1
|
Demir MF, Lin YH, Costa Cruz PH, Tajima M, Honjo T, Müller E. Blocking S100A9-signaling is detrimental to the initiation of anti-tumor immunity. Front Immunol 2024; 15:1479502. [PMID: 39497822 PMCID: PMC11532050 DOI: 10.3389/fimmu.2024.1479502] [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: 08/12/2024] [Accepted: 10/03/2024] [Indexed: 11/07/2024] Open
Abstract
S100A9, a multifunctional protein mainly expressed by neutrophils and monocytes, poses an immunological paradox. In virus infections or sterile inflammation, it functions as an alarmin attracting innate immune cells, as well as mediating proinflammatory effects through TLR4 signaling. However, in cancer, S100A9 levels have been shown to associate with poor prognosis and lack of response to immunotherapy. Its expression by myeloid cells has been related to an immune suppressive phenotype, the so-called myeloid derived suppressor cells (MDSCs). Targeting S100A9 in cancer has therefore been proposed as a potential way to relieve myeloid-mediated immune suppression. Surprisingly, we found that blocking the extracellular TLR4 signaling from S100A9 using the inhibitor Paquinimod, resulted in increased tumor growth and a detrimental effect on anti-PD-L1 efficacy in the CT26 tumor model. This effect was caused by a reduction in the tumor immune infiltration to about half of untreated controls, and the reduction was made up of a 5-fold decrease in Ly6Chigh monocytic cells. The suppressive Ly6G+ myeloid cells compartment was not reduced by Paquinimod treatment, suggesting alternative mechanisms by which S100A9 contributes to myeloid-mediated suppression. Intratumoral injection of recombinant S100A9 early after mice inoculation with CT26 cells had an anti-tumor effect. These findings indicate an important yet understudied role of S100A9 as an alarmin and immune stimulatory signal in cancer settings, and highlight the potential to exploit such signals to promote beneficial anti-tumor responses.
Collapse
Affiliation(s)
- Melike Fusun Demir
- Department of Immunology and Genomic Medicine, Kyoto University, Kyoto, Japan
- Division of Integrated High-Order Regulatory Systems, Center for Cancer Immunotherapy and Immunobiology, Kyoto, Japan
| | - Yu-Hsien Lin
- Division of Integrated High-Order Regulatory Systems, Center for Cancer Immunotherapy and Immunobiology, Kyoto, Japan
| | - Pedro Henrique Costa Cruz
- Division of Integrated High-Order Regulatory Systems, Center for Cancer Immunotherapy and Immunobiology, Kyoto, Japan
| | - Masaki Tajima
- Division of Integrated High-Order Regulatory Systems, Center for Cancer Immunotherapy and Immunobiology, Kyoto, Japan
| | - Tasuku Honjo
- Department of Immunology and Genomic Medicine, Kyoto University, Kyoto, Japan
| | - Elisabeth Müller
- Department of Immunology and Genomic Medicine, Kyoto University, Kyoto, Japan
- Division of Integrated High-Order Regulatory Systems, Center for Cancer Immunotherapy and Immunobiology, Kyoto, Japan
- Tumor Immunology Group, Institute of Pathology, Oslo University Hospital, Oslo, Norway
- Therapy Prediction In Lung Cancer, Department of Cancer Genetics, Institute of Cancer Research, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
2
|
Özbay Kurt FG, Cicortas BA, Balzasch BM, De la Torre C, Ast V, Tavukcuoglu E, Ak C, Wohlfeil SA, Cerwenka A, Utikal J, Umansky V. S100A9 and HMGB1 orchestrate MDSC-mediated immunosuppression in melanoma through TLR4 signaling. J Immunother Cancer 2024; 12:e009552. [PMID: 39266214 PMCID: PMC11409250 DOI: 10.1136/jitc-2024-009552] [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] [Accepted: 08/01/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Immunotherapies for malignant melanoma are challenged by the resistance developed in a significant proportion of patients. Myeloid-derived suppressor cells (MDSC), with their ability to inhibit antitumor T-cell responses, are a major contributor to immunosuppression and resistance to immune checkpoint therapies in melanoma. Damage-associated molecular patterns S100A8, S100A9, and HMGB1, acting as toll like receptor 4 (TLR4) and receptor for advanced glycation endproducts (RAGE) ligands, are highly expressed in the tumor microenvironment and drive MDSC activation. However, the role of TLR4 and RAGE signaling in the acquisition of MDSC immunosuppressive properties remains to be better defined. Our study investigates how the signaling via TLR4 and RAGE as well as their ligands S100A9 and HMGB1, shape MDSC-mediated immunosuppression in melanoma. METHODS MDSC were isolated from the peripheral blood of patients with advanced melanoma or generated in vitro from healthy donor-derived monocytes. Monocytes were treated with S100A9 or HMGB1 for 72 hours. The immunosuppressive capacity of treated monocytes was assessed in the inhibition of T-cell proliferation assay in the presence or absence of TLR4 and RAGE inhibitors. Plasma levels of S100A8/9 and HMGB1 were quantified by ELISA. Single-cell RNA sequencing (scRNA-seq) was performed on monocytes from patients with melanoma and healthy donors. RESULTS We showed that exposure to S100A9 and HMGB1 converted healthy donor-derived monocytes into MDSC through TLR4 signaling. Our scRNA-seq data revealed in patient monocytes enriched inflammatory genes, including S100 and those involved in NF-κB and TLR4 signaling, and a reduced major histocompatibility complex II gene expression. Furthermore, elevated plasma S100A8/9 levels correlated with shorter progression-free survival in patients with melanoma. CONCLUSIONS These findings highlight the critical role of TLR4 and, to a lesser extent, RAGE signaling in the conversion of monocytes into MDSC-like cells, underscore the potential of targeting S100A9 to prevent this conversion, and highlight the prognostic value of S100A8/9 as a plasma biomarker in melanoma.
Collapse
Affiliation(s)
- Feyza Gül Özbay Kurt
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany
- Department of Dermatology Venereology and Allergology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Beatrice-Ana Cicortas
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany
- Department of Dermatology Venereology and Allergology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Bianca M Balzasch
- Department of Immunobiochemistry, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Carolina De la Torre
- NGS Core Facility, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Volker Ast
- NGS Core Facility, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ece Tavukcuoglu
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany
- Department of Dermatology Venereology and Allergology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
| | - Cagla Ak
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany
- Department of Dermatology Venereology and Allergology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sebastian A Wohlfeil
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany
- Department of Dermatology Venereology and Allergology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
| | - Adelheid Cerwenka
- Department of Immunobiochemistry, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany
- Department of Dermatology Venereology and Allergology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany
- Department of Dermatology Venereology and Allergology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
3
|
Chisholm LO, Jeon CK, Prell JS, Harms MJ. Changing expression system alters oligomerization and proinflammatory activity of recombinant human S100A9. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.14.608001. [PMID: 39185185 PMCID: PMC11343194 DOI: 10.1101/2024.08.14.608001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
S100A9 is a Damage Associated Molecular Pattern (DAMP) that activates the innate immune system via Toll-like receptor 4 (TLR4). Despite many years of study, the mechanism of activation remains unknown. To date, much of the biochemical characterization of S100A9 has been performed using recombinant S100A9 expressed in E. coli (S100A9ec). TLR4 is the canonical receptor for LPS, a molecule found in the outer membrane of E. coli, raising the possibility of artifacts due to LPS contamination. Here we report characterization of LPS-free recombinant S100A9 expressed in insect cells (S100A9in). We show that S100A9in does not activate TLR4. This difference does not appear to be due to LPS contamination, protein misfolding, purification artifacts, or differences in phosphorylation. We show instead that S100A9in adopts an altered oligomeric state compared to S100A9ec. Disrupting oligomer formation with the E. coli disaggregase SlyD restores activity to S100A9in. Our results also indicate that the oligomeric state of S100A9 is a major factor in its ability to activate TLR4 and that this can be altered in unexpected ways by the recombinant expression system used to produce the protein.
Collapse
Affiliation(s)
- Lauren O. Chisholm
- Department of Chemistry & Biochemistry, University of Oregon, Eugene OR 97403
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403
| | - Chae Kyung Jeon
- Department of Chemistry & Biochemistry, University of Oregon, Eugene OR 97403
- Materials Science Institute, University of Oregon, Eugene, OR 97403
| | - James S. Prell
- Department of Chemistry & Biochemistry, University of Oregon, Eugene OR 97403
- Materials Science Institute, University of Oregon, Eugene, OR 97403
| | - Michael J. Harms
- Department of Chemistry & Biochemistry, University of Oregon, Eugene OR 97403
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403
| |
Collapse
|
4
|
Lodge-Tulloch NA, Paré JF, Couture C, Bernier E, Cotechini T, Girard S, Graham CH. Maternal Innate Immune Reprogramming After Complicated Pregnancy. Am J Reprod Immunol 2024; 92:e13908. [PMID: 39119763 DOI: 10.1111/aji.13908] [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: 01/30/2024] [Revised: 05/19/2024] [Accepted: 07/05/2024] [Indexed: 08/10/2024] Open
Abstract
PROBLEM Preeclampsia (PE) and fetal growth restriction (FGR) are often associated with maternal inflammation and an increased risk of cardiovascular and metabolic disease in the affected mothers. The mechanism responsible for this increased risk of subsequent disease may involve reprogramming of innate immune cells, characterized by epigenetic modifications. METHOD OF STUDY Circulating monocytes from women with PE, FGR, or uncomplicated pregnancies (control) were isolated before labor. Cytokine release from monocytes following exposure to lipopolysaccharide (LPS) and the presence of lysine 4-trimethylated histone 3 (H3K4me3) within TNF promoter sequences were evaluated. Single-cell transcriptomic profiles of circulating monocytes from women with PE or uncomplicated pregnancies were assessed. RESULTS Monocytes from women with PE or FGR exhibited increased IL-10 secretion and decreased IL-1β and GM-CSF secretion in response to LPS. While TNFα secretion was not significantly different in cultures of control monocytes versus those from complicated pregnancies with or without LPS exposure, monocytes from complicated pregnancies had significantly decreased levels of H3K4me3 associated with TNF promoter sequences. Cluster quantification and pathway analysis of differentially expressed genes revealed an increased proportion of anti-inflammatory myeloid cells and a lower proportion of inflammatory non-classical monocytes among the circulating monocyte population in women with PE. CONCLUSIONS Monocytes from women with PE and FGR exhibit an immune tolerance phenotype before initiation of labor. Further investigation is required to determine whether this tolerogenic phenotype persists after the affected pregnancy and contributes to increased risk of subsequent disease.
Collapse
Affiliation(s)
| | - Jean-François Paré
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Camille Couture
- Department of Obstetrics and Gynecology, Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
- Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada
| | - Elsa Bernier
- Department of Obstetrics and Gynecology, Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
- Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada
| | - Tiziana Cotechini
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
- Department of Obstetrics and Gynecology, Université de Montréal, Montreal, Quebec, Canada
| | - Charles H Graham
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
5
|
Boucher J, Gilbert C, Bose S, Tessier PA. S100A9: The Unusual Suspect Connecting Viral Infection and Inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1523-1529. [PMID: 38709994 PMCID: PMC11076006 DOI: 10.4049/jimmunol.2300640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/20/2024] [Indexed: 05/08/2024]
Abstract
The study of S100A9 in viral infections has seen increased interest since the COVID-19 pandemic. S100A8/A9 levels were found to be correlated with the severity of COVID-19 disease, cytokine storm, and changes in myeloid cell subsets. These data led to the hypothesis that S100A8/A9 proteins might play an active role in COVID-19 pathogenesis. This review explores the structures and functions of S100A8/9 and the current knowledge on the involvement of S100A8/A9 and its constituents in viral infections. The potential roles of S100A9 in SARS-CoV-2 infections are also discussed.
Collapse
Affiliation(s)
- Julien Boucher
- Axe de recherche sur les maladies infectieuses et immunitaires, Centre de recherche du CHU de Québec-Université Laval, and Département de microbiologie-infectiologie et d’immunologie, Faculté de Médecine, Université Laval, Quebec City, Quebec, Canada
| | - Caroline Gilbert
- Axe de recherche sur les maladies infectieuses et immunitaires, Centre de recherche du CHU de Québec-Université Laval, and Département de microbiologie-infectiologie et d’immunologie, Faculté de Médecine, Université Laval, Quebec City, Quebec, Canada
| | - Santanu Bose
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Philippe A. Tessier
- Axe de recherche sur les maladies infectieuses et immunitaires, Centre de recherche du CHU de Québec-Université Laval, and Département de microbiologie-infectiologie et d’immunologie, Faculté de Médecine, Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
6
|
Paramasivam S, Perumal SS, Ekambaram SP. Computational Deciphering of the Role of S100A8 and S100A9 Proteins and Their Changes in the Structure Assembly Influences Their Interaction with TLR4, RAGE, and CD36. Protein J 2024; 43:243-258. [PMID: 38431537 DOI: 10.1007/s10930-024-10186-0] [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] [Accepted: 02/20/2024] [Indexed: 03/05/2024]
Abstract
S100A8 and S100A9 belong to the calcium-binding, damage associated molecular pattern (DAMP) proteins shown to aggravate the pathogenesis of rheumatoid arthritis (RA) through their interaction with the TLR4, RAGE and CD36 receptors. S100A8 and S100A9 proteins tend to exist in monomeric, homo and heterodimeric forms, which have been implicated in the pathogenesis of RA, via interacting with Pattern Recognition receptors (PRRs). The study aims to assess the influence of changes in the structure and biological assembly of S100A8 and S100A9 proteins as well as their interaction with significant receptors in RA through computational methods and surface plasmon resonance (SPR) analysis. Molecular docking analysis revealed that the S100A9 homodimer and S100A8/A9 heterodimer showed higher binding affinity towards the target receptors. Most S100 proteins showed good binding affinity towards TLR4 compared to other receptors. Based on the 50 ns MD simulations, TLR4, RAGE, and CD36 formed stable complexes with the monomeric and dimeric forms of S100A8 and S100A9 proteins. However, SPR analysis showed that the S100A8/A9 heterodimers formed stable complexes and exhibited high binding affinity towards the receptors. SPR data also indicated that TLR4 and its interactions with S100A8/A9 proteins may play a primary role in the pathogenesis of RA, with additional contributions from CD36 and RAGE interactions. Subsequent in vitro and in vivo investigations are warranted to corroborate the involvement of S100A8/A9 and the expression of TLR4, RAGE, and CD36 in the pathophysiology of RA.
Collapse
Affiliation(s)
- Sivasakthi Paramasivam
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology Campus, University College of Engineering, Anna University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Senthamil Selvan Perumal
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology Campus, University College of Engineering, Anna University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Sanmuga Priya Ekambaram
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology Campus, University College of Engineering, Anna University, Tiruchirappalli, Tamil Nadu, 620 024, India.
| |
Collapse
|
7
|
Hau JL, Kremser H, Knogl-Tritschler S, Stefanski V, Steuber J, Fritz G. Fast IMAC purification of non-tagged S100A8/A9 (calprotectin) from Homo sapiens and Sus scrofa. Protein Expr Purif 2023; 208-209:106275. [PMID: 37084837 DOI: 10.1016/j.pep.2023.106275] [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: 02/24/2023] [Revised: 04/16/2023] [Accepted: 04/17/2023] [Indexed: 04/23/2023]
Abstract
S100A8/A9 (calprotectin) is a damage-associated molecular pattern molecule (DAMP) that plays a key role in the immune response of mammalia. S100A8/A9 is therefore widely used as a biomarker in human and veterinary medicine, but diagnostic tools for the detection of S100A8/A9 are rarely optimised for the specific organism, since the corresponding S100A8/A9 is often not available. There is need for an easy, reliable protocol for the production of recombinant, highly pure S100A8/A9 from various mammalia. Here we describe the expression and purification of recombinant human and porcine S100A8/A9 by immobilized metal affinity chromatography (IMAC), which takes advantage of the intrinsic, high-affinity binding of native un-tagged S100A8/A9 to metal ions. Highly pure S100A8/A9 is obtained by a combination of IMAC, ion exchange and size exclusion chromatographic steps. Considering the high sequence homology and conservation of the metal ion coordinating residues of S100A8/A9 metal binding sites, the protocol is presumably applicable to S100A8/A9 of various mammalia.
Collapse
Affiliation(s)
- Jann-Louis Hau
- Institute of Biology, University of Hohenheim, Garbenstr. 30, 70599, Stuttgart, Germany
| | - Hannes Kremser
- Institute of Biology, University of Hohenheim, Garbenstr. 30, 70599, Stuttgart, Germany
| | | | - Volker Stefanski
- Institute of Animal Science, Behavioral Physiology of Livestock, University of Hohenheim, Garbenstr. 30, 70599, Stuttgart, Germany; HoLMiR-Hohenheim Center for Livestock Microbiome Research, University of Hohenheim, 70593, Stuttgart, Germany
| | - Julia Steuber
- Institute of Biology, University of Hohenheim, Garbenstr. 30, 70599, Stuttgart, Germany; HoLMiR-Hohenheim Center for Livestock Microbiome Research, University of Hohenheim, 70593, Stuttgart, Germany.
| | - Günter Fritz
- Institute of Biology, University of Hohenheim, Garbenstr. 30, 70599, Stuttgart, Germany.
| |
Collapse
|
8
|
Jukic A, Bakiri L, Wagner EF, Tilg H, Adolph TE. Calprotectin: from biomarker to biological function. Gut 2021; 70:1978-1988. [PMID: 34145045 PMCID: PMC8458070 DOI: 10.1136/gutjnl-2021-324855] [Citation(s) in RCA: 241] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/02/2021] [Indexed: 12/15/2022]
Abstract
The incidence of inflammatory bowel diseases (IBD) emerged with Westernisation of dietary habits worldwide. Crohn's disease and ulcerative colitis are chronic debilitating conditions that afflict individuals with substantial morbidity and challenge healthcare systems across the globe. Since identification and characterisation of calprotectin (CP) in the 1980s, faecal CP emerged as significantly validated, non-invasive biomarker that allows evaluation of gut inflammation. Faecal CP discriminates between inflammatory and non-inflammatory diseases of the gut and portraits the disease course of human IBD. Recent studies revealed insights into biological functions of the CP subunits S100A8 and S100A9 during orchestration of an inflammatory response at mucosal surfaces across organ systems. In this review, we summarise longitudinal evidence for the evolution of CP from biomarker to rheostat of mucosal inflammation and suggest an algorithm for the interpretation of faecal CP in daily clinical practice. We propose that mechanistic insights into the biological function of CP in the gut and beyond may facilitate interpretation of current assays and guide patient-tailored medical therapy in IBD, a concept warranting controlled clinical trials.
Collapse
Affiliation(s)
- Almina Jukic
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Latifa Bakiri
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Erwin F Wagner
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
9
|
Charles E, Dumont BL, Bonneau S, Neagoe PE, Villeneuve L, Räkel A, White M, Sirois MG. Angiopoietin 1 release from human neutrophils is independent from neutrophil extracellular traps (NETs). BMC Immunol 2021; 22:51. [PMID: 34344299 PMCID: PMC8336418 DOI: 10.1186/s12865-021-00442-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 07/20/2021] [Indexed: 11/24/2022] Open
Abstract
Background Neutrophils induce the synthesis and release of angiopoietin 1 (Ang1), a cytosolic growth factor involved in angiogenesis and capable of inducing several pro-inflammatory activities in neutrophils. Neutrophils also synthesize and release neutrophil extracellular traps (NETs), comprised from decondensed nuclear DNA filaments carrying proteins such as neutrophil elastase (NE), myeloperoxidase (MPO), proteinase 3 (PR3) and calprotectin (S100A8/S100A9), which together, contribute to the innate immune response against pathogens (e.g., bacteria). NETs are involved in various pathological conditions through pro-inflammatory, pro-thrombotic and endothelial dysfunction effects and have recently been found in heart failure (HF) and type 2 diabetes (T2DM) patients. The aim of the present study was to investigate the role of NETs on the synthesis and release of Ang1 by the neutrophils in patients with T2DM and HF with preserved ejection fraction (HFpEF) (stable or acute decompensated; ADHFpEF) with or without T2DM. Results Our data show that at basal level (PBS) and upon treatment with LPS, levels of NETs are slightly increased in patients suffering from T2DM, HFpEF ± T2DM and ADHF without (w/o) T2DM, whereas this increase was significant in ADHFpEF + T2DM patients compared to healthy control (HC) volunteers and ADHFpEF w/o T2DM. We also observed that treatments with PMA or A23187 increase the synthesis of Ang1 (from 150 to 250%) in HC and this effect is amplified in T2DM and in all cohorts of HF patients. Ang1 is completely released (100%) by neutrophils of all groups and does not bind to NETs as opposed to calprotectin. Conclusions Our study suggests that severely ill patients with HFpEF and diabetes synthesize and release a greater abundance of NETs while Ang1 exocytosis is independent of NETs synthesis. Supplementary Information The online version contains supplementary material available at 10.1186/s12865-021-00442-8.
Collapse
Affiliation(s)
- Elcha Charles
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada.,Department of Pharmacology and Physiology , Université de Montréal, Montreal, QC, Canada
| | - Benjamin L Dumont
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada.,Department of Pharmacology and Physiology , Université de Montréal, Montreal, QC, Canada
| | - Steven Bonneau
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada.,Department of Pharmacology and Physiology , Université de Montréal, Montreal, QC, Canada
| | - Paul-Eduard Neagoe
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada
| | - Louis Villeneuve
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada
| | - Agnès Räkel
- Department of Medicine, Université de Montréal, Montreal, QC, Canada.,Faculty of Medicine, and Research Center-Centre Hospitalier de l'Université de Montréal (CHUM), Université de Montréal, Montreal, QC, Canada
| | - Michel White
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada. .,Department of Medicine, Université de Montréal, Montreal, QC, Canada.
| | - Martin G Sirois
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada. .,Department of Pharmacology and Physiology , Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
10
|
S100A9/CD163 expression profiles in classical monocytes as biomarkers to discriminate idiopathic pulmonary fibrosis from idiopathic nonspecific interstitial pneumonia. Sci Rep 2021; 11:12135. [PMID: 34108546 PMCID: PMC8190107 DOI: 10.1038/s41598-021-91407-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 05/04/2021] [Indexed: 12/22/2022] Open
Abstract
Circulating monocytes have pathogenic relevance in idiopathic pulmonary fibrosis (IPF). Here, we determined whether the cell surface levels of two markers, pro-inflammatory-related S100A9 and anti-inflammatory-related CD163, expressed on CD14strongCD16− classical monocytes by flow cytometry could discriminate IPF from idiopathic nonspecific interstitial pneumonia (iNSIP). Twenty-five patients with IPF, 25 with iNSIP, and 20 healthy volunteers were prospectively enrolled in this study. The S100A9+CD163− cell percentages in classical monocytes showed a pronounced decrease on monocytes in iNSIP compared to that in IPF. In contrast, the percentages of S100A9−CD163+ cells were significantly higher in iNSIP patients than in IPF patients and healthy volunteers. In IPF patients, there was a trend toward a correlation between the percentage of S100A9+CD163− monocytes and the surfactant protein-D (SP-D) serum levels (r = 0.4158, [95% confidence interval (CI) − 0.02042–0.7191], p = 0.051). The individual percentages of S100A9+CD163− and S100A9−CD163+ cells were also independently associated with IPF through multivariate regression analysis. The unadjusted area under the receiver operating characteristic curve (ROC-AUC) to discriminate IPF from iNSIP was (ROC-AUC 0.802, 95% CI [0.687–0.928]), suggesting that these are better biomarkers than serum SP-D (p < 0.05). This preliminary study reports the first comparative characterization of monocyte phenotypes between IPF and iNSIP.
Collapse
|
11
|
Otazu GK, Dayyani M, Badie B. Role of RAGE and Its Ligands on Inflammatory Responses to Brain Tumors. Front Cell Neurosci 2021; 15:770472. [PMID: 34975408 PMCID: PMC8716782 DOI: 10.3389/fncel.2021.770472] [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: 09/03/2021] [Accepted: 11/22/2021] [Indexed: 12/14/2022] Open
Abstract
Gliomas, the most common form of brain cancer, can range from relatively slow-growing low-grade to highly aggressive glioblastoma that has a median overall survival of only 15 months despite multimodal standard therapy. Although immunotherapy with checkpoint inhibitors has significantly improved patient survival for some cancers, to date, these agents have not shown consistent efficacy against malignant gliomas. Therefore, there is a pressing need to better understand the impact of host inflammatory responses on the efficacy of emerging immunotherapy approaches for these resistant tumors. RAGE is a multi-ligand pattern recognition receptor that is activated in various inflammatory states such as diabetes, Alzheimer's disease, cystic fibrosis, and cancer. Low levels of RAGE can be found under normal physiological conditions in neurons, immune cells, activated endothelial, and vascular smooth muscle cells, but it is over-expressed under chronic inflammation due to the accumulation of its ligands. RAGE binds to a range of damage-associated molecular pattern molecules (DAMPs) including AGEs, HMGB1, S100s, and DNA which mediate downstream cellular responses that promote tumor growth, angiogenesis, and invasion. Both in vitro and in vivo studies have shown that inhibition of RAGE signaling can disrupt inflammation and cancer progression and metastasis. Here, we will review our current understanding of the role of RAGE pathway on glioma progression and how it could be exploited to improve the efficacy of immunotherapy approaches.
Collapse
Affiliation(s)
- Griffith Kyle Otazu
- Division of Neurosurgery, City of Hope Beckman Research Institute and Medical Center, Duarte, CA, United States
| | - Mojtaba Dayyani
- Division of Neurosurgery, City of Hope Beckman Research Institute and Medical Center, Duarte, CA, United States
| | - Behnam Badie
- Division of Neurosurgery, City of Hope Beckman Research Institute and Medical Center, Duarte, CA, United States
| |
Collapse
|
12
|
Harman JL, Loes AN, Warren GD, Heaphy MC, Lampi KJ, Harms MJ. Evolution of multifunctionality through a pleiotropic substitution in the innate immune protein S100A9. eLife 2020; 9:e54100. [PMID: 32255429 PMCID: PMC7213983 DOI: 10.7554/elife.54100] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/03/2020] [Indexed: 12/16/2022] Open
Abstract
Multifunctional proteins are evolutionary puzzles: how do proteins evolve to satisfy multiple functional constraints? S100A9 is one such multifunctional protein. It potently amplifies inflammation via Toll-like receptor four and is antimicrobial as part of a heterocomplex with S100A8. These two functions are seemingly regulated by proteolysis: S100A9 is readily degraded, while S100A8/S100A9 is resistant. We take an evolutionary biochemical approach to show that S100A9 evolved both functions and lost proteolytic resistance from a weakly proinflammatory, proteolytically resistant amniote ancestor. We identify a historical substitution that has pleiotropic effects on S100A9 proinflammatory activity and proteolytic resistance but has little effect on S100A8/S100A9 antimicrobial activity. We thus propose that mammals evolved S100A8/S100A9 antimicrobial and S100A9 proinflammatory activities concomitantly with a proteolytic 'timer' to selectively regulate S100A9. This highlights how the same mutation can have pleiotropic effects on one functional state of a protein but not another, thus facilitating the evolution of multifunctionality.
Collapse
Affiliation(s)
- Joseph L Harman
- Department of Chemistry and Biochemistry, University of OregonEugeneUnited States
- Institute of Molecular Biology, University of OregonEugeneUnited States
| | - Andrea N Loes
- Department of Chemistry and Biochemistry, University of OregonEugeneUnited States
- Institute of Molecular Biology, University of OregonEugeneUnited States
| | - Gus D Warren
- Department of Chemistry and Biochemistry, University of OregonEugeneUnited States
- Institute of Molecular Biology, University of OregonEugeneUnited States
| | - Maureen C Heaphy
- Department of Chemistry and Biochemistry, University of OregonEugeneUnited States
- Institute of Molecular Biology, University of OregonEugeneUnited States
| | | | - Michael J Harms
- Department of Chemistry and Biochemistry, University of OregonEugeneUnited States
- Institute of Molecular Biology, University of OregonEugeneUnited States
| |
Collapse
|
13
|
Melief J, Pico de Coaña Y, Maas R, Fennemann FL, Wolodarski M, Hansson J, Kiessling R. High expression of ID1 in monocytes is strongly associated with phenotypic and functional MDSC markers in advanced melanoma. Cancer Immunol Immunother 2020; 69:513-522. [PMID: 31953577 PMCID: PMC7113206 DOI: 10.1007/s00262-019-02476-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 12/31/2019] [Indexed: 12/15/2022]
Abstract
The efficacy of immunotherapies for malignant melanoma is severely hampered by local and systemic immunosuppression mediated by myeloid-derived suppressor cells (MDSC). Inhibitor of differentiation 1 (ID1) is a transcriptional regulator that was shown to be centrally involved in the induction of immunosuppressive properties in myeloid cells in mice, while it was overexpressed in CD11b+ cells in the blood of late-stage melanoma patients. Therefore, we comprehensively assessed ID1 expression in PBMC from stage III and IV melanoma patients, and studied ID1 regulation in models for human monocyte differentiation towards monocyte-derived dendritic cells. A highly significant elevation of ID1 was observed in CD33+CD11b+CD14+HLA-DRlow monocytic MDSC in the blood of melanoma patients compared to their HLA-DRhigh counterparts, while expression of ID1 correlated positively with established MDSC markers S100A8/9 and iNOS. Moreover, expression of ID1 in monocytes significantly decreased in PBMC samples taken after surgical removal of melanoma metastases, compared to those taken before surgery. Finally, maturation of monocyte-derived DC coincided with a significant downregulation of ID1. Together, these data indicate that increased ID1 expression is strongly associated with expression of phenotypic and immunosuppressive markers of monocytic MDSC, while downregulation is associated with a more immunogenic myeloid phenotype. As such, ID1 may be an additional phenotypic marker for monocytic MDSC. Investigation of ID1 as a pharmacodynamic biomarker or its use as a target for modulating MDSC is warranted.
Collapse
Affiliation(s)
- Jeroen Melief
- Department of Oncology-Pathology, Karolinska Institute, Visionsgatan 4, 171 64 Solna, Stockholm, Sweden.
| | - Yago Pico de Coaña
- Department of Oncology-Pathology, Karolinska Institute, Visionsgatan 4, 171 64 Solna, Stockholm, Sweden
| | - Roeltje Maas
- Department of Oncology-Pathology, Karolinska Institute, Visionsgatan 4, 171 64 Solna, Stockholm, Sweden.,Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Felix-Lennart Fennemann
- Department of Oncology-Pathology, Karolinska Institute, Visionsgatan 4, 171 64 Solna, Stockholm, Sweden.,Department of Tumor Immunology, Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maria Wolodarski
- Department of Oncology-Pathology, Karolinska Institute, Visionsgatan 4, 171 64 Solna, Stockholm, Sweden.,Karolinska University Hospital Solna, Stockholm, Sweden
| | - Johan Hansson
- Department of Oncology-Pathology, Karolinska Institute, Visionsgatan 4, 171 64 Solna, Stockholm, Sweden
| | - Rolf Kiessling
- Department of Oncology-Pathology, Karolinska Institute, Visionsgatan 4, 171 64 Solna, Stockholm, Sweden
| |
Collapse
|
14
|
Akiyama M, Zeisbrich M, Ibrahim N, Ohtsuki S, Berry GJ, Hwang PH, Goronzy JJ, Weyand CM. Neutrophil Extracellular Traps Induce Tissue-Invasive Monocytes in Granulomatosis With Polyangiitis. Front Immunol 2019; 10:2617. [PMID: 31798577 PMCID: PMC6874157 DOI: 10.3389/fimmu.2019.02617] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 10/21/2019] [Indexed: 12/20/2022] Open
Abstract
Objective: Granulomatosis with polyangiitis (GPA) is a multi-organ vasculitic syndrome typically associated with neutrophil extracellular trap (NET) formation and aggressive tissue inflammation. Manifestations in head and neck (H&N) GPA include septal perforations, saddle-nose deformities, bony erosions of the orbital and sinus walls, middle ear damage and epiglottitis, indicative of bone, cartilage, and connective tissue destruction. Whether H&N-centric lesions engage disease pathways distinctive from the ischemic tissue damage in the lungs, kidneys, skin, and peripheral nerves is unknown. We have compared inflammatory responses triggered by neutrophilic NETs in patients with H&N GPA and systemic GPA (sGPA). Methods: Neutrophils and monocytes were isolated from the peripheral blood of patients with H&N GPA, sGPA, and age/gender matched healthy individuals. Neutrophil NETosis was induced. NETs were isolated and cocultured with monocytes. Gene induction was quantified by RT-PCR, protein upregulation by flow cytometry. Tissue invasiveness of monocytes was measured in a 3D collagen matrix system. Expression of MMP-9 in tissue-residing macrophages was assessed by immunohistochemistry in tissue biopsies. Results: Neutrophils from H&N GPA patients showed more intense NETosis with higher frequencies of netting neutrophils (P < 0.001) and release of higher amounts of NETs (P < 0.001). Isolated NETs from H&N GPA functioned as an inducer of danger-associated molecular patterns in monocytes; specifically, alarmin S100A9. NET-induced upregulation of monocyte S100A9 required recognition of DNA. S100A9 release resulted in the induction of metalloproteinases, including MMP-9, and enabled monocytes to invade into extracellular matrix. Anti-MMP-9 treatment attenuated the tissue invasiveness of monocytes primed with NETs from H&N GPA patients. MMP-9-producing macrophages dominated the tissue infiltrates in naso-sinal biopsies from H&N GPA patients. Conclusion: Distinct disease patterns in GPA are associated with differences in NET formation and NET content. H&N GPA patients with midline cartilaginous and bony lesions are highly efficient in generating NETs. H&N GPA neutrophils trigger the induction of the alarmin S100A9, followed by production of MMP-9, endowing monocytes with tissue-invasive capabilities.
Collapse
Affiliation(s)
- Mitsuhiro Akiyama
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Markus Zeisbrich
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Nour Ibrahim
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Stanford, CA, United States
| | - Shozo Ohtsuki
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Gerald J Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Peter H Hwang
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Stanford, CA, United States
| | - Jörg J Goronzy
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Cornelia M Weyand
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
15
|
Nys G, Cobraiville G, Servais AC, Malaise MG, de Seny D, Fillet M. Targeted proteomics reveals serum amyloid A variants and alarmins S100A8-S100A9 as key plasma biomarkers of rheumatoid arthritis. Talanta 2019; 204:507-517. [PMID: 31357327 DOI: 10.1016/j.talanta.2019.06.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/07/2019] [Accepted: 06/10/2019] [Indexed: 01/18/2023]
Abstract
Serum amyloid A (SAA) and S100 (S100A8, S100A9 and S100A12) proteins were previously identified as biomarkers of interest for rheumatoid arthritis (RA). Among SAA family, two closely related isoforms (SAA-1 and SAA-2) are linked to the acute-phase of inflammation. They respectively exist under the form of three (α, β, and γ) and two (α and β) allelic variants. We developed a single run quantitative method for these protein variants and investigated their clinical relevance in the context of RA. The method was developed and validated according to regulations before being applied on plasma coming from RA patients (n = 46), other related inflammatory pathologies (n = 116) and controls (n = 62). Depending on the activity score of RA, SAA1 isoforms (mainly of SAA1α and SAA1β subtypes) were found to be differentially present in plasma revealing their dual role during the development of RA. In addition, the weight of SAA1α in the total SAA response varied from 32 to 80% depending on the pathology studied. A negative correlation between SAA1α and SAA1β was also highlighted for RA early-onset (r = -0.41). SAA2 and S100A8/S100A9 proteins were significantly overexpressed compared to control samples regardless of RA stage. The pathophysiological relevance of these quantitative and qualitative characteristics of the SAA response remains unknown. However, the significant negative correlation observed between SAA1α and SAA1β levels in RA early-onset suggests the existence of still unknown regulatory mechanisms in these diseases.
Collapse
Affiliation(s)
- Gwenaël Nys
- Laboratory for the Analysis of Medicines, Center for Interdisciplinary Research on Medicines (CIRM), ULiege, Quartier Hopital, Avenue Hippocrate 15, 4000 Liege, Belgium
| | - Gaël Cobraiville
- Laboratory for the Analysis of Medicines, Center for Interdisciplinary Research on Medicines (CIRM), ULiege, Quartier Hopital, Avenue Hippocrate 15, 4000 Liege, Belgium; Laboratory of Rheumatology, GIGA-Inflammation, Infection & Immunity, ULiege and CHU de Liege, Quartier Hopital, Avenue Hippocrate 15, 4000 Liege, Belgium
| | - Anne-Catherine Servais
- Laboratory for the Analysis of Medicines, Center for Interdisciplinary Research on Medicines (CIRM), ULiege, Quartier Hopital, Avenue Hippocrate 15, 4000 Liege, Belgium
| | - Michel G Malaise
- Laboratory of Rheumatology, GIGA-Inflammation, Infection & Immunity, ULiege and CHU de Liege, Quartier Hopital, Avenue Hippocrate 15, 4000 Liege, Belgium
| | - Dominique de Seny
- Laboratory of Rheumatology, GIGA-Inflammation, Infection & Immunity, ULiege and CHU de Liege, Quartier Hopital, Avenue Hippocrate 15, 4000 Liege, Belgium
| | - Marianne Fillet
- Laboratory for the Analysis of Medicines, Center for Interdisciplinary Research on Medicines (CIRM), ULiege, Quartier Hopital, Avenue Hippocrate 15, 4000 Liege, Belgium.
| |
Collapse
|
16
|
Lim RR, Vaidya T, Gadde SG, Yadav NK, Sethu S, Hainsworth DP, Mohan RR, Ghosh A, Chaurasia SS. Correlation between systemic S100A8 and S100A9 levels and severity of diabetic retinopathy in patients with type 2 diabetes mellitus. Diabetes Metab Syndr 2019; 13:1581-1589. [PMID: 31336525 DOI: 10.1016/j.dsx.2019.03.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 03/05/2019] [Indexed: 02/06/2023]
Abstract
AIMS S100A8 and S100A9 are myeloid-related damage-associated molecular patterns (DAMPs) primarily involved in the modulation of innate immune response to cellular injury. This study evaluated the correlation between circulating concentrations of S100A8 and S100A9 proteins with the severity of diabetic retinopathy (DR) in patients with type 2 diabetes (T2DM). METHODS T2DM patients with HbA1c levels >7%, fasting blood glucose >126 mg/dl and history of diabetes were included in this study. DR severity was graded based on ETDRS and Gloucestershire classifications. Plasma samples were evaluated for S100A8 and S100A9 levels using ELISA. RESULTS In this comparative study, DR patients (n = 89) had increased plasma S100A8 and S100A9 proteins compared to age-matched T2DM controls (n = 28), which was directly related to the severity of DR. Female DR subjects had increased S100A8 expression compared to their male counterparts. Substantial retention of S100A8 and S100A9 production was seen in DR patients above 50 years of age. Duration of T2DM was not found to affect protein levels, however T2DM onset at >50 years old significantly increased S100A8 and S100A9 concentrations. CONCLUSIONS Our findings suggest that systemic circulation levels of S100A8 and S100A9 are correlated with the progression of DR in T2DM patients, indicating their potential role in DR pathogenesis.
Collapse
Affiliation(s)
- Rayne R Lim
- Ocular Immunology and Angiogenesis Lab, Department of Veterinary Medicine & Surgery, University of Missouri, Columbia, MO, 65211, USA; Department of Biomedical Sciences, University of Missouri, Columbia, MO, 65211, USA; Harry S. Truman Memorial Veteran Hospital, Columbia, MO, 65201, USA
| | - Tanuja Vaidya
- GROW Research Laboratory, Narayana Nethralaya, Bangalore, 560099, India
| | - Santosh G Gadde
- Vitreoretina Department, Narayana Nethralaya, Bangalore, 560099, India
| | - Naresh K Yadav
- Vitreoretina Department, Narayana Nethralaya, Bangalore, 560099, India
| | - Swaminathan Sethu
- GROW Research Laboratory, Narayana Nethralaya, Bangalore, 560099, India
| | - Dean P Hainsworth
- Vitreoretinal Service, Ophthalmology, Mason Eye Institute, University of Missouri, Columbia, MO, 65211, USA
| | - Rajiv R Mohan
- Ocular Immunology and Angiogenesis Lab, Department of Veterinary Medicine & Surgery, University of Missouri, Columbia, MO, 65211, USA; Department of Biomedical Sciences, University of Missouri, Columbia, MO, 65211, USA; Harry S. Truman Memorial Veteran Hospital, Columbia, MO, 65201, USA
| | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Nethralaya, Bangalore, 560099, India.
| | - Shyam S Chaurasia
- Ocular Immunology and Angiogenesis Lab, Department of Veterinary Medicine & Surgery, University of Missouri, Columbia, MO, 65211, USA; Department of Biomedical Sciences, University of Missouri, Columbia, MO, 65211, USA; Harry S. Truman Memorial Veteran Hospital, Columbia, MO, 65201, USA.
| |
Collapse
|
17
|
Proteomic features of delayed ocular symptoms caused by exposure to sulfur mustard: As studied by protein profiling of corneal epithelium. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2017; 1865:1445-1454. [DOI: 10.1016/j.bbapap.2017.08.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 08/11/2017] [Accepted: 08/31/2017] [Indexed: 12/21/2022]
|
18
|
S100-A9 protein in exosomes from chronic lymphocytic leukemia cells promotes NF-κB activity during disease progression. Blood 2017; 130:777-788. [DOI: 10.1182/blood-2017-02-769851] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 05/29/2017] [Indexed: 12/20/2022] Open
Abstract
Key Points
Plasma-derived exosomes from patients with CLL exhibit different protein cargo compositions depending on disease status and progression. S100-A9 protein is overexpressed and S100-A9 cargo in exosomes activates NF-κB pathway in patients with CLL during disease progression.
Collapse
|
19
|
Cai Y, Shi Z, Bai Y. Review of Rosai-Dorfman Disease: New Insights into the Pathogenesis of This Rare Disorder. Acta Haematol 2017; 138:14-23. [PMID: 28614806 DOI: 10.1159/000475588] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 04/10/2017] [Indexed: 12/22/2022]
Abstract
Rosai-Dorfman disease (RDD) is a rare histiocytosis typically with bilateral painless cervical lymphadenopathy. Laboratory data are nonspecific, and the presence of emperipolesis in large foamy S-100+ CD1a- histiocytes is the prominent histologic feature. The pathogenesis of RDD still remains elusive. According to published studies, we propose that RDD cells might represent intermediate recruiting monocytes with differentiation blockade. Both disturbance of homoeostasis and inherent genomic alterations could contribute to initiation of the disorder through signal transduction. Several inflammatory molecules such as macrophage colony-stimulating factor, IL-1β, IL-6, and tumor necrosis factor-α also play a pivotal role in the development of this rare entity. Additional studies are needed to further elucidate the essence of the disease.
Collapse
Affiliation(s)
- Yanan Cai
- Department of Hematology/Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
| | | | | |
Collapse
|
20
|
Kennedy DE, Knight KL. Inflammatory Changes in Bone Marrow Microenvironment Associated with Declining B Lymphopoiesis. THE JOURNAL OF IMMUNOLOGY 2017; 198:3471-3479. [PMID: 28320833 DOI: 10.4049/jimmunol.1601643] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 02/27/2017] [Indexed: 12/22/2022]
Abstract
B lymphopoiesis arrests precipitously in rabbits such that by 2-4 mo of age, before sexual maturity, little to no B lymphopoiesis occurs in the bone marrow (BM). Previously, we showed that in mice, adipocytes inhibit B lymphopoiesis in vitro by inducing inflammatory myeloid cells, which produce IL-1β. In this study, we characterized rabbit BM after the arrest of B lymphopoiesis and found a dramatic increase in fat, increased CD11b+ myeloid cells, and upregulated expression of the inflammatory molecules, IL-1β and S100A9, by the myeloid cells. We added BM fat, CD11b+ myeloid cells, and recombinant S100A9 to B lymphopoiesis cultures and found that they inhibited B lymphopoiesis and enhanced myelopoiesis. Unlike IL-1β, which inhibits B lymphopoiesis by acting on early lymphoid progenitors, S100A9 inhibits B lymphopoiesis by acting on myeloid cells and promoting the release of inflammatory molecules, including IL-1β. Many molecules produced by adipocytes activate the NLRP3 inflammasome, and the NLRP3 inhibitor, glibenclamide, restored B lymphopoiesis and minimized induction of myeloid cells induced by adipocyte-conditioned medium in vitro. We suggest that fat provides an inflammatory microenvironment in the BM and promotes/activates myeloid cells to produce inflammatory molecules such as IL-1β and S100A9, which negatively regulate B lymphopoiesis.
Collapse
Affiliation(s)
- Domenick E Kennedy
- Department of Microbiology and Immunology, Loyola University, Chicago, IL 60153
| | - Katherine L Knight
- Department of Microbiology and Immunology, Loyola University, Chicago, IL 60153
| |
Collapse
|
21
|
Allaoui R, Bergenfelz C, Mohlin S, Hagerling C, Salari K, Werb Z, Anderson RL, Ethier SP, Jirström K, Påhlman S, Bexell D, Tahin B, Johansson ME, Larsson C, Leandersson K. Cancer-associated fibroblast-secreted CXCL16 attracts monocytes to promote stroma activation in triple-negative breast cancers. Nat Commun 2016; 7:13050. [PMID: 27725631 PMCID: PMC5062608 DOI: 10.1038/ncomms13050] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 08/22/2016] [Indexed: 02/06/2023] Open
Abstract
Triple-negative (TN) breast cancers (ER−PR−HER2−) are highly metastatic and associated with poor prognosis. Within this subtype, invasive, stroma-rich tumours with infiltration of inflammatory cells are even more aggressive. The effect of myeloid cells on reactive stroma formation in TN breast cancer is largely unknown. Here, we show that primary human monocytes have a survival advantage, proliferate in vivo and develop into immunosuppressive myeloid cells expressing the myeloid-derived suppressor cell marker S100A9 only in a TN breast cancer environment. This results in activation of cancer-associated fibroblasts and expression of CXCL16, which we show to be a monocyte chemoattractant. We propose that this migratory feedback loop amplifies the formation of a reactive stroma, contributing to the aggressive phenotype of TN breast tumours. These insights could help select more suitable therapies targeting the stromal component of these tumours, and could aid prediction of drug resistance. A reactive tumour stroma is associated with poor prognosis. Here, the authors show that in patients with triple negative breast cancer resident monocytes activate cancer-associated fibroblasts and induce production of CXCL16, which acts as a monocyte chemoattractant, resulting in an amplificatory feedback loop.
Collapse
Affiliation(s)
- Roni Allaoui
- Department of Translational Medicine, Cancer Immunology, Lund University, Malmö 205 02, Sweden
| | - Caroline Bergenfelz
- Department of Translational Medicine, Cancer Immunology, Lund University, Malmö 205 02, Sweden
| | - Sofie Mohlin
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund 223 63, Sweden
| | - Catharina Hagerling
- Department of Translational Medicine, Cancer Immunology, Lund University, Malmö 205 02, Sweden.,Department of Anatomy and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California 94143-0452, USA
| | - Kiarash Salari
- Department of Anatomy and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California 94143-0452, USA
| | - Zena Werb
- Department of Anatomy and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California 94143-0452, USA
| | - Robin L Anderson
- Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre, The University of Melbourne, Melbourne 8006, Australia
| | - Stephen P Ethier
- Department of Pathology and Laboratory Medicine, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | - Karin Jirström
- Department of Clinical Sciences Lund, Oncology and Pathology, Lund University, Lund 221 85, Sweden
| | - Sven Påhlman
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund 223 63, Sweden
| | - Daniel Bexell
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund 223 63, Sweden
| | - Balázs Tahin
- Department of Translational Medicine, Clinical Pathology, Skånes Universitetssjukhus, Malmö 205 02, Sweden
| | - Martin E Johansson
- Department of Translational Medicine, Cancer Immunology, Lund University, Malmö 205 02, Sweden.,Department of Translational Medicine, Clinical Pathology, Skånes Universitetssjukhus, Malmö 205 02, Sweden
| | - Christer Larsson
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund 223 63, Sweden
| | - Karin Leandersson
- Department of Translational Medicine, Cancer Immunology, Lund University, Malmö 205 02, Sweden
| |
Collapse
|
22
|
Interaction of an esophageal MEG protein from schistosomes with a human S100 protein involved in inflammatory response. Biochim Biophys Acta Gen Subj 2016; 1861:3490-3497. [PMID: 27639541 DOI: 10.1016/j.bbagen.2016.09.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 08/09/2016] [Accepted: 09/01/2016] [Indexed: 11/24/2022]
Abstract
BACKGROUND The Micro-Exon Gene-14 (MEG-14) displays a remarkable structure that allows the generation of antigenic variation in Schistosomes. Previous studies showed that the soluble portion of the MEG-14 protein displays features of an intrinsically disordered protein and is expressed exclusively in the parasite esophageal gland. These features indicated a potential for interaction with host proteins present in the plasma and cells from ingested blood. METHODS A yeast two-hybrid experiment using as bait the soluble domain of Schistosoma mansoni MEG-14 (sMEG-14) against a human leukocyte cDNA library was performed. Pull-down and surface plasmon resonance (SPR) experiments were used to validate the interaction between sMEG-14 and human S100A9. Synchrotron radiation circular dichroism (SRCD) were used to detect structural changes upon interaction between sMEG-14 and human S100A9. Feeding of live parasites with S100A9 attached to a fluorophore allowed the tracking of the fate of this protein in the parasite digestive system. RESULTS S100A9 interacted with sMEG-14 consistently in yeast two-hybrid assay, pull-down and SPR experiments. SRCD suggested that MEG-14 acquired a more regular structure as a result of the interaction with S100A9. Accumulation of recombinant S100A9 in the parasite's esophageal gland, when ingested by live worms suggests that such interaction may occur in vivo. CONCLUSION S100A9, a protein previously described to be involved in modulation of inflammatory response, was found to interact with sMEG-14. GENERAL SIGNIFICANCE Our results allow proposing a mechanism involving MEG-14 for the parasite to block inflammatory signaling, which would occur upon release of S100A9 when ingested blood cells are lysed.
Collapse
|
23
|
Stephan JR, Nolan EM. Calcium-induced Tetramerization and Zinc Chelation Shield Human Calprotectin from Degradation by Host and Bacterial Extracellular Proteases. Chem Sci 2016; 7:1962-1975. [PMID: 26925211 PMCID: PMC4763987 DOI: 10.1039/c5sc03287c] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/17/2015] [Indexed: 12/19/2022] Open
Abstract
Calprotectin (CP, S100A8/S100A9 oligomer, MRP-8/14 oligomer, calgranulins A and B) is a protein component of the innate immune system that contributes to the metal-withholding response by sequestering bioavailable transition metal ions at sites of infection. Human CP employs Ca(II) ions to modulate its quaternary structure, transition metal binding properties, and antimicrobial activity. In this work, we report the discovery that Ca(II)-induced self-association of human CP to afford heterotetramers protects the protein scaffold from degradation by host serine proteases. We present the design and characterization of two new human CP-Ser variants, S100A8(C42S)(I60E)/S100A9(C3S) and S100A8(C42S)(I60K)/S100A9(C3S), that exhibit defective tetramerization properties. Analytical size exclusion chromatography and analytical ultracentrifugation reveal that both variants, hereafter I60E and I60K, persist as heterodimers in the presence of Ca(II) only, and form heterotetramers in the presence of Mn(II) only and both Ca(II) and Mn(II). Coordination to Fe(II) also causes I60E and I60K to form heterotetramers in both the absence and presence of Ca(II). The Ca(II)-bound I60E and I60K heterodimers are readily degraded by trypsin, chymotrypsin and human neutrophil elastase, whereas the Ca(II)-bound CP-Ser heterotetramers and the Ca(II)- and Mn(II)-bound I60E and I60K heterotetramers are resistant to degradation by these host proteases. The staphylococcal extracellular protease GluC cuts the S100A8 subunit of CP-Ser at the C-terminal end of residue 89 to afford a ΔSKHE variant. The GluC cleavage site is in close proximity to the His3Asp metal-binding site, which coordinates Zn(II) with high affinity, and Zn(II) chelation protects the S100A8 subunit from GluC cleavage. Taken together, these results provide new insight into how Ca(II) ions and transition metals modulate the chemistry and biology of CP, and indicate that coordination to divalent cations transforms human CP into a protease-resistant form and enables innate immune function in the hostile conditions of an infection site.
Collapse
Affiliation(s)
- Jules R. Stephan
- Department of Chemistry , Massachusetts Institute of Technology , Cambridge , MA 02139 , USA . ; Fax: +1-617-324-0505 ; Tel: +1-617-452-2495
| | - Elizabeth M. Nolan
- Department of Chemistry , Massachusetts Institute of Technology , Cambridge , MA 02139 , USA . ; Fax: +1-617-324-0505 ; Tel: +1-617-452-2495
| |
Collapse
|
24
|
Chakraborty P, Bjork P, Källberg E, Olsson A, Riva M, Mörgelin M, Liberg D, Ivars F, Leanderson T. Vesicular Location and Transport of S100A8 and S100A9 Proteins in Monocytoid Cells. PLoS One 2015; 10:e0145217. [PMID: 26661255 PMCID: PMC4678419 DOI: 10.1371/journal.pone.0145217] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 11/30/2015] [Indexed: 12/18/2022] Open
Abstract
We show here, by using surface biotinylation, followed by Western blotting or surface plasmon resonance analysis, that very low levels of S100A8 and/or S100A9 can be detected on the surface of THP-1 cells or freshly isolated human monocytes. This was supported by immune-electron microscopy where we observed membrane-associated expression of the proteins restricted to small patches. By using confocal microscopy we could determine that S100A8 and S100A9 protein in THP-1 cells or freshly isolated human monocytes was mostly present in vesicular structures. This finding was confirmed using immune-electron microscopy. Subcellular fractionation and confocal microscopy showed that these vesicular structures are mainly early endosomes and endolysosomes. Our subsequent studies showed that accumulation of S100A8 and S100A9 in the endolysosomal compartment is associated with induction of their release from the cells. Furthermore, an inhibitor of lysosomal activity could modulate the release of S100A8 and S100A9 in the extracellular milieu. Our current results suggest that the S100A8 and S100A9 proteins are primarily associated with certain kinds of cytosolic vesicles and may be secreted via an endolysosomal pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Tomas Leanderson
- Immunology Group, Lund University, Lund, Sweden
- Active Biotech AB, Lund, Sweden
- * E-mail:
| |
Collapse
|
25
|
Van Crombruggen K, Vogl T, Pérez-Novo C, Holtappels G, Bachert C. Differential release and deposition of S100A8/A9 proteins in inflamed upper airway tissue. Eur Respir J 2015; 47:264-74. [DOI: 10.1183/13993003.00159-2015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 07/04/2015] [Indexed: 12/15/2022]
Abstract
Intracellular Ca2+-binding S100A8/A9 proteins gain novel functions when released during inflammation. The exact outcome of their extracellular function depends on the local tissue environment in which they are released; both anti-inflammatory and pro-inflammatory responses are described, modulating the immune system by binding Toll-like receptor (TLR)-4 or the receptor for advanced glycation end-products (RAGE). However, the contribution of the proteins in the pathophysiology of chronic rhinosinusitis (CRS) remains unclear.Homomeric S100A8 and S100A9, and heteromeric S100A8/A9 proteins were evaluated in CRS with/without nasal polyps (CRSw/sNP) and controls. Functional responses were assessed in polyp tissue stimulated with S100 proteins in the presence of TLR-4 and RAGE blocking antibodies.S100A8, S100A9 and S100A8/A9 protein levels were significantly higher in CRSwNP patients, showing increased deposition on extracellular matrix (ECM) structures of CRSwNP tissue in contrast to CRSsNP and controls. In the presence ofStaphylococcus aureus, S100A8/A9 is released from neutrophils and from the ECM. Extracellular S100A8 and S100A9 proteins induced increased levels of diverse inflammatory mediatorsviaTLR-4 engagement.The inflammatory/remodelling characteristics of CRSwNP specifically allow increased retention of S100A8, S100A9 and S100A8/A9 proteins in the ECM of CRSwNP tissue. Upon release, homodimeric proteins act as a local danger signal inducing inflammatory mediators, predominantlyviaTLR-4 activation.
Collapse
|
26
|
S100A9 expressed in ER(-)PgR(-) breast cancers induces inflammatory cytokines and is associated with an impaired overall survival. Br J Cancer 2015; 113:1234-43. [PMID: 26448179 PMCID: PMC4647879 DOI: 10.1038/bjc.2015.346] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 08/03/2015] [Accepted: 09/04/2015] [Indexed: 12/30/2022] Open
Abstract
Background: Breast cancer is the most common cancer form among women today. Depending on hormone receptor status, breast cancers are divided into different subtypes with vastly varying prognosis. S100A9 is a calcium-binding protein that is associated with inflammation and expressed not only in myeloid cells but also in some tumours. The role for S100A9 in the malignant cells is not well characterised; however, previous studies have shown that the protein could have important immune-modulating properties. Methods: Using a human breast cancer cohort consisting of 144 tumour samples and in vitro analysis of human breast cancer cell lines, we investigated the expression and function of S100A9 in human breast cancer. Results: We show that S100A9 expression in breast cancer correlated with the ER−PgR− breast tumour subtype (P<0.001) and with Ki67 (P=0.024) and was expressed both in the malignant cells and in the tumour-infiltrating anti-inflammatory CD163+ myeloid cells (P<0.001). Stromal expression of S100A9 also correlated to nodal stage, tumour size and Her2 positivity. Within the ER−PgR− subgroup, all Her2+ and EGFR+ tumours expressed S100A9 in the cytoplasm. Both cytoplasmic staining in the malignant cells as well as stromal S100A9 expression in myeloid cells correlated with a decreased overall survival in breast cancer patients. Furthermore, rS100A9 homodimers induced expression of pro-inflammatory cytokines (IL-6, IL-8 and IL-1β) in a TLR4- and EGFR-dependent manner in human breast cancer cells in vitro. Conclusion: We suggest that S100A9 could be viewed as a novel therapeutic target for patients with ER−PgR− breast cancers.
Collapse
|
27
|
Chen B, Miller AL, Rebelatto M, Brewah Y, Rowe DC, Clarke L, Czapiga M, Rosenthal K, Imamichi T, Chen Y, Chang CS, Chowdhury PS, Naiman B, Wang Y, Yang D, Humbles AA, Herbst R, Sims GP. S100A9 induced inflammatory responses are mediated by distinct damage associated molecular patterns (DAMP) receptors in vitro and in vivo. PLoS One 2015; 10:e0115828. [PMID: 25706559 PMCID: PMC4338059 DOI: 10.1371/journal.pone.0115828] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 12/02/2014] [Indexed: 01/12/2023] Open
Abstract
Release of endogenous damage associated molecular patterns (DAMPs), including members of the S100 family, are associated with infection, cellular stress, tissue damage and cancer. The extracellular functions of this family of calcium binding proteins, particularly S100A8, S100A9 and S100A12, are being delineated. They appear to mediate their functions via receptor for advanced glycation endproducts (RAGE) or TLR4, but there remains considerable uncertainty over the relative physiological roles of these DAMPs and their pattern recognition receptors. In this study, we surveyed the capacity of S100 proteins to induce proinflammatory cytokines and cell migration, and the contribution RAGE and TLR4 to mediate these responses in vitro. Using adenoviral delivery of murine S100A9, we also examined the potential for S100A9 homodimers to trigger lung inflammation in vivo. S100A8, S100A9 and S100A12, but not the S100A8/A9 heterodimer, induced modest levels of TLR4-mediated cytokine production from human PBMC. In contrast, for most S100s including S100A9, RAGE blockade inhibited S100-mediated cell migration of THP1 cells and major leukocyte populations, whereas TLR4-blockade had no effect. Intranasal administration of murine S100A9 adenovirus induced a specific, time-dependent predominately macrophage infiltration that coincided with elevated S100A9 levels and proinflammatory cytokines in the BAL fluid. Inflammatory cytokines were markedly ablated in the TLR4-defective mice, but unexpectedly the loss of TLR4 signaling or RAGE-deficiency did not appreciably impact the S100A9-mediated lung pathology or the inflammatory cell infiltrate in the alveolar space. These data demonstrate that physiological levels of S100A9 homodimers can trigger an inflammatory response in vivo, and despite the capacity of RAGE and TLR4 blockade to inhibit responses in vitro, the response is predominately independent of both these receptors.
Collapse
Affiliation(s)
- Bo Chen
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Allison L. Miller
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Marlon Rebelatto
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Yambasu Brewah
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Daniel C. Rowe
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Lori Clarke
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Meggan Czapiga
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Kim Rosenthal
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Tomozumi Imamichi
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
- Laboratory of Human Retrovirology, Applied and Developmental Directorate, Building 550 Room 126, Leidos Biomedical Research Inc, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, United States of America
| | - Yan Chen
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Chew-Shun Chang
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Partha S. Chowdhury
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Brian Naiman
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Yue Wang
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - De Yang
- Laboratory of Molecular Immunoregulation, National Cancer Institute, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, United States of America
| | - Alison A. Humbles
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Ronald Herbst
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Gary P. Sims
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
- * E-mail:
| |
Collapse
|
28
|
Target-selective protein S-nitrosylation by sequence motif recognition. Cell 2014; 159:623-34. [PMID: 25417112 DOI: 10.1016/j.cell.2014.09.032] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 08/07/2014] [Accepted: 09/10/2014] [Indexed: 11/20/2022]
Abstract
S-nitrosylation is a ubiquitous protein modification emerging as a principal mechanism of nitric oxide (NO)-mediated signal transduction and cell function. S-nitrosylases can use NO synthase (NOS)-derived NO to modify selected cysteines in target proteins. Despite proteomic identification of over a thousand S-nitrosylated proteins, few S-nitrosylases have been identified. Moreover, mechanisms underlying site-selective S-nitrosylation and the potential role of specific sequence motifs remain largely unknown. Here, we describe a stimulus-inducible, heterotrimeric S-nitrosylase complex consisting of inducible NOS (iNOS), S100A8, and S100A9. S100A9 exhibits transnitrosylase activity, shuttling NO from iNOS to the target protein, whereas S100A8 and S100A9 coordinately direct site selection. A family of proteins S-nitrosylated by iNOS-S100A8/A9 were revealed by proteomic analysis. A conserved I/L-X-C-X2-D/E motif was necessary and sufficient for iNOS-S100A8/A9-mediated S-nitrosylation. These results reveal an elusive parallel between protein S-nitrosylation and phosphorylation, namely, stimulus-dependent posttranslational modification of selected targets by primary sequence motif recognition.
Collapse
|
29
|
Sivaprasad U, Kinker KG, Ericksen MB, Lindsey M, Gibson AM, Bass SA, Hershey NS, Deng J, Medvedovic M, Khurana Hershey GK. SERPINB3/B4 contributes to early inflammation and barrier dysfunction in an experimental murine model of atopic dermatitis. J Invest Dermatol 2014; 135:160-169. [PMID: 25111616 PMCID: PMC4268075 DOI: 10.1038/jid.2014.353] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 07/08/2014] [Accepted: 07/31/2014] [Indexed: 01/21/2023]
Abstract
Serine proteases are critical for epidermal barrier homeostasis, and their aberrant expression and/or activity is associated with chronic skin diseases. Elevated levels of the serine protease inhibitors SERPINB3 and SERPINB4 are seen in patients with atopic dermatitis and psoriasis. However, their mechanistic role in the skin is unknown. To evaluate the contribution of Serpinb3a (mouse homolog of SERPINB3 and SERPINB4) in atopic dermatitis, we examined the effect of topical Aspergillus fumigatus extract exposure in wild-type and Serpinb3a-null mice on transepidermal water loss (TEWL), sensitization, and inflammation. Allergen exposure induced Serpinb3a expression in the skin, along with increased TEWL, epidermal thickness, and skin inflammation, all of which were attenuated in the absence of Serpinb3a. Attenuated TEWL correlated with decreased expression of the pro-inflammatory marker S100A8. Silencing of SERPINB3/B4 in human keratinocytes decreased S100A8 expression, supporting a role for SERPINB3/B4 in the initiation of the acute inflammatory response. RNA-seq analysis following allergen exposure identified a network of pro-inflammatory genes induced in wild-type mice that was absent in Serpinb3a-null mice. In conclusion, Serpinb3a deficiency attenuates barrier dysfunction and the early inflammatory response following cutaneous allergen exposure, supporting a role for Serpinb3a (mice) and SERPINB3/B4 (humans) early in atopic dermatitis.
Collapse
Affiliation(s)
- Umasundari Sivaprasad
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Kayla G Kinker
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Mark B Ericksen
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Mark Lindsey
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Aaron M Gibson
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Stacey A Bass
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Nicolas S Hershey
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jingyuan Deng
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA
| | - Mario Medvedovic
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA
| | - Gurjit K Khurana Hershey
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA.
| |
Collapse
|
30
|
Boonla C, Tosukhowong P, Spittau B, Schlosser A, Pimratana C, Krieglstein K. Inflammatory and fibrotic proteins proteomically identified as key protein constituents in urine and stone matrix of patients with kidney calculi. Clin Chim Acta 2013; 429:81-9. [PMID: 24333391 DOI: 10.1016/j.cca.2013.11.036] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 11/27/2013] [Accepted: 11/27/2013] [Indexed: 11/19/2022]
Abstract
To uncover whether urinary proteins are incorporated into stones, the proteomic profiles of kidney stones and urine collected from the same patients have to be explored. We employed 1D-PAGE and nanoHPLC-ESI-MS/MS to analyze the proteomes of kidney stone matrix (n=16), nephrolithiatic urine (n=14) and healthy urine (n=3). We identified 62, 66 and 22 proteins in stone matrix, nephrolithiatic urine and healthy urine, respectively. Inflammation- and fibrosis-associated proteins were frequently detected in the stone matrix and nephrolithiatic urine. Eighteen proteins were exclusively found in the stone matrix and nephrolithiatic urine, considered as candidate biomarkers for kidney stone formation. S100A8 and fibronectin, representatives of inflammation and fibrosis, respectively, were up-regulated in nephrolithiasis renal tissues. S100A8 was strongly expressed in infiltrated leukocytes. Fibronectin was over-expressed in renal tubular cells. S100A8 and fibronectin were immunologically confirmed to exist in nephrolithiatic urine and stone matrix, but in healthy urine they were undetectable. Conclusion, both kidney stones and urine obtained from the same patients greatly contained inflammatory and fibrotic proteins. S100A8 and fibronectin were up-regulated in stone-baring kidneys and nephrolithiatic urine. Therefore, inflammation and fibrosis are suggested to be involved in the formation of kidney calculi.
Collapse
Affiliation(s)
- Chanchai Boonla
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330 Thailand.
| | - Piyaratana Tosukhowong
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330 Thailand
| | - Björn Spittau
- Department of Molecular Embryology, Institute for Anatomy and Cell Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Andreas Schlosser
- Center for Biological Systems Analysis (ZBSA), Core Facility Proteomics, University of Freiburg, 79104 Freiburg, Germany
| | - Chaowat Pimratana
- Division of Urological Surgery, Khon Kaen Hospital, Khon Kaen 40000 Thailand
| | - Kerstin Krieglstein
- Department of Molecular Embryology, Institute for Anatomy and Cell Biology, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
31
|
Cortés-Malagón EM, Bonilla-Delgado J, Díaz-Chávez J, Hidalgo-Miranda A, Romero-Cordoba S, Uren A, Celik H, McCormick M, Munguía-Moreno JA, Ibarra-Sierra E, Escobar-Herrera J, Lambert PF, Mendoza-Villanueva D, Bermudez-Cruz RM, Gariglio P. Gene expression profile regulated by the HPV16 E7 oncoprotein and estradiol in cervical tissue. Virology 2013; 447:155-65. [PMID: 24210110 DOI: 10.1016/j.virol.2013.08.036] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 07/15/2013] [Accepted: 08/30/2013] [Indexed: 12/11/2022]
Abstract
The HPV16 E7 oncoprotein and 17β-estradiol are important factors for the induction of premalignant lesions and cervical cancer. The study of these factors is crucial for a better understanding of cervical tumorigenesis. Here, we assessed the global gene expression profiles induced by the HPV16 E7 oncoprotein and/or 17β-estradiol in cervical tissue of FvB and K14E7 transgenic mice. We found that the most dramatic changes in gene expression occurred in K14E7 and FvB groups treated with 17β-estradiol. A large number of differentially expressed genes involved in the immune response were observed in 17β-estradiol treated groups. The E7 oncoprotein mainly affected the expression of genes involved in cellular metabolism. Our microarray data also identified differentially expressed genes that have not previously been reported in cervical cancer. The identification of genes regulated by E7 and 17β-estradiol, provides the basis for further studies on their role in cervical carcinogenesis.
Collapse
Affiliation(s)
- Enoc M Cortés-Malagón
- Department of Genetics and Molecular Biology, Centro de Investigación y de Estudios Avanzados (Cinvestav), Mexico City 07360, Mexico; Research Unit, Hospital Juárez de México, Mexico City 07760, Mexico.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|