1
|
Hintermann E, Tondello C, Fuchs S, Bayer M, Pfeilschifter JM, Taubert R, Mollenhauer M, Elferink RPJO, Manns MP, Christen U. Blockade of neutrophil extracellular trap components ameliorates cholestatic liver disease in Mdr2 (Abcb4) knockout mice. J Autoimmun 2024; 146:103229. [PMID: 38653165 DOI: 10.1016/j.jaut.2024.103229] [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: 12/27/2023] [Revised: 03/21/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
Primary sclerosing cholangitis (PSC) is an (auto)immune-mediated cholestatic liver disease with a yet unclear etiology. Increasing evidence points to an involvement of neutrophils in chronic liver inflammation and cirrhosis but also liver repair. Here, we investigate the role of the neutrophil extracellular trap (NET) component myeloperoxidase (MPO) and the therapeutic potential of DNase I and of neutrophil elastase (NE) inhibitor GW311616A on disease outcome in the multidrug resistance 2 knockout (Mdr2-/-) mouse, a PSC animal model. Initially, we observed the recruitment of MPO expressing cells and the formation of NETs in liver biopsies of PSC patients and in Mdr2-/- livers. Furthermore, sera of Mdr2-/- mice contained perinuclear anti-neutrophil cytoplasmic antibody (p-ANCA)-like reactivity similar to PSC patient sera. Also, hepatic NE activity was significantly higher in Mdr2-/- mice than in wild type littermates. Flow cytometry analyses revealed that during disease development a highly active neutrophil subpopulation established specifically in the liver of Mdr2-/- mice. However, absence of their MPO activity, as in MPO-deficient Mdr2-/- mice, showed no effect on hepatobiliary disease severity. In contrast, clearance of extracellular DNA by DNase I reduced the frequency of liver-resident neutrophils, plasmacytoid dendritic cells (pDCs) and CD103+ conventional DCs and decreased cholangiocyte injury. Combination of DNase I with a pDC-depleting antibody was additionally hepatocyte-protective. Most importantly, GW311616A, an orally bioavailable inhibitor of human NE, attenuated hepatobiliary injury in a TNFα-dependent manner and damped hyperproliferation of biliary epithelial cells. Further, hepatic immigration and activity of CD11b+ DCs as well as the secretion of IFNγ by hepatic CD4 and CD8 T cells were reduced. Our findings delineate neutrophils as important participants in the immune cell crosstalk that drives cholestatic liver disease and identify NET components as potential therapeutic targets.
Collapse
Affiliation(s)
- Edith Hintermann
- Pharmazentrum Frankfurt / ZAFES, Goethe University, University Hospital Frankfurt, Frankfurt am Main, Germany.
| | - Camilla Tondello
- Pharmazentrum Frankfurt / ZAFES, Goethe University, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Sina Fuchs
- Pharmazentrum Frankfurt / ZAFES, Goethe University, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Monika Bayer
- Pharmazentrum Frankfurt / ZAFES, Goethe University, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Josef M Pfeilschifter
- Pharmazentrum Frankfurt / ZAFES, Goethe University, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Richard Taubert
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Germany
| | - Martin Mollenhauer
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Roland P J Oude Elferink
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Germany
| | - Michael P Manns
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Germany
| | - Urs Christen
- Pharmazentrum Frankfurt / ZAFES, Goethe University, University Hospital Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
2
|
Tatsiy O, Mayer TZ, de Carvalho Oliveira V, Sylvain-Prévost S, Isabel M, Dubois CM, McDonald PP. Cytokine Production and NET Formation by Monosodium Urate-Activated Human Neutrophils Involves Early and Late Events, and Requires Upstream TAK1 and Syk. Front Immunol 2020; 10:2996. [PMID: 32010124 PMCID: PMC6974451 DOI: 10.3389/fimmu.2019.02996] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 12/05/2019] [Indexed: 01/08/2023] Open
Abstract
Gout is a prevalent and incapacitating disease triggered by the deposition of monosodium urate (MSU) crystals in joints, which are also massively infiltrated by neutrophils. The interaction of the latter with MSU crystals triggers several responses, including the generation of inflammatory mediators and of neutrophil extracellular traps (NETs). Though some of the signaling events mobilized by MSU in neutrophils have been described (e.g., Src family kinases, Syk, PKC, PI3K), the picture remains fragmentary. Likewise, the impact of these signaling events on cellular responses is incompletely understood. In this study, we examined transcriptomic changes triggered by MSU in neutrophils and their impact on the corresponding proteins, as well as the role of various signaling pathways in prominent functional responses. We report for the first time that neutrophils can secrete the monocyte chemoattractant, CCL4, in response to MSU. Accordingly, we found that transcription factors NF-κB, CREB, and C/EBP are belatedly activated by MSU crystals, and at least the former is involved in chemokine generation. Moreover, we show that MAPKs and Akt are activated by MSU in neutrophils, that they are under the control of TAK1 and Syk, and that they participate in cytokine generation and NETosis. In the latter instance, we found the phenomenon to be independent of endogenous ROS, but under the control of PAD4. We finally provide evidence that endogenous factors contribute to the belated phosphorylation of kinases and transcription factors in response to MSU. Collectively, our findings unveil potentially important therapeutic targets for gouty arthritis.
Collapse
Affiliation(s)
- Olga Tatsiy
- Pulmonary Division, Faculty of Medicine, Université de Sherbrooke and Centre de recherche du CHUS (CRCHUS), Sherbrooke, QC, Canada.,Department of Immunology and Cell Biology, Faculty of Medicine, Université de Sherbrooke and CRCHUS, Sherbrooke, QC, Canada
| | - Thomas Z Mayer
- Pulmonary Division, Faculty of Medicine, Université de Sherbrooke and Centre de recherche du CHUS (CRCHUS), Sherbrooke, QC, Canada.,Department of Immunology and Cell Biology, Faculty of Medicine, Université de Sherbrooke and CRCHUS, Sherbrooke, QC, Canada
| | - Vanessa de Carvalho Oliveira
- Pulmonary Division, Faculty of Medicine, Université de Sherbrooke and Centre de recherche du CHUS (CRCHUS), Sherbrooke, QC, Canada.,Department of Immunology and Cell Biology, Faculty of Medicine, Université de Sherbrooke and CRCHUS, Sherbrooke, QC, Canada
| | - Stéphanie Sylvain-Prévost
- Pulmonary Division, Faculty of Medicine, Université de Sherbrooke and Centre de recherche du CHUS (CRCHUS), Sherbrooke, QC, Canada
| | - Marilyn Isabel
- Pulmonary Division, Faculty of Medicine, Université de Sherbrooke and Centre de recherche du CHUS (CRCHUS), Sherbrooke, QC, Canada
| | - Claire M Dubois
- Department of Immunology and Cell Biology, Faculty of Medicine, Université de Sherbrooke and CRCHUS, Sherbrooke, QC, Canada
| | - Patrick P McDonald
- Pulmonary Division, Faculty of Medicine, Université de Sherbrooke and Centre de recherche du CHUS (CRCHUS), Sherbrooke, QC, Canada
| |
Collapse
|
3
|
Fonseca Z, Díaz-Godínez C, Mora N, Alemán OR, Uribe-Querol E, Carrero JC, Rosales C. Entamoeba histolytica Induce Signaling via Raf/MEK/ERK for Neutrophil Extracellular Trap (NET) Formation. Front Cell Infect Microbiol 2018; 8:226. [PMID: 30023352 PMCID: PMC6039748 DOI: 10.3389/fcimb.2018.00226] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 06/15/2018] [Indexed: 12/25/2022] Open
Abstract
Amoebiasis, the disease caused by Entamoeba histolytica is the third leading cause of human deaths among parasite infections. E. histolytica was reported associated with around 100 million cases of amoebic dysentery, colitis and amoebic liver abscess that lead to almost 50,000 fatalities worldwide in 2010. E. histolytica infection is associated with the induction of inflammation characterized by a large number of infiltrating neutrophils. These neutrophils have been implicated in defense against this parasite, by mechanisms not completely described. The neutrophil antimicrobial mechanisms include phagocytosis, degranulation, and formation of neutrophil extracellular traps (NETs). Recently, our group reported that NETs are also produced in response to E. histolytica trophozoites. But, the mechanism for NETs induction remains unknown. In this report we explored the possibility that E. histolytica leads to NETs formation via a signaling pathway similar to the pathways activated by PMA or the Fc receptor FcγRIIIb. Neutrophils were stimulated by E. histolytica trophozoites and the effect of various pharmacological inhibitors on amoeba-induced NETs formation was assessed. Selective inhibitors of Raf, MEK, and NF-κB prevented E. histolytica-induced NET formation. In contrast, inhibitors of PKC, TAK1, and NADPH-oxidase did not block E. histolytica-induced NETs formation. E. histolytica induced phosphorylation of ERK in a Raf and MEK dependent manner. These data show that E. histolytica activates a signaling pathway to induce NETs formation, that involves Raf/MEK/ERK, but it is independent of PKC, TAK1, and reactive oxygen species (ROS). Thus, amoebas activate neutrophils via a different pathway from the pathways activated by PMA or the IgG receptor FcγRIIIb.
Collapse
Affiliation(s)
- Zayda Fonseca
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - César Díaz-Godínez
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Nancy Mora
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Omar R Alemán
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Eileen Uribe-Querol
- División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Julio C Carrero
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
4
|
Mahon OR, Dunne A. Disease-Associated Particulates and Joint Inflammation; Mechanistic Insights and Potential Therapeutic Targets. Front Immunol 2018; 9:1145. [PMID: 29892292 PMCID: PMC5985611 DOI: 10.3389/fimmu.2018.01145] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 05/07/2018] [Indexed: 12/27/2022] Open
Abstract
It is now well established that intra-articular deposition of endogenous particulates, such as osteoarthritis-associated basic calcium phosphate crystals, gout-associated monosodium urate crystals, and calcium deposition disease-associated calcium pyrophosphate crystals, contributes to joint destruction through the production of cartilage-degrading enzymes and pro-inflammatory cytokines. Furthermore, exogenous wear-debris particles, generated from prosthetic implants, drive periprosthetic osteolysis which impacts on the longevity of total joint replacements. Over the last few years, significant insight has been gained into the mechanisms through which these particulates exert their effects. Not only has this increased our understanding of the pathological processes associated with crystal deposition but it has also led to the identification of a number of therapeutic targets to treat particulate-associated disease. In this review, we discuss recent developments regarding the cellular events triggered by joint-associated particulates, as well as future directions in therapy for particulate-related arthropathies.
Collapse
Affiliation(s)
- Olwyn R Mahon
- School of Biochemistry and Immunology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, The University of Dublin, Dublin, Ireland
| | - Aisling Dunne
- School of Biochemistry and Immunology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, The University of Dublin, Dublin, Ireland
| |
Collapse
|
5
|
Alemán OR, Mora N, Cortes-Vieyra R, Uribe-Querol E, Rosales C. Transforming Growth Factor-β-Activated Kinase 1 Is Required for Human FcγRIIIb-Induced Neutrophil Extracellular Trap Formation. Front Immunol 2016; 7:277. [PMID: 27486461 PMCID: PMC4947870 DOI: 10.3389/fimmu.2016.00277] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 07/04/2016] [Indexed: 01/24/2023] Open
Abstract
Neutrophils (PMNs) are the most abundant leukocytes in the blood. PMN migrates from the circulation to sites of infection where they are responsible for antimicrobial functions. PMN uses phagocytosis, degranulation, and formation of neutrophil extracellular traps (NETs) to kill microbes. Several stimuli, including bacteria, fungi, and parasites, and some pharmacological compounds, such as Phorbol 12-myristate 13-acetate (PMA), are efficient inducers of NETs. Antigen-antibody complexes are also capable of inducing NET formation. Recently, it was reported that FcγRIIIb cross-linking induced NET formation similarly to PMA stimulation. Direct cross-linking of FcγRIIA or integrins did not promote NET formation. FcγRIIIb-induced NET formation presented different kinetics from PMA-induced NET formation, suggesting differences in signaling. Because FcγRIIIb also induces a strong activation of extracellular signal-regulated kinase (ERK) and nuclear factor Elk-1, and the transforming growth factor-β-activated kinase 1 (TAK1) has recently been implicated in ERK signaling, in the present report, we explored the role of TAK1 in the signaling pathway activated by FcγRIIIb leading to NET formation. FcγRIIIb was stimulated by specific monoclonal antibodies, and NET formation was evaluated in the presence or absence of pharmacological inhibitors. The antibiotic LL Z1640-2, a selective inhibitor of TAK1 prevented FcγRIIIb-induced, but not PMA-induced NET formation. Both PMA and FcγRIIIb cross-linking induced phosphorylation of ERK. But, LL Z1640-2 only inhibited the FcγRIIIb-mediated activation of ERK. Also, only FcγRIIIb, similarly to transforming growth factor-β-induced TAK1 phosphorylation. A MEK (ERK kinase)-specific inhibitor was able to prevent ERK phosphorylation induced by both PMA and FcγRIIIb. These data show for the first time that FcγRIIIb cross-linking activates TAK1, and that this kinase is required for triggering the MEK/ERK signaling pathway to NETosis.
Collapse
Affiliation(s)
- Omar Rafael Alemán
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Nancy Mora
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Ricarda Cortes-Vieyra
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Eileen Uribe-Querol
- División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
6
|
Alemán OR, Mora N, Cortes-Vieyra R, Uribe-Querol E, Rosales C. Differential Use of Human Neutrophil Fcγ Receptors for Inducing Neutrophil Extracellular Trap Formation. J Immunol Res 2016; 2016:2908034. [PMID: 27034964 PMCID: PMC4806689 DOI: 10.1155/2016/2908034] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/11/2015] [Accepted: 12/13/2015] [Indexed: 12/19/2022] Open
Abstract
Neutrophils (PMN) are the most abundant leukocytes in the blood. PMN migrate from the circulation to sites of infection, where they are responsible for antimicrobial functions. PMN use phagocytosis, degranulation, and formation of neutrophil extracellular traps (NETs) to kill microbes. NETs are fibers composed of chromatin and neutrophil-granule proteins. Several pathogens, including bacteria, fungi, and parasites, and also some pharmacological stimuli such as phorbol 12-myristate 13-acetate (PMA) are efficient inducers of NETs. Antigen-antibody complexes are also capable of inducing NET formation. However the particular Fcγ receptor involved in triggering this function is a matter of controversy. In order to provide some insight into what Fcγ receptor is responsible for NET formation, each of the two human Fcγ receptors was stimulated individually by specific monoclonal antibodies and NET formation was evaluated. FcγRIIa cross-linking did not promote NET formation. Cross-linking other receptors such as integrins also did not promote NET formation. In contrast FcγRIIIb cross-linking induced NET formation similarly to PMA stimulation. NET formation was dependent on NADPH-oxidase, PKC, and ERK activation. These data show that cross-linking FcγRIIIb is responsible for NET formation by the human neutrophil.
Collapse
Affiliation(s)
- Omar Rafael Alemán
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510 México, DF, Mexico
| | - Nancy Mora
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510 México, DF, Mexico
| | - Ricarda Cortes-Vieyra
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510 México, DF, Mexico
| | - Eileen Uribe-Querol
- División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, 04510 México, DF, Mexico
| | - Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510 México, DF, Mexico
| |
Collapse
|
7
|
Behnen M, Leschczyk C, Möller S, Batel T, Klinger M, Solbach W, Laskay T. Immobilized immune complexes induce neutrophil extracellular trap release by human neutrophil granulocytes via FcγRIIIB and Mac-1. THE JOURNAL OF IMMUNOLOGY 2014; 193:1954-65. [PMID: 25024378 DOI: 10.4049/jimmunol.1400478] [Citation(s) in RCA: 182] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Canonical neutrophil antimicrobial effector mechanisms, such as degranulation, production of reactive oxygen species, and release of neutrophil extracellular traps (NETs), can result in severe pathology. Activation of neutrophils through immune complexes (ICs) plays a central role in the pathogenesis of many autoimmune inflammatory diseases. In this study, we report that immobilized ICs (iICs), which are hallmarks of several autoimmune diseases, induce the release of NETs from primary human neutrophils. The iIC-induced NET formation was found to require production of reactive oxygen species by NADPH oxidase and myeloperoxidase and to be mediated by FcγRIIIb. Blocking of the β2 integrin macrophage-1 Ag but not lymphocyte function-associated Ag-1 abolished iIC-induced NET formation. This suggests that FcγRIIIb signals in association with macrophage-1 Ag. As intracellular signaling pathways involved in iIC-induced NET formation we identified the tyrosine kinase Src/Syk pathway, which downstream regulates the PI3K/Akt, p38 MAPK, and ERK1/2 pathways. To our knowledge, the present study shows for the first time that iICs induce NET formation. Thus, we conclude that NETs contribute to pathology in autoimmune inflammatory disorders associated with surface-bound ICs.
Collapse
Affiliation(s)
- Martina Behnen
- Institute for Medical Microbiology and Hygiene, University of Lübeck, D-23538 Lübeck, Germany; and
| | - Christoph Leschczyk
- Institute for Medical Microbiology and Hygiene, University of Lübeck, D-23538 Lübeck, Germany; and
| | - Sonja Möller
- Institute for Medical Microbiology and Hygiene, University of Lübeck, D-23538 Lübeck, Germany; and
| | - Tobit Batel
- Institute for Medical Microbiology and Hygiene, University of Lübeck, D-23538 Lübeck, Germany; and
| | - Matthias Klinger
- Institute of Anatomy, University of Lübeck, D-23538 Lübeck, Germany
| | - Werner Solbach
- Institute for Medical Microbiology and Hygiene, University of Lübeck, D-23538 Lübeck, Germany; and
| | - Tamás Laskay
- Institute for Medical Microbiology and Hygiene, University of Lübeck, D-23538 Lübeck, Germany; and
| |
Collapse
|
8
|
Migita K, Izumi Y, Jiuchi Y, Kozuru H, Kawahara C, Nakamura M, Nakamura T, Agematsu K, Masumoto J, Yasunami M, Kawakami A, Eguchi K. Serum amyloid A induces NLRP-3-mediated IL-1β secretion in neutrophils. PLoS One 2014; 9:e96703. [PMID: 24846290 PMCID: PMC4028190 DOI: 10.1371/journal.pone.0096703] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 04/10/2014] [Indexed: 11/18/2022] Open
Abstract
Background/Aims Serum amyloid A (SAA) is an acute phase reactant with significant immunological activities, including effects on cytokine synthesis and neutrophil chemotaxis. Neutrophils can also release cytokines with proinflammatory properties. IL-1β is a key proinflammatory cytokine, the secretion of which is controlled by inflammasome. We investigated the proinflammatory effects of SAA invitro in relation to the NLRP3 inflammasome in neutrophils. Methodology/Principal Findings Human neutrophils isolated form healthy subjects were stimulated with serum amyloid A (SAA). The cellular supernatants were analyzed by western blot using anti-IL-1β or anti-caspase-1 antibodies. IL-1β or Nod-like receptor family, pyrin domain containing 3 (NLRP3) mRNA expressions were analyzed by real-time PCR or reverse transcription-PCR (RT-PCR) method. SAA stimulation induced pro-IL-1β mRNA expression in neutrophils. Furthermore, SAA engaged the caspase-1-activating inflammasome, resulting in the production of active IL-1β. SAA-induced pro-IL-1β expression was marginally suppressed by the Syk specific inhibitor, R406, and SAA-induced pro-IL-1β processing in neutrophils was prevented by R406. Furthermore, SAA-induced NLRP3 mRNA expression was completely blocked by R406. Analysis of intracellular signaling revealed that SAA stimulation activated the tyrosine kinase Syk and mitogen-activated protein kinase (MAPK). Conclusions/Significance These results demonstrate that the innate neutrophil immune response against SAA involves a two-step activation process: an initial signal promoting expression of pro-IL-1β and a second signal involving Syk-dependent activation of the NLRP3 inflammasome and caspase-1, allowing processing of pro-IL-1β and secretion of mature IL-1β.
Collapse
Affiliation(s)
- Kiyoshi Migita
- Department of Rheumatology and Clinical Research Center, Nagasaki Medical Center, Omura, Nagasaki, Japan
- * E-mail:
| | - Yasumori Izumi
- Department of Rheumatology and Clinical Research Center, Nagasaki Medical Center, Omura, Nagasaki, Japan
| | - Yuka Jiuchi
- Department of Rheumatology and Clinical Research Center, Nagasaki Medical Center, Omura, Nagasaki, Japan
| | - Hideko Kozuru
- Department of Rheumatology and Clinical Research Center, Nagasaki Medical Center, Omura, Nagasaki, Japan
| | - Chieko Kawahara
- Department of Rheumatology and Clinical Research Center, Nagasaki Medical Center, Omura, Nagasaki, Japan
| | - Minoru Nakamura
- Department of Rheumatology and Clinical Research Center, Nagasaki Medical Center, Omura, Nagasaki, Japan
| | - Tadashi Nakamura
- Department of Rheumatology, Shinto Kumamoto Hospital, Kumamoto, Japan
| | - Kazunaga Agematsu
- Department of Infection and Host Defense, Graduate School of Medicine, Shinshu University, Matsumoto, Nagano, Japan
| | - Junya Masumoto
- Department of Pathology, Ehime University Graduate School of Medicine and Proteo-Science Center, Toon, Ehime, Japan
| | - Michio Yasunami
- Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki University, Nagasaki, Japan
| | - Atsushi Kawakami
- Department of Rheumatology, Nagasaki University Hospital, Nagasaki, Japan
| | - Katsumi Eguchi
- Department of Rheumatology, Sasebo City General Hospital, Sasebo, Nagasaki, Japan
| |
Collapse
|
9
|
Crane JK, Mongiardo KM. Pro-inflammatory effects of uric acid in the gastrointestinal tract. Immunol Invest 2013; 43:255-66. [PMID: 24377830 DOI: 10.3109/08820139.2013.864667] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Uric acid can be generated in the gastrointestinal (GI) tract from the breakdown of nucleotides ingested in the diet or from purines released from host cells as a result of pathogen-induced cell damage. Xanthine oxidase (XO) is the enzyme that converts hypoxanthine or xanthine into uric acid, a reaction that also generates hydrogen peroxide. It has been assumed that the product of XO responsible for the pro-inflammatory effects of this enzyme is hydrogen peroxide. Recent literature on uric acid, however, has indicated that uric acid itself may have biological effects. We tested whether uric acid itself has detectable pro-inflammatory effects using an in vivo model using ligated rabbit intestinal segments ("loops") as well as in vitro assays using cultured cells. Addition of exogenous uric acid increased the influx of heterophils into rabbit intestinal loops, as measured by myeloperoxidase activity. In addition, white blood cells adhered avidly to uric acid crystals, forming large aggregates of cells. Uric acid acts as a leukocyte chemoattractant in the GI tract. The role of uric acid in enteric infections and in non-infectious disorders of the GI tract deserves more attention.
Collapse
Affiliation(s)
- John K Crane
- Deparment of Medicine, Division of Infectious Diseases, University at Buffalo , Buffalo, New York , USA
| | | |
Collapse
|
10
|
Crane JK. Role of host xanthine oxidase in infection due to enteropathogenic and Shiga-toxigenic Escherichia coli. Gut Microbes 2013; 4:388-91. [PMID: 23811846 PMCID: PMC3839983 DOI: 10.4161/gmic.25584] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Xanthine oxidase (XO) has been recognized as an important host defense enzyme for decades. In our recent study in Infection and Immunity, we found that enteropathogenic and Shiga-toxigenic E. coli (EPEC and STEC) were far more resistant to killing by the XO pathway than laboratory E. coli strains used in the past. Although XO plus hypoxanthine substrate rarely generated enough H 2O 2 to kill EPEC and STEC, the pathogens were able to sense the H2O2 and react to it with an increase in expression of virulence factors, most notably Shiga toxin (Stx). H 2O 2 produced by XO also triggered a chloride secretory response in T84 cell monolayers studied in the Ussing chamber. Adding exogenous XO plus its substrate in vivo did not decrease the number of STEC bacteria recovered from ligated intestinal loops, but instead appeared to worsen the infection and increased the amount of Stx2 toxin produced. XO plus hypoxanthine also increases the ability of Stx2 to translocate across intestinal monolayers. With regard to EPEC and STEC, the role of XO appears more complex and subtle than what has been reported in the past, since XO also plays a role in host-pathogen signaling, in regulating virulence in pathogens, in Stx production and in toxin translocation. Uric acid produced by XO may also be in itself an immune modulator in the intestinal tract.
Collapse
|
11
|
Wang X, Wanyan P, Wang JM, Tian JH, Hu L, Shen XP, Yang KH. A Randomized, Controlled Trial to Assess the Efficacy of Arthroscopic Debridement in Combination with Oral Medication Versus Oral Medication in Patients with Gouty Knee Arthritis. Indian J Surg 2013; 77:628-34. [PMID: 26730077 DOI: 10.1007/s12262-013-0949-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 07/05/2013] [Indexed: 11/29/2022] Open
Abstract
Gouty knee arthritis refers to a form of inflammatory diseases caused by deposits of needle-like crystals of uric acid in knee joint. The aim of this study was to assess the efficacy and safety of arthroscopic debridement in combination with oral medication versus oral medication alone for the treatment of gouty knee arthritis. A total of 60 patients with gouty knee arthritis were randomized to receive either arthroscopic surgery in combination with oral medication or oral medication alone. Efficacy was assessed with the angle of motion, functions, and visual analog scale (VAS). These indices were measured prior to treatment and at 2, 4, 12, 24, and 48 weeks posttreatment. Surgery- and medication-related complications were observed. Significant differences in flexion and extension of the knee joint, lymphoma scores, and VAS were detected between the two groups at 2, 4, and 12 weeks posttreatment (P < 0.05) but not at weeks 24 and 48 posttreatment (P > 0.05) . Significant differences in these indices were detected at different time points in each group (P < 0.05), except between weeks 24 and 48 (P > 0.05). Arthroscopic surgery in combination with oral medication is superior to single oral medication in the flexion and extension of the knee joint, lymphoma scores, and pain relief (VAS) before 24 weeks, although no statistical differences were detected in the efficacy after 24 weeks, and in medication-related safety between the two groups. Although arthroscopic debridement cannot replace systemic uric acid-lowering treatments such as medication and dietary control, it is still an effective approach.
Collapse
Affiliation(s)
- Xin Wang
- First Clinical Medical College of Lanzhou University, the First Hospital of Lanzhou University, Dong Gang West Road, No. 199, Lanzhou, 730000 Gansu China
| | - Pingping Wanyan
- The Second Hospital of Lanzhou University, Cui Ying Men, Lanzhou, 730030 Gansu China
| | - Jian Min Wang
- The First Hospital of Lanzhou University, Dong Gang West Road, No. 199, Lanzhou, 730000 Gansu China
| | - Jin Hui Tian
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Dong Gang West Road, No. 199, Lanzhou, 730000 Gansu China
| | - Long Hu
- Orthopedics Department, The First Hospital of Lanzhou University, Lanzhou, Gansu China
| | - Xi Ping Shen
- Department of Epidemiology and Health Statistics, School of Public Health, Lanzhou University, Lanzhou, Gansu China
| | - Ke Hu Yang
- Evidence-Based Medicine Center, The First Clinical Medical College of Lanzhou University, Dong Gang West Road, No. 199, Lanzhou, 730000 Gansu China
| |
Collapse
|
12
|
Mechanistic aspects of inflammation and clinical management of inflammation in acute gouty arthritis. J Clin Rheumatol 2013; 19:19-29. [PMID: 23319019 DOI: 10.1097/rhu.0b013e31827d8790] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
It has been recently demonstrated that interleukin 1β (IL-1β) plays a central role in monosodium urate crystal-induced inflammation and that the NALP3 inflammasome plays a major role in IL-1β production. These discoveries have offered new insights into the pathogenesis of acute gouty arthritis. In this review, we discuss the molecular mechanisms by which monosodium urate crystals induce acute inflammation and examine the mechanisms of action (MOAs) of traditional anti-inflammatory drugs (e.g., nonsteroidal anti-inflammatory drugs, colchicine, and glucocorticoids) and biologic agents (e.g., the IL-1β antagonists anakinra, rilonacept, and canakinumab) to understand how their MOAs contribute to their safety profiles. Traditional anti-inflammatory agents may act on the IL-1β pathway at some level; however, their MOAs are broad-ranging, unspecific, and biologically complex. This lack of specificity may explain the range of systemic adverse effects associated with them. The therapeutic margins of nonsteroidal anti-inflammatory drugs, colchicine, and glucocorticoids are particularly low in elderly patients and in patients with cardiovascular, metabolic, or renal comorbidities that are frequently associated with gouty arthritis. In contrast, the IL-1β antagonists act on very specific targets of inflammation, which may decrease the potential for systemic adverse effects, although infrequent but serious adverse events (including infection and administration reactions) have been reported. Because these IL-1β antagonists target an early event immediately downstream from NALP3 inflammasome activation, they may provide effective alternatives to traditional agents with minimal systemic adverse effects. Results of ongoing trials of IL-1β antagonists will likely provide clarification of their potential role in the management of acute gouty arthritis.
Collapse
|
13
|
Neutrophils, IL-1β, and gout: is there a link? Semin Immunopathol 2013; 35:501-12. [DOI: 10.1007/s00281-013-0361-0] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Accepted: 01/09/2013] [Indexed: 01/09/2023]
|
14
|
Molecular mechanisms of N-formyl-methionyl-leucyl-phenylalanine-induced superoxide generation and degranulation in mouse neutrophils: phospholipase D is dispensable. Mol Cell Biol 2012; 33:136-45. [PMID: 23109426 DOI: 10.1128/mcb.00869-12] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Phospholipase D (PLD), which produces the lipid messenger phosphatidic acid (PA), has been implicated in superoxide generation and degranulation in neutrophils. The basis for this conclusion is the observation that primary alcohols, which interfere with PLD-catalyzed PA production, inhibit these neutrophil functions. However, off-target effects of primary alcohols cannot be totally excluded. Here, we generated PLD(-/-) mice in order to reevaluate the involvement of PLD in and investigate the molecular mechanisms of these neutrophil functions. Surprisingly, N-formyl-methionyl-leucyl-phenylalanine (fMLP) induced these functions in PLD(-/-) neutrophils, and these functions were suppressed by ethanol. These results indicate that PLD is dispensable for these neutrophil functions and that ethanol nonspecifically inhibits them, warning against the use of primary alcohols as specific inhibitors of PLD-mediated PA formation. The calcium ionophore ionomycin and the membrane-permeative compound 1-oleoyl-2-acetyl-sn-glycerol (OADG) synergistically induced superoxide generation. On the other hand, ionomycin alone induced degranulation, which was further augmented by OADG. These results demonstrate that conventional protein kinase C (cPKC) is crucial for superoxide generation, and a Ca(2+)-dependent signaling pathway(s) and cPKC are involved in degranulation in mouse neutrophils.
Collapse
|
15
|
Abstract
Uric acid is a waste product of purine catabolism. This molecule comes to clinical attention when it nucleates to form crystals of monosodium urate (MSU) in joints or other tissues, and thereby causes the inflammatory disease of gout. Patients with gout frequently suffer from a number of comorbid conditions including hypertension, diabetes mellitus and cardiovascular disease. Why MSU crystals trigger inflammation and are associated with comorbidities of gout has been unclear, but recent studies provide new insights into these issues. Rather than simply being a waste product, uric acid could serve a pathophysiological role as a local alarm signal that alerts the immune system to cell injury and helps to trigger both innate and adaptive immune responses. The inflammatory component of these immune responses is caused when urate crystals trigger both inflammasome-dependent and independent pathways to generate the proinflammatory cytokine IL-1. The resulting bioactive IL-1 stimulates the inflammation of gout and might contribute to the development of other comorbidities. Surprisingly, the same mechanisms underlie the inflammatory response to a number of irritant particles, many of which also cause disease. These new insights help to explain the pathogenesis of gout and point to potential new therapeutic targets for this and other sterile inflammatory diseases.
Collapse
|
16
|
|
17
|
Mitroulis I, Kambas K, Chrysanthopoulou A, Skendros P, Apostolidou E, Kourtzelis I, Drosos GI, Boumpas DT, Ritis K. Neutrophil extracellular trap formation is associated with IL-1β and autophagy-related signaling in gout. PLoS One 2011; 6:e29318. [PMID: 22195044 PMCID: PMC3241704 DOI: 10.1371/journal.pone.0029318] [Citation(s) in RCA: 296] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 11/24/2011] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Gout is a prevalent inflammatory arthritis affecting 1-2% of adults characterized by activation of innate immune cells by monosodium urate (MSU) crystals resulting in the secretion of interleukin-1β (IL-1β). Since neutrophils play a major role in gout we sought to determine whether their activation may involve the formation of proinflammatory neutrophil extracellular traps (NETs) in relation to autophagy and IL-1β. METHODOLOGY/PRINCIPAL FINDINGS Synovial fluid neutrophils from six patients with gout crisis and peripheral blood neutrophils from six patients with acute gout and six control subjects were isolated. MSU crystals, as well as synovial fluid or serum obtained from patients with acute gout, were used for the treatment of control neutrophils. NET formation was assessed using immunofluorescence microscopy. MSU crystals or synovial fluid or serum from patients induced NET formation in control neutrophils. Importantly, NET production was observed in neutrophils isolated from synovial fluid or peripheral blood from patients with acute gout. NETs contained the alarmin high mobility group box 1 (HMGB1) supporting their pro-inflammatory potential. Inhibition of phosphatidylinositol 3-kinase signaling or phagolysosomal fusion prevented NET formation, implicating autophagy in this process. NET formation was driven at least in part by IL-1β as demonstrated by experiments involving IL-1β and its inhibitor anakinra. CONCLUSIONS/SIGNIFICANCE These findings document for the first time that activation of neutrophils in gout is associated with the formation of proinflammatory NETs and links this process to both autophagy and IL-1β. Modulation of the autophagic machinery may represent an additional therapeutic study in crystalline arthritides.
Collapse
Affiliation(s)
- Ioannis Mitroulis
- First Department of Internal Medicine, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Konstantinos Kambas
- First Department of Internal Medicine, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Akrivi Chrysanthopoulou
- First Department of Internal Medicine, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Panagiotis Skendros
- First Department of Internal Medicine, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Eirini Apostolidou
- First Department of Internal Medicine, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Ioannis Kourtzelis
- First Department of Internal Medicine, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Georgios I. Drosos
- Department of Orthopaedic Surgery, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Dimitrios T. Boumpas
- Department of Rheumatology, Clinical Immunology and Allergy, University Hospital, Medical School, University of Crete, Heraklion, Greece
| | - Konstantinos Ritis
- First Department of Internal Medicine, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
- * E-mail:
| |
Collapse
|
18
|
Uratsuji H, Tada Y, Kawashima T, Kamata M, Hau CS, Asano Y, Sugaya M, Kadono T, Asahina A, Sato S, Tamaki K. P2Y6 receptor signaling pathway mediates inflammatory responses induced by monosodium urate crystals. THE JOURNAL OF IMMUNOLOGY 2011; 188:436-44. [PMID: 22102722 DOI: 10.4049/jimmunol.1003746] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Gout occurs in individuals with hyperuricemia when monosodium urate (MSU) crystals precipitate in tissues and induce acute inflammation via phagocytic cells such as monocytes. MSU crystals have been demonstrated in skin diseases such as tophaceous gout or psoriasis; however, the importance of MSU crystals in the skin is totally unknown. In this study, we found that MSU crystals, through P2Y(6) receptors, stimulated normal human keratinocytes (NHK) to produce IL-1α, IL-8/CXCL8, and IL-6. P2Y(6) receptor expression increased in MSU-stimulated NHK. Both P2Y(6)-specific antagonist and P2Y(6) antisense oligonucleotides significantly inhibited the production of IL-1α, IL-8/CXCL8, and IL-6 by NHK. Similarly, the P2Y(6)-specific antagonist completely inhibited the MSU-induced production of IL-1β by THP-1 cells, a human monocytic cell line. Remarkably, the P2Y(6)-specific antagonist significantly reduced neutrophil influx in both mouse air pouch and peritonitis models. Thus, these results indicate that the P2Y(6) receptor signaling pathway may be a potential therapeutic target for MSU-associated inflammatory diseases, such as tophaceous gout.
Collapse
Affiliation(s)
- Hideya Uratsuji
- Department of Dermatology, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
DCIR-mediated enhancement of HIV-1 infection requires the ITIM-associated signal transduction pathway. Blood 2011; 117:6589-99. [DOI: 10.1182/blood-2011-01-331363] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Dendritic cell immunoreceptor (DCIR) is a C-type lectin receptor expressed at high levels on dendritic cells (DCs). This surface molecule acts as an attachment factor for HIV-1 on DCs and contributes to trans- and cis-infection pathways. Moreover, DICR is induced by HIV-1 in CD4+ T cells and promotes virus replication in this cell type. Nothing is known hitherto about the DCIR-dependent signaling, which is induced following HIV-1 ligation. First, specific pharmacologic inhibitors were tested on HIV-1 binding/entry and, second, specific antisense oligonucleotides targeted, more specifically kinases and phosphatases, were used. Our results show that SHP-1, SHP-2, Syk, and Src kinases (ie, Src, Fyn, and Hck) as well as PKC-α and MAP kinases (ie, Erk1/2 and p38) are all involved in the DCIR-mediated signal transduction pathway triggered by HIV-1. By mutagenesis and through the use of intracellular phosphorylated peptides, we show as well a pivotal role for the tyrosine and threonine residues of the DCIR immunoreceptor tyrosine-based inhibitory motif (ITIM). Our data suggest for the first time an involvement of ITIM domain in HIV-1–mediated signaling events and a relationship between phosphorylation events and DCIR function with respect to HIV-1 biology.
Collapse
|
20
|
Marois L, Vaillancourt M, Paré G, Gagné V, Fernandes MJG, Rollet-Labelle E, Naccache PH. CIN85 modulates the down-regulation of Fc gammaRIIa expression and function by c-Cbl in a PKC-dependent manner in human neutrophils. J Biol Chem 2011; 286:15073-84. [PMID: 21372129 PMCID: PMC3083175 DOI: 10.1074/jbc.m110.213660] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 02/21/2011] [Indexed: 01/04/2023] Open
Abstract
We previously described a non-classical mechanism that arrests FcγRIIa signaling in human neutrophils once engaged by immune complexes or opsonized pathogens. The engagement of FcγRIIa leads to its ubiquitination by the ubiquitin ligase c-Cbl and degradation by the proteasome. Herein, we further examined some of the events regulating this novel pathway. The adaptor protein CIN85 was described in other systems to be involved in the regulation of the c-Cbl-dependent pathway. We found that CIN85 is expressed in human neutrophils and that it translocates like c-Cbl from the cytosol to the plasma membrane following receptor cross-linking. CIN85 was also recruited to the same subset of high density detergent-resistant membrane fractions in which stimulated FcγRIIa partitioned with c-Cbl. The integrity of these microdomains is essential to the FcγRIIa degradation process because the cholesterol-depleting agent methyl-β-cyclodextrin inhibits this event. Silencing the expression of CIN85 by siRNA in dibutyryl cyclic AMP-differentiated PLB 985 cells prevented FcγRIIa degradation and increased IgG-mediated phagocytosis. Confocal microscopy revealed that the presence of CIN85 is essential to the proper sorting of FcγRIIa during endocytosis. We also provide direct evidence that CIN85 is a substrate of serine/threonine kinase PKCs. Classical PKCs positively regulate FcγRIIa ubiquitination and degradation because these events were inhibited by Gö6976, a classical PKC inhibitor. We conclude that the ubiquitination and degradation of stimulated FcγRIIa mediated by c-Cbl are positively regulated by the adaptor protein CIN85 in a PKC-dependent manner and that these events contribute to the termination of FcγRIIa signaling.
Collapse
Affiliation(s)
- Louis Marois
- From the Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du CHUQ, Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, Québec G1V 4G2, Canada
| | - Myriam Vaillancourt
- From the Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du CHUQ, Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, Québec G1V 4G2, Canada
| | - Guillaume Paré
- From the Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du CHUQ, Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, Québec G1V 4G2, Canada
| | - Valérie Gagné
- From the Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du CHUQ, Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, Québec G1V 4G2, Canada
| | - Maria J. G. Fernandes
- From the Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du CHUQ, Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, Québec G1V 4G2, Canada
| | - Emmanuelle Rollet-Labelle
- From the Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du CHUQ, Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, Québec G1V 4G2, Canada
| | - Paul H. Naccache
- From the Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du CHUQ, Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, Québec G1V 4G2, Canada
| |
Collapse
|
21
|
Bertram A, Ley K. Protein kinase C isoforms in neutrophil adhesion and activation. Arch Immunol Ther Exp (Warsz) 2011; 59:79-87. [PMID: 21298489 DOI: 10.1007/s00005-011-0112-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Accepted: 10/08/2010] [Indexed: 01/13/2023]
Abstract
Neutrophils are the first line of defense against bacterial and mycotic pathogens. In order to reach the pathogens, neutrophils need to transmigrate through the vascular endothelium and migrate to the site of infection. Defense strategies against pathogens include phagocytosis, production and release of oxygen radicals through the oxidative burst, and degranulation of antimicrobial and inflammatory molecules. Protein kinase C (PKC)-δ is required for full assembly of NADPH oxidase and activation of the respiratory burst. Neutrophils also express PKC-α and -β, which may be involved in adhesion, degranulation and phagocytosis, but the evidence is not conclusive yet. This review focuses on the potential impact of protein kinase C isoforms on neutrophil adhesion and activation.
Collapse
Affiliation(s)
- Anna Bertram
- Department of Nephrology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover, Germany
| | | |
Collapse
|
22
|
Cohen G, Raupachova J, Ilic D, Werzowa J, Hörl WH. Effect of leptin on polymorphonuclear leucocyte functions in healthy subjects and haemodialysis patients. Nephrol Dial Transplant 2011; 26:2271-81. [PMID: 21216885 DOI: 10.1093/ndt/gfq731] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Dysfunction of polymorphonuclear leucocytes (PMNLs) in end-stage renal disease (ESRD) patients contributes to a diminished immune defence. The serum levels of leptin are elevated in patients with ESRD. We analysed in vitro effects of leptin on PMNLs from healthy subjects (HS; n = 12) and haemodialysis (HD) patients (n = 15) before and after HD. METHODS PMNL oxidative burst and phagocytosis were tested by flow cytometry in whole blood. Chemotaxis of isolated PMNLs was assessed by the under-agarose method. To assess the involvement of leptin in PMNL signalling pathways, signal transduction inhibitors were used and the activity of intracellular kinases was investigated by western blotting, in vitro kinase assays and the Luminex technology. RESULTS Increasing the leptin level in the blood of HS leads to a reduced activation of the oxidative burst by Escherichia coli and phorbol 12-myristate 13-acetate. Activation of the oxidative burst is reduced in the blood of HD patients and the addition of leptin does not lead to further PMNL inhibition. Leptin at a concentration measured in HD patients significantly reduces the chemotaxis of PMNLs from HS but had no effect on PMNLs from ESRD patients before and also after HD treatment with high-flux dialysers. The phosphoinositide 3-kinase/Akt pathway is involved in the inhibitory effects of leptin. CONCLUSIONS In the presence of leptin, PMNLs from HS and HD patients respond differently to stimuli. The lack of response to leptin in PMNLs from HD patients cannot be influenced by HD.
Collapse
Affiliation(s)
- Gerald Cohen
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Cincinnati, Cincinnati, OH, USA.
| | | | | | | | | |
Collapse
|
23
|
Litherland GJ, Elias MS, Hui W, Macdonald CD, Catterall JB, Barter MJ, Farren MJ, Jefferson M, Rowan AD. Protein kinase C isoforms zeta and iota mediate collagenase expression and cartilage destruction via STAT3- and ERK-dependent c-fos induction. J Biol Chem 2010; 285:22414-25. [PMID: 20463008 PMCID: PMC2903406 DOI: 10.1074/jbc.m110.120121] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Revised: 04/30/2010] [Indexed: 12/27/2022] Open
Abstract
The protein kinase C (PKC) signaling pathway is a major regulator of cellular functions and is implicated in pathologies involving extracellular matrix remodeling. Inflammatory joint disease is characterized by excessive extracellular matrix catabolism, and here we assess the role of PKC in the induction of the collagenases, matrix metalloproteinase (MMP)-1 and MMP-13, in human chondrocytes by the potent cytokine stimulus interleukin-1 (IL-1) in combination with oncostatin M (OSM). IL-1 + OSM-stimulated collagenolysis and gelatinase activity were ameliorated by pharmacological PKC inhibition in bovine cartilage, as was collagenase gene induction in human chondrocytes. Small interfering RNA-mediated silencing of PKC gene expression showed that both novel (nPKC delta, nPKC eta) and atypical (aPKC zeta, aPKC iota) isoforms were involved in collagenase induction by IL-1. However, MMP1 and MMP13 induction by IL-1 + OSM was inhibited only by aPKC silencing, suggesting that only atypical isoforms play a significant role in complex inflammatory milieus. Silencing of either aPKC led to diminished IL-1 + OSM-dependent extracellular signal-regulated kinase (ERK) and signal transducer and activator of transcription (STAT) 3 phosphorylation, and c-fos expression. STAT3 gene silencing or ERK pathway inhibition also resulted in loss of IL-1 + OSM-stimulated c-fos and collagenase expression. Silencing of c-fos and c-jun expression was sufficient to abrogate IL-1 + OSM-stimulated collagenase gene induction, and overexpression of both c-fos and c-jun was sufficient to drive transcription from the MMP1 promoter in the absence of a stimulus. Our data identify atypical PKC isozymes as STAT and ERK activators that mediate c-fos and collagenase expression during IL-1 + OSM synergy in human chondrocytes. aPKCs may constitute potential therapeutic targets for inflammatory joint diseases involving increased collagenase expression.
Collapse
Affiliation(s)
- Gary J. Litherland
- From the Cell Signalling, Injury and Repair Group, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle-upon-Tyne NE2 4HH, United Kingdom
| | - Martina S. Elias
- From the Cell Signalling, Injury and Repair Group, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle-upon-Tyne NE2 4HH, United Kingdom
| | - Wang Hui
- From the Cell Signalling, Injury and Repair Group, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle-upon-Tyne NE2 4HH, United Kingdom
| | - Christopher D. Macdonald
- From the Cell Signalling, Injury and Repair Group, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle-upon-Tyne NE2 4HH, United Kingdom
| | - Jonathon B. Catterall
- From the Cell Signalling, Injury and Repair Group, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle-upon-Tyne NE2 4HH, United Kingdom
| | - Matt J. Barter
- From the Cell Signalling, Injury and Repair Group, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle-upon-Tyne NE2 4HH, United Kingdom
| | - Matthew J. Farren
- From the Cell Signalling, Injury and Repair Group, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle-upon-Tyne NE2 4HH, United Kingdom
| | - Matthew Jefferson
- From the Cell Signalling, Injury and Repair Group, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle-upon-Tyne NE2 4HH, United Kingdom
| | - Andrew D. Rowan
- From the Cell Signalling, Injury and Repair Group, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle-upon-Tyne NE2 4HH, United Kingdom
| |
Collapse
|
24
|
Affiliation(s)
- Oana Popa‐Nita
- Centre de Recherche en Rhumatologie et Immunologie du Centre de Recherche du CHUL‐CHUQ, Department of Microbiology and Immunology, Faculty of Medicine, Laval University Québec Canada
| | - Paul H Naccache
- Centre de Recherche en Rhumatologie et Immunologie du Centre de Recherche du CHUL‐CHUQ, Department of Microbiology and Immunology, Faculty of Medicine, Laval University Québec Canada
| |
Collapse
|