CONCLUSIONS Nox2 deficiency enhances macrophage secretion of IL-1β and matrix metalloproteinase-9, disrupting tissue-remodeling functions in AAA lesions. These actions are unfavorable if NOX2 is to serve as a molecular target for AAA.
Collapse
Affiliation(s)
- Yasuyoshi Kigawa
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.)
| | - Takuro Miyazaki
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.).
| | - Xiao-Feng Lei
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.)
| | - Tomoya Nakamachi
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.)
| | - Tatsunori Oguchi
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.)
| | - Joo-ri Kim-Kaneyama
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.)
| | - Matsuo Taniyama
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.)
| | - Shohko Tsunawaki
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.)
| | - Seiji Shioda
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.)
| | - Akira Miyazaki
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.)
| |
Collapse
|
48
|
Tyrosine phosphorylation in Toll-like receptor signaling. Cytokine Growth Factor Rev 2014; 25:533-41. [PMID: 25022196 DOI: 10.1016/j.cytogfr.2014.06.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 06/16/2014] [Indexed: 12/28/2022]
Abstract
There is a wealth of knowledge about how different Ser/Thr protein kinases participate in Toll-like receptor (TLR) signaling. In many cases, we know the identities of the Ser/Thr residues of various components of the TLR-signaling pathways that are phosphorylated, the functional consequences of the phosphorylation and the responsible protein kinases. In contrast, the analysis of Tyr-phosphorylation of TLRs and their signaling proteins is currently incomplete, because several existing analyses are not systematic or they do not rely on robust experimental data. Nevertheless, it is clear that many TLRs require, for signaling, ligand-dependent phosphorylation of specific Tyr residues in their cytoplasmic domains; the list includes TLR2, TLR3, TLR4, TLR5, TLR8 and TLR9. In this article, we discuss the current status of knowledge of the effect of Tyr-phosphorylation of TLRs and their signaling proteins on their biochemical and biological functions, the possible identities of the relevant protein tyrosine kinases (PTKs) and the nature of regulations of PTK-mediated activation of TLR signaling pathways.
Collapse
|
49
|
Soluble CEACAM8 interacts with CEACAM1 inhibiting TLR2-triggered immune responses. PLoS One 2014; 9:e94106. [PMID: 24743304 PMCID: PMC3990526 DOI: 10.1371/journal.pone.0094106] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 03/12/2014] [Indexed: 12/21/2022] Open
Abstract
Lower respiratory tract bacterial infections are characterized by neutrophilic inflammation in the airways. The carcinoembryonic antigen-related cell adhesion molecule (CEACAM) 8 is expressed in and released by human granulocytes. Our study demonstrates that human granulocytes release CEACAM8 in response to bacterial DNA in a TLR9-dependent manner. Individuals with a high percentage of bronchial lavage fluid (BALF) granulocytes were more likely to have detectable levels of released CEACAM8 in the BALF than those with a normal granulocyte count. Soluble, recombinant CEACAM8-Fc binds to CEACAM1 expressed on human airway epithelium. Application of CEACAM8-Fc to CEACAM1-positive human pulmonary epithelial cells resulted in reduced TLR2-dependent inflammatory responses. These inhibitory effects were accompanied by tyrosine phosphorylation of the immunoreceptor tyrosine-based inhibitory motif (ITIM) of CEACAM1 and by recruitment of the phosphatase SHP-1, which could negatively regulate Toll-like receptor 2-dependent activation of the phosphatidylinositol 3-OH kinase-Akt kinase pathway. Our results suggest a new mechanism by which granulocytes reduce pro-inflammatory immune responses in human airways via secretion of CEACAM8 in neutrophil-driven bacterial infections.
Collapse
|
50
|
Chargui A, El May MV. Autophagy mediates neutrophil responses to bacterial infection. APMIS 2014; 122:1047-58. [PMID: 24735202 DOI: 10.1111/apm.12271] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 02/13/2014] [Indexed: 12/30/2022]
Abstract
Neutrophils constitute the first line of cellular defense against pathogens and autophagy is a fundamental cellular homeostasis pathway that operates with the intracellular degradation/recycling system. Induction of the autophagic process in neutrophils, in response to invading pathogens, constitutes a crucial mechanism in innate immunity. Exploration of autophagy has greatly progressed and diverse strategies have been reported for studying this molecular process in different biological systems; especially in infectious and inflammatory diseases. Furthermore, the role of autophagy in neutrophils, during pathogenic infection, continues to be of interest, due to the role of the cell in immunity function, its recruitment to the site of infection and its implication in inflammatory diseases. This review focuses on the known role of autophagy in neutrophils defence against pathogenic infections. A more detailed discussion will concern the recent findings highlighting the role of autophagy in inflammation and cell death in infected neutrophils.
Collapse
Affiliation(s)
- Abderrahman Chargui
- Laboratory of Histology, Embryology and Cell Biology, Faculty of Medicine, Tunis, Tunisia; Higher School of Agriculture, Kef, Tunisia
| | | |
Collapse
|