51
|
Leite Dantas R, Bettenworth D, Varga G, Weinhage T, Wami HT, Dobrindt U, Roth J, Vogl T, Ludwig S, Wixler V. Spontaneous onset of TNFα-triggered colonic inflammation depends on functional T lymphocytes, S100A8/A9 alarmins, and MHC H-2 haplotype. J Pathol 2020; 251:388-399. [PMID: 32449525 DOI: 10.1002/path.5473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 05/05/2020] [Accepted: 05/18/2020] [Indexed: 12/17/2022]
Abstract
Recently, we established a doxycycline-inducible human tumor necrosis factor alpha (TNFα)-transgenic mouse line, ihTNFtg. Non-induced young and elderly mice showed low but constitutive expression of hTNFα due to promoter leakiness. The persistently present hTNFα stimulated endogenous pro-inflammatory mouse mS100A8/A9 alarmins. Secreted mS100A8/A9 in turn induced the expression and release of mouse mTNFα. The continuous upregulation of pro-inflammatory mTNFα and mS100A8/A9 proteins, due to their mutual expression dependency, gradually led to increased levels in colon tissue and blood. This finally exceeded the threshold levels tolerated by the healthy organism, leading to the onset of intestinal inflammation. Here, recombinant hTNFα functioned as an initial trigger for the development of chronic inflammation. Crossing ihTNFtg mice with S100A9KO mice lacking active S100A8/A9 alarmins or with Rag1KO mice lacking T and B lymphocytes completely abrogated the development of colonic inflammation, despite the still leaky hTNFα promoter. Furthermore, both the intensity of the immune response and the strength of immunosuppressive Treg induction was found to depend on the major histocompatibility complex (MHC) genetic composition. In summary, the onset of intestinal inflammation in elderly mice depends on at least four factors that have to be present simultaneously: TNFα upregulation, S100A8/A9 protein expression, functional T lymphocytes and genetic composition, with the MHC haplotype being of central importance. Only joint action of these factors leads to chronic intestinal inflammation, while absence of any of these determinants abrogates the development of the autoimmune disorder. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Rafael Leite Dantas
- Institute of Molecular Virology, Westfaelische Wilhelms University, Muenster, Germany
| | - Dominik Bettenworth
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Muenster, Muenster, Germany
| | - Georg Varga
- Pediatric Rheumatology and Immunology, Westfaelische Wilhelms University, Muenster, Germany
| | - Toni Weinhage
- Pediatric Rheumatology and Immunology, Westfaelische Wilhelms University, Muenster, Germany
| | | | - Ulrich Dobrindt
- Institute of Hygiene, Westfaelische Wilhelms University, Muenster, Germany
| | - Johannes Roth
- Institute of Immunology, Westfaelische Wilhelms University, Muenster, Germany
| | - Thomas Vogl
- Institute of Immunology, Westfaelische Wilhelms University, Muenster, Germany
| | - Stephan Ludwig
- Institute of Molecular Virology, Westfaelische Wilhelms University, Muenster, Germany
| | - Viktor Wixler
- Institute of Molecular Virology, Westfaelische Wilhelms University, Muenster, Germany
| |
Collapse
|
52
|
Zhu Y, Yun Y, Jin M, Li G, Li H, Miao P, Ding X, Feng X, Xu L, Sun B. Identification of novel biomarkers for neonatal hypoxic-ischemic encephalopathy using iTRAQ. Ital J Pediatr 2020; 46:67. [PMID: 32448169 PMCID: PMC7245890 DOI: 10.1186/s13052-020-00822-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Background A prompt diagnosis of HIE remains a challenge clinically. This study aimed to identify potential biomarkers of neonatal hypoxic-ischemic encephalopathy (HIE) via a novel proteomic approach, the isobaric tags for absolute and relative quantification (iTRAQ) method. Methods Blood samples were collected from neonates with mild (n = 4), moderate (n = 4), or severe (n = 4) HIE who were admitted to the neonatal intensive care unit of Children’s Hospital of Soochow University between Oct 2015 and Oct 2017. iTRAQ was performed in HIE patients and healthy controls (n = 4). Bioinformatics analyses including Gene Ontology and KEGG pathway enrichment analysis were performed to evaluate the potential features and capabilities of the identified differentially expressed proteins. Results A total of 51 commonly differentially expressed proteins were identified among the comparisons between mild, moderate, and severe HIE as well as healthy controls. Haptoglobin (HP) and S100A8 were most significantly up-regulated in patients with HIE and further validated via real-time PCR and western blotting. The differentially expressed proteins represented multiple biological processes, cellular components and molecular functions and were markedly enriched in complement and coagulation cascades. Conclusions HP and S100A8 may serve as a potential biomarker for neonatal HIE and reflects the severity of HIE. The complement and coagulation cascades play crucial roles in the development of neonatal HIE.
Collapse
Affiliation(s)
- Yuanyuan Zhu
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| | - Yajing Yun
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| | - Meifang Jin
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Gen Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Hong Li
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| | - Po Miao
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| | - Xin Ding
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| | - Xing Feng
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| | - Lixiao Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China.
| | - Bin Sun
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
53
|
Viemann D. S100-Alarmins Are Essential Pilots of Postnatal Innate Immune Adaptation. Front Immunol 2020; 11:688. [PMID: 32425933 PMCID: PMC7203218 DOI: 10.3389/fimmu.2020.00688] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/26/2020] [Indexed: 12/14/2022] Open
Abstract
The restricted capacity of newborn infants to mount inflammatory responses toward microbial challenges has traditionally been linked to the high risk of septic diseases during the neonatal period. In recent years, substantial evidence has been provided that this characteristic of the neonatal immune system is actually a meaningful physiologic state that is based on specific transiently active cellular and molecular mechanisms and required for a favorable course of postnatal immune adaptation. The identification of physiologically high amounts of S100-alarmins in neonates has been one of the crucial pieces in the puzzle that contributed to the change of concept. In this context, innate immune immaturity could be redefined and assigned to the epigenetic silence of adult-like cell-autonomous regulation at the beginning of life. S100-alarmins represent an alternative age-specific mechanism of immune regulation that protects neonates from hyperinflammatory immune responses. Here, we summarize how infants are provided with S100-alarmins and why these allow an uneventful clash between the innate immune system and the extrauterine world. The mode of action of S100-alarmins is highlighted including their tuning functions at multiple levels for establishing a state of homeostasis with the environment in the newborn individual.
Collapse
Affiliation(s)
- Dorothee Viemann
- Department of Pediatric Pneumology, Allergy and Neonatology, Hannover Medical School, Hanover, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hanover, Germany.,PRIMAL Consortium, Hanover, Germany
| |
Collapse
|
54
|
Di Ceglie I, Kruisbergen NNL, van den Bosch MHJ, van Lent PLEM. Fc-gamma receptors and S100A8/A9 cause bone erosion during rheumatoid arthritis. Do they act as partners in crime? Rheumatology (Oxford) 2020; 58:1331-1343. [PMID: 31180451 DOI: 10.1093/rheumatology/kez218] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 04/25/2019] [Indexed: 12/19/2022] Open
Abstract
Bone erosion is one of the central hallmarks of RA and is caused by excessive differentiation and activation of osteoclasts. Presence of autoantibodies in seropositive arthritis is associated with radiographic disease progression. ICs, formed by autoantibodies and their antigens, activate Fcγ-receptor signalling in immune cells, and as such stimulate inflammation-mediated bone erosion. Interestingly, ICs can also directly activate osteoclasts by binding to FcγRs on their surface. Next to autoantibodies, high levels of alarmins, among which is S100A8/A9, are typical for RA and they can further activate the immune system but also directly promote osteoclast function. Therefore, IC-activated FcγRs and S100A8/A9 might act as partners in crime to stimulate inflammation and osteoclasts differentiation and function, thereby stimulating bone erosion. This review discusses the separate roles of ICs, FcγRs and alarmins in bone erosion and sheds new light on the possible interplay between them, which could fuel bone erosion.
Collapse
Affiliation(s)
- Irene Di Ceglie
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nik N L Kruisbergen
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Peter L E M van Lent
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
55
|
Narciclasine improves outcome in sepsis among neonatal rats via inhibition of calprotectin and alleviating inflammatory responses. Sci Rep 2020; 10:2947. [PMID: 32076015 PMCID: PMC7031385 DOI: 10.1038/s41598-020-59716-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 02/03/2020] [Indexed: 12/31/2022] Open
Abstract
Sepsis is associated with exacerbated inflammatory response which subsequently results in multiple organ dysfunction. Sepsis accounts for high mortality and morbidity among newborns worldwide. Narciclasine is a plant alkaloid which has shown to possess anti-inflammatory properties. In this study we investigated the effect and mechanism of action of narciclasine in neonatal sepsis rat models. The excessive release of S100A8/A9 or calprotectin in neonatal sepsis could be detrimental as it could exacerbate the inflammatory responses. We found that narciclasine significantly reduced the plasma levels of S100A8/A9 and also suppressed its expression in the liver and lung. The systemic and local bacterial load was also reduced in the narciclasine treated rats. The systemic and local production of pro-inflammatory cytokines in plasma and organs (liver and lungs) was significantly reduced in the narciclasine treated rats. The histopathological studies showed that narciclasine prevents the organ damage associated with sepsis and improved the survival of neonatal rats. Sepsis increased the phosphorylated NF-κβ p65 protein expression in the liver. Narciclasine suppressed the phosphorylation of NF-κβ p65 and the degradation of NF-κβ inhibitory protein alpha. It could also suppress the expression of adaptor proteins of the toll like receptor signaling pathway viz., myeloid differentiation factor 88 (MyD88), Interleukin-1 receptor-associated kinase 1 (IRAK1) and TNF receptor associated factor 6 (TRAF6). These results suggest that narciclasine protects against sepsis in neonatal rats through the inhibition of calprotectin, pro-inflammatory cytokines and suppression of NF-κβ signaling pathway.
Collapse
|
56
|
Ishizuka K, Fujii W, Azuma N, Mizobuchi H, Morimoto A, Sanjoba C, Matsumoto Y, Goto Y. Pathological roles of MRP14 in anemia and splenomegaly during experimental visceral leishmaniasis. PLoS Negl Trop Dis 2020; 14:e0008020. [PMID: 31961866 PMCID: PMC6994150 DOI: 10.1371/journal.pntd.0008020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 01/31/2020] [Accepted: 01/01/2020] [Indexed: 01/03/2023] Open
Abstract
Myeloid-related protein 14 (MRP14) belongs to the S100 calcium-binding protein family and is expressed in neutrophils and inflammatory macrophages. Increase in the number of MRP14+ cells or serum level of MRP14 is associated with various diseases such as autoimmune diseases and infectious diseases, suggesting the involvement of the molecule in pathogenesis of those diseases. In this study, to examine the pathological involvement of MRP14 during cutaneous and visceral leishmaniasis, wild-type (WT) and MRP14 knockout (MRP14KO) mice were infected with Leishmania major and L. donovani. Increase in the number of MRP14+ cells at the infection sites in wild-type mice was commonly found in the skin during L. major infection as well as the spleen and liver during L. donovani infection. In contrast, the influence of MRP14 to the pathology seemed different between the two infections. MRP14 depletion exacerbated the lesion development and ulcer formation in L. major infection. On the other hand, the depletion improved anemia and splenomegaly but not hepatomegaly at 24 weeks of L. donovani infection. These results suggest that, distinct from its protective role in CL, MRP14 is involved in exacerbation of some symptoms during VL.
Collapse
Affiliation(s)
- Kanna Ishizuka
- Laboratory of Molecular Immunology, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Wataru Fujii
- Laboratory of Applied Genetics, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Natsuho Azuma
- Laboratory of Molecular Immunology, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Haruka Mizobuchi
- Laboratory of Molecular Immunology, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Ayako Morimoto
- Laboratory of Molecular Immunology, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Chizu Sanjoba
- Laboratory of Molecular Immunology, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yoshitsugu Matsumoto
- Laboratory of Molecular Immunology, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yasuyuki Goto
- Laboratory of Molecular Immunology, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
57
|
Sreejit G, Flynn MC, Patil M, Krishnamurthy P, Murphy AJ, Nagareddy PR. S100 family proteins in inflammation and beyond. Adv Clin Chem 2020; 98:173-231. [PMID: 32564786 DOI: 10.1016/bs.acc.2020.02.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The S100 family proteins possess a variety of intracellular and extracellular functions. They interact with multiple receptors and signal transducers to regulate pathways that govern inflammation, cell differentiation, proliferation, energy metabolism, apoptosis, calcium homeostasis, cell cytoskeleton and microbial resistance. S100 proteins are also emerging as novel diagnostic markers for identifying and monitoring various diseases. Strategies aimed at targeting S100-mediated signaling pathways hold a great potential in developing novel therapeutics for multiple diseases. In this chapter, we aim to summarize the current knowledge about the role of S100 family proteins in health and disease with a major focus on their role in inflammatory conditions.
Collapse
Affiliation(s)
| | - Michelle C Flynn
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mallikarjun Patil
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andrew J Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Department of Immunology, Monash University, Melbourne, VIC, Australia
| | | |
Collapse
|
58
|
Zandstra J, van de Geer A, Tanck MWT, van Stijn-Bringas Dimitriades D, Aarts CEM, Dietz SM, van Bruggen R, Schweintzger NA, Zenz W, Emonts M, Zavadska D, Pokorn M, Usuf E, Moll HA, Schlapbach LJ, Carrol ED, Paulus S, Tsolia M, Fink C, Yeung S, Shimizu C, Tremoulet A, Galassini R, Wright VJ, Martinón-Torres F, Herberg J, Burns J, Levin M, Kuijpers TW. Biomarkers for the Discrimination of Acute Kawasaki Disease From Infections in Childhood. Front Pediatr 2020; 8:355. [PMID: 32775314 PMCID: PMC7388698 DOI: 10.3389/fped.2020.00355] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 05/28/2020] [Indexed: 01/08/2023] Open
Abstract
Background: Kawasaki disease (KD) is a vasculitis of early childhood mimicking several infectious diseases. Differentiation between KD and infectious diseases is essential as KD's most important complication-the development of coronary artery aneurysms (CAA)-can be largely avoided by timely treatment with intravenous immunoglobulins (IVIG). Currently, KD diagnosis is only based on clinical criteria. The aim of this study was to evaluate whether routine C-reactive protein (CRP) and additional inflammatory parameters myeloid-related protein 8/14 (MRP8/14 or S100A8/9) and human neutrophil-derived elastase (HNE) could distinguish KD from infectious diseases. Methods and Results: The cross-sectional study included KD patients and children with proven infections as well as febrile controls. Patients were recruited between July 2006 and December 2018 in Europe and USA. MRP8/14, CRP, and HNE were assessed for their discriminatory ability by multiple logistic regression analysis with backward selection and receiver operator characteristic (ROC) curves. In the discovery cohort, the combination of MRP8/14+CRP discriminated KD patients (n = 48) from patients with infection (n = 105), with area under the ROC curve (AUC) of 0.88. The HNE values did not improve discrimination. The first validation cohort confirmed the predictive value of MRP8/14+CRP to discriminate acute KD patients (n = 26) from those with infections (n = 150), with an AUC of 0.78. The second validation cohort of acute KD patients (n = 25) and febrile controls (n = 50) showed an AUC of 0.72, which improved to 0.84 when HNE was included. Conclusion: When used in combination, the plasma markers MRP8/14, CRP, and HNE may assist in the discrimination of KD from both proven and suspected infection.
Collapse
Affiliation(s)
- Judith Zandstra
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Annemarie van de Geer
- Sanquin Research and Landsteiner Laboratory, Department of Blood Cell Research, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Michael W T Tanck
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Diana van Stijn-Bringas Dimitriades
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Cathelijn E M Aarts
- Sanquin Research and Landsteiner Laboratory, Department of Blood Cell Research, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Sanne M Dietz
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Robin van Bruggen
- Sanquin Research and Landsteiner Laboratory, Department of Blood Cell Research, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Nina A Schweintzger
- Department of General Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Werner Zenz
- Department of General Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Marieke Emonts
- Pediatric Infectious Diseases and Immunology Department, Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Dace Zavadska
- Department of Pediatrics, Riga Stradins University, Riga, Latvia
| | - Marko Pokorn
- Department of Infectious Diseases, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Effua Usuf
- Medical Research Council Unit the Gambia (MRCG) at LSHTM, Serrekunda, Gambia
| | - Henriette A Moll
- Department of General Pediatrics, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands
| | - Luregn J Schlapbach
- Pediatric Intensive Care Unit, Lady Cilento Children's Hospital, Pediatric Critical Care Research Group, Brisbane, QLD, Australia
| | - Enitan D Carrol
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool Institute of Infection and Global Health, Liverpool, United Kingdom
| | - Stephane Paulus
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool Institute of Infection and Global Health, Liverpool, United Kingdom
| | - Maria Tsolia
- Second Department of Pediatrics, P. & A. Kyriakou Children's Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Colin Fink
- Micropathology Ltd., University of Warwick, Warwick, United Kingdom
| | - Shunmay Yeung
- Department of Clinical Research, Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, United Kingdom.,Section of Paediatric Infectious Diseases, Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Chisato Shimizu
- Kawasaki Disease Research Center, Rady's Children's Hospital-San Diego, University of California, San Diego, San Diego, CA, United States
| | - Adriana Tremoulet
- Kawasaki Disease Research Center, Rady's Children's Hospital-San Diego, University of California, San Diego, San Diego, CA, United States
| | - Rachel Galassini
- Section of Paediatric Infectious Diseases, Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Victoria J Wright
- Section of Paediatric Infectious Diseases, Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Federico Martinón-Torres
- Translational Pediatrics and Infectious Diseases, Hospital Clínico Universitario de Santiago, University of Santiago, Santiago de Compostela, Spain
| | - Jethro Herberg
- Section of Paediatric Infectious Diseases, Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Jane Burns
- Kawasaki Disease Research Center, Rady's Children's Hospital-San Diego, University of California, San Diego, San Diego, CA, United States
| | - Michael Levin
- Section of Paediatric Infectious Diseases, Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Taco W Kuijpers
- Sanquin Research and Landsteiner Laboratory, Department of Blood Cell Research, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | | |
Collapse
|
59
|
Abstract
Rheumatoid arthritis (RA) is an inflammatory autoimmune disease characterized by inflammatory cell infiltration, high levels of cytokines, and erosion of cartilage and bone in joints. Calprotectin (CLP), as a recently described member of S100 family proteins, is a heterodimeric complex of S100A8 and S100A9. Currently, plenty of studies have indicated significantly increased serum and synovial fluid levels of CLP in patients with RA. It was reported that CLP was related to cell differentiation, migration, apoptosis, and production of pro-inflammatory factors in RA. In addition, there are the positive relationships between serum, synovial CLP and traditional acute phase reactants, disease activity, ultrasound and radiographic progression of joints, and treatment response of RA. In this review, we mainly discuss the role of CLP in the pathogenesis of RA as well as its potential to estimate clinical disease progression of RA patients.
Collapse
|
60
|
Wang C, Kong Y, Ding Y, Sun J, Chen T. Serum Calprotectin Levels and Outcome Following Percutaneous Coronary Intervention in Patients with Diabetes and Acute Coronary Syndrome. Med Sci Monit 2019; 25:9517-9523. [PMID: 31834876 PMCID: PMC6929552 DOI: 10.12659/msm.918126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background A retrospective study of data from a prospective clinical registry was conducted to evaluate the prognostic role of serum calprotectin in patients with diabetes who underwent percutaneous coronary intervention (PCI) for acute coronary syndrome (ACS). Material/Methods Data were retrieved for 273 patients with diabetes mellitus who underwent PCI for primary ACS in a single center. Serum calprotectin levels were measured before PCI. Baseline clinical data included the Global Registry of Acute Coronary Events (GRACE) risk score for ACS. All patients underwent regular follow-up for major adverse cardiovascular events (MACE) during 12 months after PCI, including target vessel revascularization (TVR), defined as the need for an unplanned repeat PCI or coronary artery procedure. The predictive value of serum calprotectin for MACE was analyzed by using univariate and multivariate analysis and receiver operating characteristic (ROC) curve analysis. Results At the final follow-up, 47 of the 273 patients studies experienced MACE. Optimal cutoff values for serum calprotectin levels predictive for MACE stratified patients into a high calprotectin group and a low calprotectin group. The incidence of MACE and TVR in the high calprotectin group was significantly greater than in the low calprotectin group (21.9% vs. 11.5%; P=0.02). Multivariate analysis, adjusted for confounders, showed that the serum level of calprotectin was an independent risk factor for MACE (HR, 1.56; 95% CI, 1.08–4.62; P=0.01). Conclusions In patients with diabetes and the co-morbidity of ACS, a high serum level of calprotectin was associated with a significantly increased risk for MACE following PCI.
Collapse
Affiliation(s)
- Chengji Wang
- Fifth Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China (mainland)
| | - Yu Kong
- Fifth Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China (mainland)
| | - Yuanyuan Ding
- Department of Gastroenterology, Jining No. 1 People's Hospital, Jining, Shandong, China (mainland)
| | - Jingzhi Sun
- Fifth Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China (mainland)
| | - Tao Chen
- Department of Critical Care Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong, China (mainland)
| |
Collapse
|
61
|
Pan S, Hu Y, Hu M, Xu Y, Chen M, Du C, Cui J, Zheng P, Lai J, Zhang Y, Bai J, Jiang P, Zhu J, He Y, Wang J. S100A8 facilitates cholangiocarcinoma metastasis via upregulation of VEGF through TLR4/NF‑κB pathway activation. Int J Oncol 2019; 56:101-112. [PMID: 31746424 PMCID: PMC6910197 DOI: 10.3892/ijo.2019.4907] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 10/03/2019] [Indexed: 12/12/2022] Open
Abstract
A growing body of evidence indicates that S100 calcium-binding protein A8 (S100A8) is frequently overexpressed in malignant tumor tissues and regulates tumor progression; however, the role of S100A8 in cholangiocarcinoma (CCA) remains unclear. The present study demonstrated that the protein expression of S100A8 was significantly higher in pathological tissues compared with adjacent normal tissues from patients with CCA. In addition, S100A8 expression was significantly associated with differentiation, lymph node metastasis and poor prognosis in patients following surgical resection of CCA. Furthermore, both in vitro and in vivo experiments revealed that overexpression of S100A6 promoted, while S100A8 knockdown attenuated, the migration and metastasis of CCA cells. Of note, the present results indicated that S100A8 promoted the CCA tumor cell-induced migration of vascular endothelial cells. Finally, S100A8 was demonstrated to positively regulate the expression of vascular endothelial growth factor (VEGF) in CCA cells, which was mediated by activation of the Toll-like receptor 4 (TLR4)/NF-κB pathway. In conclusion, the present study demonstrated that S100A8 had an important role in facilitating CCA cell migration and metastasis via upregulation of VEGF expression by activating the TLR4/NF-κB pathway. These findings may provide a novel target for CCA treatment.
Collapse
Affiliation(s)
- Shuguang Pan
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Centre for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Ying Hu
- Oncology Department, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Mengjia Hu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Centre for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yang Xu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Centre for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Mo Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Centre for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Changhong Du
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Centre for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Jinchi Cui
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Ping Zheng
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Jiejuan Lai
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yujun Zhang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Jie Bai
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Peng Jiang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Jin Zhu
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yu He
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Junping Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Centre for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
62
|
Wei L, Liu M, Xiong H. Role of Calprotectin as a Biomarker in Periodontal Disease. Mediators Inflamm 2019; 2019:3515026. [PMID: 31530995 PMCID: PMC6721252 DOI: 10.1155/2019/3515026] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/30/2019] [Accepted: 08/10/2019] [Indexed: 12/28/2022] Open
Abstract
Periodontal disease (PD) is a common infectious and inflammatory disease characterised by inflammation of tissues surrounding and supporting the teeth and destruction of the associated alveolar bone, eventually resulting in tooth loss. This disease is caused by periodontopathic bacteria in plaque biofilm and resultant innate and adaptive immune responses in periodontal tissues. Calprotectin (CLP) is a calcium-binding protein of the S-100 protein family and is found to be induced by activated granulocytes, monocytes, and epithelial cells. CLP has been shown to play an important role in numerous inflammatory diseases and disorders. Increasing evidence indicates that CLP is involved in the progression of PD, and its levels may be associated with disease severity and outcome of periodontal treatments. This review will summarise recent studies regarding the presence, regulation, and function of CLP in PD. The findings indicate that CLP may be an effective biomarker for diagnosis and treatment for the PD.
Collapse
Affiliation(s)
- Lili Wei
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Mingwen Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Haofei Xiong
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
63
|
Marinković G, Grauen Larsen H, Yndigegn T, Szabo IA, Mares RG, de Camp L, Weiland M, Tomas L, Goncalves I, Nilsson J, Jovinge S, Schiopu A. Inhibition of pro-inflammatory myeloid cell responses by short-term S100A9 blockade improves cardiac function after myocardial infarction. Eur Heart J 2019; 40:2713-2723. [DOI: 10.1093/eurheartj/ehz461] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 05/20/2019] [Accepted: 07/01/2019] [Indexed: 11/13/2022] Open
Abstract
Abstract
Aims
Neutrophils have both detrimental and beneficial effects in myocardial infarction (MI), but little is known about the underlying pathways. S100A8/A9 is a pro-inflammatory alarmin abundantly expressed in neutrophils that is rapidly released in the myocardium and circulation after myocardial ischaemia. We investigated the role of S100A8/A9 in the innate immune response to MI.
Methods and results
In 524 patients with acute coronary syndrome (ACS), we found that high plasma S100A8/A9 at the time of the acute event was associated with lower left ventricular ejection fraction (EF) at 1-year and increased hospitalization for heart failure (HF) during follow-up. In wild-type C57BL/6 mice with MI induced by permanent coronary artery ligation, treatment with the S100A9 blocker ABR-238901 during the inflammatory phase of the immune response inhibited haematopoietic stem cell proliferation and myeloid cell egression from the bone marrow. The treatment reduced the numbers of neutrophils and monocytes/macrophages in the myocardium, promoted an anti-inflammatory environment, and significantly improved cardiac function compared with MI controls. To mimic the clinical scenario, we further confirmed the effects of the treatment in a mouse model of ischaemia/reperfusion. Compared with untreated mice, 3-day ABR-238901 treatment significantly improved left ventricular EF (48% vs. 35%, P = 0.002) and cardiac output (15.7 vs. 11.1 mL/min, P = 0.002) by Day 21 post-MI.
Conclusion
Short-term S100A9 blockade inhibits inflammation and improves cardiac function in murine models of MI. As an excessive S100A8/A9 release is linked to incident HF, S100A9 blockade might represent a feasible strategy to improve prognosis in ACS patients.
Collapse
Affiliation(s)
- Goran Marinković
- Department of Clinical Sciences Malmö, Lund University, CRC 91:12, Jan Waldenströms gata 35, SE-214 28, Malmö, Sweden
| | - Helena Grauen Larsen
- Department of Cardiology, Skane University Hospital Malmö, Carl-Bertil Laurells gata 9, SE-214 28 Malmö, Sweden
| | - Troels Yndigegn
- Department of Cardiology, Skane University Hospital Malmö, Carl-Bertil Laurells gata 9, SE-214 28 Malmö, Sweden
| | - Istvan Adorjan Szabo
- Department of Pathophysiology, University of Medicine, Pharmacy, Sciences and Technology of Targu-Mures, Gheorghe Marinescu str. 38, 540139 Targu-Mures, Romania
| | - Razvan Gheorghita Mares
- Department of Pathophysiology, University of Medicine, Pharmacy, Sciences and Technology of Targu-Mures, Gheorghe Marinescu str. 38, 540139 Targu-Mures, Romania
| | - Lisa de Camp
- DeVos Cardiovascular Research Program, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
| | - Matthew Weiland
- DeVos Cardiovascular Research Program, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
| | - Lukas Tomas
- Department of Clinical Sciences Malmö, Lund University, CRC 91:12, Jan Waldenströms gata 35, SE-214 28, Malmö, Sweden
| | - Isabel Goncalves
- Department of Clinical Sciences Malmö, Lund University, CRC 91:12, Jan Waldenströms gata 35, SE-214 28, Malmö, Sweden
- Department of Cardiology, Skane University Hospital Malmö, Carl-Bertil Laurells gata 9, SE-214 28 Malmö, Sweden
| | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, CRC 91:12, Jan Waldenströms gata 35, SE-214 28, Malmö, Sweden
| | - Stefan Jovinge
- DeVos Cardiovascular Research Program, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
- DeVos Cardiovascular Research Program, Fredrik Meijer Heart & Vascular Institute, Spectrum Health, 100 Michigan Street NE, Grand Rapids, MI 49503, USA
- Cardiovascular Institute, Stanford Medical School, 265 Campus Drive, Stanford, CA 94305, USA
| | - Alexandru Schiopu
- Department of Clinical Sciences Malmö, Lund University, CRC 91:12, Jan Waldenströms gata 35, SE-214 28, Malmö, Sweden
- Department of Cardiology, Skane University Hospital Malmö, Carl-Bertil Laurells gata 9, SE-214 28 Malmö, Sweden
- Department of Pathophysiology, University of Medicine, Pharmacy, Sciences and Technology of Targu-Mures, Gheorghe Marinescu str. 38, 540139 Targu-Mures, Romania
| |
Collapse
|
64
|
Bickes MS, Pirr S, Heinemann AS, Fehlhaber B, Halle S, Völlger L, Willers M, Richter M, Böhne C, Albrecht M, Langer M, Pfeifer S, Jonigk D, Vieten G, Ure B, Kaisenberg C, Förster R, Köckritz-Blickwede M, Hansen G, Viemann D. Constitutive TNF‐α signaling in neonates is essential for the development of tissue‐resident leukocyte profiles at barrier sites. FASEB J 2019; 33:10633-10647. [DOI: 10.1096/fj.201900796r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Marie Sophie Bickes
- Department of Pediatric Pneumology, Allergology, and NeonatologyHannover Medical SchoolHannoverGermany
| | - Sabine Pirr
- Department of Pediatric Pneumology, Allergology, and NeonatologyHannover Medical SchoolHannoverGermany
- Cluster of Excellence Resolving Infection Susceptibility (RESIST; EXC)Hannover Medical SchoolHannoverGermany
| | - Anna Sophie Heinemann
- Department of Pediatric Pneumology, Allergology, and NeonatologyHannover Medical SchoolHannoverGermany
| | - Beate Fehlhaber
- Department of Pediatric Pneumology, Allergology, and NeonatologyHannover Medical SchoolHannoverGermany
| | - Stephan Halle
- Institute of ImmunologyHannover Medical SchoolHannoverGermany
| | - Lena Völlger
- Department of Pediatric Pneumology, Allergology, and NeonatologyHannover Medical SchoolHannoverGermany
| | - Maike Willers
- Department of Pediatric Pneumology, Allergology, and NeonatologyHannover Medical SchoolHannoverGermany
| | - Manuela Richter
- Department of Pediatric Pneumology, Allergology, and NeonatologyHannover Medical SchoolHannoverGermany
- Children's Hospital Auf der BultHannoverGermany
| | - Carolin Böhne
- Department of Pediatric Pneumology, Allergology, and NeonatologyHannover Medical SchoolHannoverGermany
| | - Melanie Albrecht
- Department of Pediatric Pneumology, Allergology, and NeonatologyHannover Medical SchoolHannoverGermany
| | - Melissa Langer
- Department of Physiological ChemistryResearch Center for Emerging Infections and Zoonoses (RIZ)University of Veterinary Medicine HannoverHannoverGermany
| | - Sandra Pfeifer
- Department of Physiological ChemistryResearch Center for Emerging Infections and Zoonoses (RIZ)University of Veterinary Medicine HannoverHannoverGermany
| | - Danny Jonigk
- Department of PathologyHannover Medical SchoolHannoverGermany
| | - Gertrud Vieten
- Department of Pediatric SurgeryHannover Medical SchoolHannoverGermany
| | - Benno Ure
- Department of Pediatric SurgeryHannover Medical SchoolHannoverGermany
| | | | - Reinhold Förster
- Cluster of Excellence Resolving Infection Susceptibility (RESIST; EXC)Hannover Medical SchoolHannoverGermany
- Institute of ImmunologyHannover Medical SchoolHannoverGermany
| | - Maren Köckritz-Blickwede
- Department of Physiological ChemistryResearch Center for Emerging Infections and Zoonoses (RIZ)University of Veterinary Medicine HannoverHannoverGermany
| | - Gesine Hansen
- Department of Pediatric Pneumology, Allergology, and NeonatologyHannover Medical SchoolHannoverGermany
- Cluster of Excellence Resolving Infection Susceptibility (RESIST; EXC)Hannover Medical SchoolHannoverGermany
| | - Dorothee Viemann
- Department of Pediatric Pneumology, Allergology, and NeonatologyHannover Medical SchoolHannoverGermany
- Cluster of Excellence Resolving Infection Susceptibility (RESIST; EXC)Hannover Medical SchoolHannoverGermany
| |
Collapse
|
65
|
Di Ceglie I, Blom AB, Davar R, Logie C, Martens JHA, Habibi E, Böttcher LM, Roth J, Vogl T, Goodyear CS, van der Kraan PM, van Lent PL, van den Bosch MH. The alarmin S100A9 hampers osteoclast differentiation from human circulating precursors by reducing the expression of RANK. FASEB J 2019; 33:10104-10115. [PMID: 31199668 DOI: 10.1096/fj.201802691rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The alarmin S100A8/A9 is implicated in sterile inflammation-induced bone resorption and has been shown to increase the bone-resorptive capacity of mature osteoclasts. Here, we investigated the effects of S100A9 on osteoclast differentiation from human CD14+ circulating precursors. Hereto, human CD14+ monocytes were isolated and differentiated toward osteoclasts with M-CSF and receptor activator of NF-κB (RANK) ligand (RANKL) in the presence or absence of S100A9. Tartrate-resistant acid phosphatase staining showed that exposure to S100A9 during monocyte-to-osteoclast differentiation strongly decreased the numbers of multinucleated osteoclasts. This was underlined by a decreased resorption of a hydroxyapatite-like coating. The thus differentiated cells showed a high mRNA and protein production of proinflammatory factors after 16 h of exposure. In contrast, at d 4, the cells showed a decreased production of the osteoclast-promoting protein TNF-α. Interestingly, S100A9 exposure during the first 16 h of culture only was sufficient to reduce osteoclastogenesis. Using fluorescently labeled RANKL, we showed that, within this time frame, S100A9 inhibited the M-CSF-mediated induction of RANK. Chromatin immunoprecipitation showed that this was associated with changes in various histone marks at the epigenetic level. This S100A9-induced reduction in RANK was in part recovered by blocking TNF-α but not IL-1. Together, our data show that S100A9 impedes monocyte-to-osteoclast differentiation, probably via a reduction in RANK expression.-Di Ceglie, I., Blom, A. B., Davar, R., Logie, C., Martens, J. H. A., Habibi, E., Böttcher, L.-M., Roth, J., Vogl, T., Goodyear, C. S., van der Kraan, P. M., van Lent, P. L., van den Bosch, M. H. The alarmin S100A9 hampers osteoclast differentiation from human circulating precursors by reducing the expression of RANK.
Collapse
Affiliation(s)
- Irene Di Ceglie
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Arjen B Blom
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Robab Davar
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Colin Logie
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Joost H A Martens
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Ehsan Habibi
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Lisa-Marie Böttcher
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johannes Roth
- Institute of Immunology, University of Münster, Münster, Germany
| | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany
| | - Carl S Goodyear
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Peter M van der Kraan
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter L van Lent
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | |
Collapse
|
66
|
Armaroli G, Verweyen E, Pretzer C, Kessel K, Hirono K, Ichida F, Okabe M, Cabral DA, Foell D, Brown KL, Kessel C. Monocyte-Derived Interleukin-1β As the Driver of S100A12-Induced Sterile Inflammatory Activation of Human Coronary Artery Endothelial Cells: Implications for the Pathogenesis of Kawasaki Disease. Arthritis Rheumatol 2019; 71:792-804. [PMID: 30447136 DOI: 10.1002/art.40784] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 11/13/2018] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Kawasaki disease (KD) is an acute vasculitis of childhood, predominantly affecting the coronary arteries. S100A12, a granulocyte-derived agonist of both the receptor for advanced glycation end products (RAGE) and Toll-like receptor 4 (TLR-4), is strongly up-regulated in KD. This study was undertaken to investigate the potential contributions of S100A12 to the pathogenesis of KD. METHODS Serum samples from patients with KD (n = 30) at different stages pre- and post-intravenous immunoglobulin (IVIG) treatment were analyzed for the expression of S100A12, cytokines, chemokines, and soluble markers of endothelial cell activation. Primary human coronary artery endothelial cells (HCAECs) were analyzed for responsiveness to direct stimulation with S100A12 or lipopolysaccharide (LPS), as assessed by real-time quantitative reverse transcription-polymerase chain reaction analysis of cytokine and endothelial cell adhesion molecule messenger RNA expression. Alternatively, HCAECs were cultured in conditioned medium obtained from primary human monocytes that were stimulated with LPS or S100A12 in the absence or presence of IVIG or cytokine antagonists. RESULTS In the serum of patients with KD, pretreatment S100A12 levels were associated with soluble vascular cell adhesion molecule 1 titers in the course of IVIG therapy (rs = -0.6, P = 0.0003). Yet, HCAECs were not responsive to direct S100A12 stimulation, despite the presence of appropriate receptors (RAGE, TLR-4). HCAECs did, however, respond to supernatants obtained from S100A12-stimulated primary human monocytes, as evidenced by the gene expression of inflammatory cytokines and adhesion molecules. This response was strictly dependent on interleukin-1β (IL-1β) signaling (P < 0.001). CONCLUSION In its role as a highly expressed mediator of sterile inflammation in KD, S100A12 appears to activate HCAECs in an IL-1β-dependent manner. These data provide new mechanistic insights into the contributions of S100A12 and IL-1β to disease pathogenesis, and may therefore support current IL-1-targeting studies in the treatment of patients with KD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mako Okabe
- University of Toyama, Toyama City, Japan
| | - David A Cabral
- University of British Columbia, British Columbia Children's Hospital, Vancouver, British Columbia, Canada
| | - Dirk Foell
- University Children's Hospital, Munster, Germany
| | - Kelly L Brown
- University of British Columbia, British Columbia Children's Hospital, Vancouver, British Columbia, Canada
| | | |
Collapse
|
67
|
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
|
68
|
Zheng X, Huo X, Zhang Y, Wang Q, Zhang Y, Xu X. Cardiovascular endothelial inflammation by chronic coexposure to lead (Pb) and polycyclic aromatic hydrocarbons from preschool children in an e-waste recycling area. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 246:587-596. [PMID: 30597391 DOI: 10.1016/j.envpol.2018.12.055] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 11/22/2018] [Accepted: 12/17/2018] [Indexed: 02/05/2023]
Abstract
Lead (Pb) and polycyclic aromatic hydrocarbon (PAH) exposure is positively associated with cardiovascular disease (CVD), and the possible potential mechanism may be caused by damage to the endothelium by modulation of inflammatory processes. No comprehensive research shows co-exposure of Pb and PAH on cardiovascular endothelial inflammation in electronic waste (e-waste) exposed populations. Given this, the aim of this study is to provide evidence for a relationship between Pb and PAH co-exposure and cardiovascular endothelial inflammation, in an e-waste-exposed population, to delineate the link between a potential mechanism for CVD and environmental exposure. We recruited 203 preschool children (3-7 years) were enrolled from Guiyu (e-waste-exposed group, n = 105) and Haojiang (reference group, n = 98). Blood Pb levels and urinary PAH metabolites were measured. Percentages of T cells, CD4+ T cells and CD8+ T cells, complete blood counts, endothelial inflammation biomarker (serum S100A8/A9), and other inflammatory biomarkers [serum interleukin (IL)-6, IL-12p70, gamma interferon-inducible protein 10 (IP-10)] levels were evaluated. Blood Pb, total urinary hydroxylated PAH (ΣOHPAH), total hydroxynaphthalene (ΣOHNap) and total hydroxyfluorene (ΣOHFlu) levels, S100A8/A9, IL-6, IL-12p70 and IP-10 concentrations, absolute counts of monocytes, neutrophils, and leukocytes, as well as CD4+ T cell percentages were significantly higher in exposed children. Elevated blood Pb, urinary 2-hydroxynaphthalene (2-OHNap) and ΣOHFlu levels were associated with higher levels of IL-6, IL-12p70, IP-10, CD4+ T cell percentages, neutrophil and monocyte counts. Mediator models indicated that neutrophils exert the significant mediation effect on the relationship between blood Pb levels and S100A8/A9. IL-6 exerts a significant mediation effect on the relationship between blood Pb levels and IP-10, as well as the relationship between urinary ΣOHFlu levels and IP-10. Our results indicate that children with elevated exposure levels of Pb and PAHs have exacerbated vascular endothelial inflammation, which may indicate future CVD risk in e-waste recycling areas.
Collapse
Affiliation(s)
- Xiangbin Zheng
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Yu Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, 9713, GZ, the Netherlands
| | - Qihua Wang
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Yuling Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China; Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, 515041, Guangdong, China.
| |
Collapse
|
69
|
Holzinger D, Tenbrock K, Roth J. Alarmins of the S100-Family in Juvenile Autoimmune and Auto-Inflammatory Diseases. Front Immunol 2019; 10:182. [PMID: 30828327 PMCID: PMC6384255 DOI: 10.3389/fimmu.2019.00182] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 01/21/2019] [Indexed: 12/17/2022] Open
Abstract
Autoimmune and auto-inflammatory diseases in children are causing chronic inflammation, organ damage, and pain. Although several options for treatment are nowadays available a significant number of patients does not respond sufficiently to current therapies. In these diseases inflammatory processes are triggered by numerous exogenous and endogenous factors. There is now increasing evidence that especially a novel family of pro-inflammatory molecules, named alarmins, play a significant role in inflammatory processes underlying these diseases. Alarmins are endogenous proteins released during stress reactions that confer inflammatory signaling via Pattern Recognition Receptors (PRRs), like the Toll-like receptor 4 (TLR4). The most abundant alarmins in juvenile rheumatic diseases belong to the family of pro-inflammatory calcium-binding S100-proteins. In this review we will give a general introduction in S100-biology. We will demonstrate the functional relevance of these proteins in animal models of autoimmune and auto-inflammatory diseases. We will show the expression patterns of S100-alarmins and correlation to disease activity in different forms of juvenile idiopathic arthritis, auto-inflammatory diseases, and systemic autoimmune disorders. Finally, we will discuss the clinical use of S100-alarmins as biomarkers for diagnosis and monitoring of rheumatic diseases in children and will point out potential future therapeutic approaches targeting inflammatory effects mediated by S100-alarmins.
Collapse
Affiliation(s)
- Dirk Holzinger
- Department of Pediatric Hematology-Oncology, University of Duisburg-Essen, Essen, Germany
| | - Klaus Tenbrock
- Department of Pediatrics, Medical Faculty, RWTH Aachen, Aachen, Germany
| | - Johannes Roth
- Institute of Immunology, University of Muenster, Münster, Germany
| |
Collapse
|
70
|
Crowe LAN, McLean M, Kitson SM, Melchor EG, Patommel K, Cao HM, Reilly JH, Leach WJ, Rooney BP, Spencer SJ, Mullen M, Chambers M, Murrell GAC, McInnes IB, Akbar M, Millar NL. S100A8 & S100A9: Alarmin mediated inflammation in tendinopathy. Sci Rep 2019; 9:1463. [PMID: 30728384 PMCID: PMC6365574 DOI: 10.1038/s41598-018-37684-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 12/12/2018] [Indexed: 11/11/2022] Open
Abstract
Alarmins S100A8 and S100A9 are endogenous molecules released in response to environmental triggers and cellular damage. They are constitutively expressed in immune cells such as monocytes and neutrophils and their expression is upregulated under inflammatory conditions. The molecular mechanisms that regulate inflammatory pathways in tendinopathy are largely unknown therefore identifying early immune effectors is essential to understanding the pathology. Based on our previous investigations highlighting tendinopathy as an alarmin mediated pathology we sought evidence of S100A8 & A9 expression in a human model of tendinopathy and thereafter, to explore mechanisms whereby S100 proteins may regulate release of inflammatory mediators and matrix synthesis in human tenocytes. Immunohistochemistry and quantitative RT-PCR showed S100A8 & A9 expression was significantly upregulated in tendinopathic tissue compared with control. Furthermore, treating primary human tenocytes with exogenous S100A8 & A9 significantly increased protein release of IL-6, IL-8, CCL2, CCL20 and CXCL10; however, no alterations in genes associated with matrix remodelling were observed at a transcript level. We propose S100A8 & A9 participate in early pathology by modulating the stromal microenvironment and influencing the inflammatory profile observed in tendinopathy. S100A8 and S100A9 may participate in a positive feedback mechanism involving enhanced leukocyte recruitment and release of pro-inflammatory cytokines from tenocytes that perpetuates the inflammatory response within the tendon in the early stages of disease.
Collapse
Affiliation(s)
- Lindsay A N Crowe
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences University of Glasgow, Glasgow, Scotland, UK
| | - Michael McLean
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences University of Glasgow, Glasgow, Scotland, UK
| | - Susan M Kitson
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences University of Glasgow, Glasgow, Scotland, UK
| | - Emma Garcia Melchor
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences University of Glasgow, Glasgow, Scotland, UK
| | - Katharina Patommel
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences University of Glasgow, Glasgow, Scotland, UK
| | - Hai Man Cao
- Orthopaedic Research Institute, Department of Orthopaedic Surgery, St George Hospital Campus, University of New South Wales, New South Wales, Australia
| | - James H Reilly
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences University of Glasgow, Glasgow, Scotland, UK
| | - William J Leach
- Department of Orthopaedic Surgery, Queen Elizabeth University Hospital Glasgow, Glasgow, Scotland, UK
| | - Brain P Rooney
- Department of Orthopaedic Surgery, Queen Elizabeth University Hospital Glasgow, Glasgow, Scotland, UK
| | - Simon J Spencer
- Department of Orthopaedic Surgery, Queen Elizabeth University Hospital Glasgow, Glasgow, Scotland, UK
| | - Michael Mullen
- Department of Orthopaedic Surgery, Queen Elizabeth University Hospital Glasgow, Glasgow, Scotland, UK
| | - Max Chambers
- Department of Orthopaedic Surgery, Queen Elizabeth University Hospital Glasgow, Glasgow, Scotland, UK
| | - George A C Murrell
- Orthopaedic Research Institute, Department of Orthopaedic Surgery, St George Hospital Campus, University of New South Wales, New South Wales, Australia
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences University of Glasgow, Glasgow, Scotland, UK
| | - Moeed Akbar
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences University of Glasgow, Glasgow, Scotland, UK
| | - Neal L Millar
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences University of Glasgow, Glasgow, Scotland, UK.
| |
Collapse
|
71
|
Serum calprotectin and ischemia modified albumin levels as markers of disease activity in Behçet's disease. Postepy Dermatol Alergol 2019; 35:609-613. [PMID: 30618530 PMCID: PMC6320477 DOI: 10.5114/pdia.2017.71269] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 10/16/2017] [Indexed: 02/02/2023] Open
Abstract
Introduction Behçet’s disease (BD) is a complex multisystemic inflammatory disorder which is characterized by recurrent attacks of acute inflammation. As there is no universally recognized pathognomonic laboratory marker of BD, its diagnosis is still based on clinical findings. Aim To evaluate the role of calprotectin and ischemia modified albumin (IMA) as biomarkers in the assessment of disease activity of BD. Material and methods A total of 93 patients with BD and 62 age- and gender-matched healthy controls were included in the study. Disease activity was assessed with the BD Current Activity Form (BDCAF) score. Serum levels of calprotectin, high-sensitivity C-reactive protein (hsCRP) and IMA were measured in the patient and control groups. Results Serum levels of calprotectin, IMA and hsCRP in patients with BD were higher than those of the healthy control group (p < 0.001 for all). No correlations between calprotectin and IMA, hsCRP, erythrocyte sedimentation rate, CRP, or BDCAF score were found. Conclusions As the calprotectin level are increased in BD patients, it could be a candidate biomarker which plays a role in BD pathogenesis.
Collapse
|
72
|
Krombach J, Hennel R, Brix N, Orth M, Schoetz U, Ernst A, Schuster J, Zuchtriegel G, Reichel CA, Bierschenk S, Sperandio M, Vogl T, Unkel S, Belka C, Lauber K. Priming anti-tumor immunity by radiotherapy: Dying tumor cell-derived DAMPs trigger endothelial cell activation and recruitment of myeloid cells. Oncoimmunology 2018; 8:e1523097. [PMID: 30546963 PMCID: PMC6287777 DOI: 10.1080/2162402x.2018.1523097] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/04/2018] [Accepted: 09/08/2018] [Indexed: 12/30/2022] Open
Abstract
The major goal of radiotherapy is the induction of tumor cell death. Additionally, radiotherapy can function as in situ cancer vaccination by exposing tumor antigens and providing adjuvants for anti-tumor immune priming. In this regard, the mode of tumor cell death and the repertoire of released damage-associated molecular patterns (DAMPs) are crucial. However, optimal dosing and fractionation of radiotherapy remain controversial. Here, we examined the initial steps of anti-tumor immune priming by different radiation regimens (20 Gy, 4 × 2 Gy, 2 Gy, 0 Gy) with cell lines of triple-negative breast cancer in vitro and in vivo. Previously, we have shown that especially high single doses (20 Gy) induce a delayed type of primary necrosis with characteristics of mitotic catastrophe and plasma membrane disintegration. Now, we provide evidence that protein DAMPs released by these dying cells stimulate sequential recruitment of neutrophils and monocytes in vivo. Key players in this regard appear to be endothelial cells revealing a distinct state of activation upon exposure to supernatants of irradiated tumor cells as characterized by high surface expression of adhesion molecules and production of a discrete cytokine/chemokine pattern. Furthermore, irradiated tumor cell-derived protein DAMPs enforced differentiation and maturation of dendritic cells as hallmarked by upregulation of co-stimulatory molecules and improved T cell-priming. Consistently, a recurring pattern was observed: The strongest effects were detected with 20 Gy-irradiated cells. Obviously, the initial steps of radiotherapy-induced anti-tumor immune priming are preferentially triggered by high single doses – at least in models of triple-negative breast cancer.
Collapse
Affiliation(s)
- Julia Krombach
- Department of Radiation Oncology, University Hospital, Munich, Germany
| | - Roman Hennel
- Department of Radiation Oncology, University Hospital, Munich, Germany
| | - Nikko Brix
- Department of Radiation Oncology, University Hospital, Munich, Germany
| | - Michael Orth
- Department of Radiation Oncology, University Hospital, Munich, Germany.,German Cancer Consortium (DKTK), Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrike Schoetz
- Department of Radiation Oncology, University Hospital, Munich, Germany.,Department of Radiotherapy and Radiooncology, Philipps-University Marburg, University Hospital Gießen and Marburg, Marburg, Germany
| | - Anne Ernst
- Department of Radiation Oncology, University Hospital, Munich, Germany.,Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Jessica Schuster
- Department of Radiation Oncology, University Hospital, Munich, Germany
| | - Gabriele Zuchtriegel
- Department of Otorhinolaryngology, University Hospital, Munich, Germany.,Walter Brendel Centre of Experimental Medicine, University Hospital, Munich, Germany.,Translational research in haematology/oncology, Institute of Experimental Infectious Diseases and Cancer Research, Division of the University Children's Hospital of Zurich, Zurich, Switzerland
| | - Christoph A Reichel
- Department of Otorhinolaryngology, University Hospital, Munich, Germany.,Walter Brendel Centre of Experimental Medicine, University Hospital, Munich, Germany
| | - Susanne Bierschenk
- Walter Brendel Centre of Experimental Medicine, University Hospital, Munich, Germany
| | - Markus Sperandio
- Walter Brendel Centre of Experimental Medicine, University Hospital, Munich, Germany
| | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany
| | - Steffen Unkel
- Department of Medical Statistics, University Medical Center, Goettingen, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, Munich, Germany.,German Cancer Consortium (DKTK), Munich, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, Munich, Germany.,German Cancer Consortium (DKTK), Munich, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| |
Collapse
|
73
|
Tammaro A, Florquin S, Brok M, Claessen N, Butter LM, Teske GJD, de Boer OJ, Vogl T, Leemans JC, Dessing MC. S100A8/A9 promotes parenchymal damage and renal fibrosis in obstructive nephropathy. Clin Exp Immunol 2018; 193:361-375. [PMID: 29746703 PMCID: PMC6150262 DOI: 10.1111/cei.13154] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2018] [Indexed: 12/11/2022] Open
Abstract
Despite advances in our understanding of the mechanisms underlying the progression of chronic kidney disease and the development of fibrosis, only limited efficacious therapies exist. The calcium binding protein S100A8/A9 is a damage-associated molecular pattern which can activate Toll-like receptor (TLR)-4 or receptor for advanced glycation end-products (RAGE). Activation of these receptors is involved in the progression of renal fibrosis; however, the role of S100A8/A9 herein remains unknown. Therefore, we analysed S100A8/A9 expression in patients and mice with obstructive nephropathy and subjected wild-type and S100A9 knock-out mice lacking the heterodimer S100A8/A9 to unilateral ureteral obstruction (UUO). We found profound S100A8/A9 expression in granulocytes that infiltrated human and murine kidney, together with enhanced renal expression over time, following UUO. S100A9 KO mice were protected from UUO-induced renal fibrosis, independently of leucocyte infiltration and inflammation. Loss of S100A8/A9 protected tubular epithelial cells from UUO-induced apoptosis and critical epithelial-mesenchymal transition steps. In-vitro studies revealed S100A8/A9 as a novel mediator of epithelial cell injury through loss of cell polarity, cell cycle arrest and subsequent cell death. In conclusion, we demonstrate that S100A8/A9 mediates renal damage and fibrosis, presumably through loss of tubular epithelial cell contacts and irreversible damage. Suppression of S100A8/A9 could be a therapeutic strategy to halt renal fibrosis in patients with chronic kidney disease.
Collapse
Affiliation(s)
- A. Tammaro
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - S. Florquin
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - M. Brok
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - N. Claessen
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - L. M. Butter
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - G. J. D. Teske
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - O. J. de Boer
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - T. Vogl
- Institute of ImmunologyUniversity of MünsterMünsterGermany
| | - J. C. Leemans
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - M. C. Dessing
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| |
Collapse
|
74
|
Calprotectin (S100A8/S100A9): a key protein between inflammation and cancer. Inflamm Res 2018; 67:801-812. [PMID: 30083975 DOI: 10.1007/s00011-018-1173-4] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/19/2018] [Accepted: 07/25/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Calprotectin (S100A8/S100A9), a heterodimeric EF-hand Ca2+ binding protein, are abundant in cytosol of neutrophils and are involved in inflammatory processes and several cancerous pathogens. OBJECTIVE The purpose of the present systematic review is to evaluate the pro- and anti-tumorigenic functions of calprotectin and its relation to inflammation. MATERIALS AND METHODS We conducted a review of studies published in the Medline (1966-2018), Scopus (2004-2018), ClinicalTrials.gov (2008-2018) and Google Scholar (2004-2018) databases, combined with studies found in the reference lists of the included studies. RESULTS Elevated levels of S100A8/S100A9 were detected in inflammation, neoplastic tumor cells and various human cancers. Recent data have explained that many cancers arise from sites of infection, chronic irritation, and inflammation. The inflammatory microenvironment which largely includes calprotectin, has an essential role on high producing of inflammatory factors and then on neoplastic process and metastasis. CONCLUSION Scientists have shown different outcomes in inflammation, malignancy and apoptosis whether the source of the aforementioned protein is extracellular or intracellular. These findings are offering new insights that anti-inflammatory therapeutic agents and anti-tumorigenic functions of calprotectin can lead to control cancer development.
Collapse
|
75
|
Kunutsor SK, Flores-Guerrero JL, Kieneker LM, Nilsen T, Hidden C, Sundrehagen E, Seidu S, Dullaart RPF, Bakker SJL. Plasma calprotectin and risk of cardiovascular disease: Findings from the PREVEND prospective cohort study. Atherosclerosis 2018; 275:205-213. [PMID: 29957458 DOI: 10.1016/j.atherosclerosis.2018.06.817] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/07/2018] [Accepted: 06/13/2018] [Indexed: 10/14/2022]
Abstract
BACKGROUND AND AIMS We aimed to assess the association of circulating calprotectin, an inflammation-associated protein, with cardiovascular disease (CVD) risk and determine whether it improves risk prediction. METHODS Plasma calprotectin measurements were made at baseline in 5290 participants in the PREVEND prospective study. Hazard ratios (95% confidence intervals [CI]) for CVD were calculated. RESULTS After a median follow-up of 8.3 years, 339 first CVD events were recorded. Calprotectin concentration was correlated with several conventional risk factors as well as with high-sensitivity C-reactive protein (hsCRP) (r = 0.42). Calprotectin was log-linearly associated with CVD risk. The risk for CVD adjusted for conventional cardiovascular risk factors was 1.26 (95% CI, 1.13-1.41) per 1 standard deviation higher baseline loge calprotectin, and was 1.24 (95% CI, 1.11-1.39) following further adjustment for triglycerides, body mass index, and other potential confounders. The association remained present after further adjustment for hsCRP 1.15 (95% CI, 1.02-1.30). Comparing extreme quartiles of plasma calprotectin levels, the corresponding adjusted HRs for CVD were 1.96 (1.37-2.82), 1.89 (1.31-2.72), and 1.56 (1.07-2.29). The association of calprotectin with CVD risk did not vary importantly in several relevant clinical subgroups. Adding calprotectin to the Framingham CVD Risk Score was associated with a C-index change (0.0016; p=0.42) difference in -2 log likelihood (p=0.038), IDI (0.0080; p < 0.001), and NRI (4.03%; p=0.024). CONCLUSIONS There is a log-linear association of calprotectin concentration with risk of CVD, which may be partly dependent on hsCRP. Adding calprotectin to conventional risk factors improves CVD risk assessment using measures of reclassification and -2 log likelihood.
Collapse
Affiliation(s)
- Setor K Kunutsor
- National Institute for Health Research Bristol Biomedical Research Centre, University Hospitals Bristol NHS Foundation Trust and University of Bristol, Bristol, UK; Translational Health Sciences, Bristol Medical School, Musculoskeletal Research Unit, University of Bristol, Learning & Research Building (Level 1), Southmead Hospital, Bristol, BS10 5NB, UK.
| | - Jose Luis Flores-Guerrero
- Department of Internal Medicine, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Lyanne M Kieneker
- Department of Internal Medicine, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | | | | | | | - Samuel Seidu
- Leicester Diabetes Centre, Leicester General Hospital, Gwendolen Road, Leicester, LE5 4WP, UK; Diabetes Research Centre, University of Leicester, Leicester General Hospital, Gwendolen Road, Leicester, LE5 4WP, UK
| | - Robin P F Dullaart
- Department of Internal Medicine, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
76
|
Wang S, Song R, Wang Z, Jing Z, Wang S, Ma J. S100A8/A9 in Inflammation. Front Immunol 2018; 9:1298. [PMID: 29942307 PMCID: PMC6004386 DOI: 10.3389/fimmu.2018.01298] [Citation(s) in RCA: 862] [Impact Index Per Article: 123.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/24/2018] [Indexed: 12/11/2022] Open
Abstract
S100A8 and S100A9 (also known as MRP8 and MRP14, respectively) are Ca2+ binding proteins belonging to the S100 family. They often exist in the form of heterodimer, while homodimer exists very little because of the stability. S100A8/A9 is constitutively expressed in neutrophils and monocytes as a Ca2+ sensor, participating in cytoskeleton rearrangement and arachidonic acid metabolism. During inflammation, S100A8/A9 is released actively and exerts a critical role in modulating the inflammatory response by stimulating leukocyte recruitment and inducing cytokine secretion. S100A8/A9 serves as a candidate biomarker for diagnosis and follow-up as well as a predictive indicator of therapeutic responses to inflammation-associated diseases. As blockade of S100A8/A9 activity using small-molecule inhibitors or antibodies improves pathological conditions in murine models, the heterodimer has potential as a therapeutic target. In this review, we provide a comprehensive and detailed overview of the distribution and biological functions of S100A8/A9 and highlight its application as a diagnostic and therapeutic target in inflammation-associated diseases.
Collapse
Affiliation(s)
- Siwen Wang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Xiangya School of Medicine, Cancer Research Institute, Central South University, Changsha, China
| | - Rui Song
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Xiangya School of Medicine, Cancer Research Institute, Central South University, Changsha, China
| | - Ziyi Wang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Xiangya School of Medicine, Cancer Research Institute, Central South University, Changsha, China
| | - Zhaocheng Jing
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Xiangya School of Medicine, Cancer Research Institute, Central South University, Changsha, China
| | - Shaoxiong Wang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Xiangya School of Medicine, Cancer Research Institute, Central South University, Changsha, China
| | - Jian Ma
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Xiangya School of Medicine, Cancer Research Institute, Central South University, Changsha, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Key Laboratory of Carcinogenesis of Ministry of Health, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, China
| |
Collapse
|
77
|
Luo X, Zheng T, Mao C, Dong X, Mou X, Xu C, Lu Q, Liu B, Wang S, Xiao Y. Aberrant MRP14 expression in thyroid follicular cells mediates chemokine secretion through the IL-1β/MAPK pathway in Hashimoto's thyroiditis. Endocr Connect 2018; 7:850-858. [PMID: 29764904 PMCID: PMC6000753 DOI: 10.1530/ec-18-0019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 05/15/2018] [Indexed: 01/10/2023]
Abstract
Myeloid-related protein 14 (MRP14) is responsible for inflammatory reactions. However, the correlation between MRP14 and Hashimoto's thyroiditis (HT) is still not clear. In this study, we examined the status of MRP14 in thyroid tissues and sera of HT patients and explored the mechanism of IL-1β-mediated regulation of MRP14 expression, as well as the effects of MRP14 on pro-inflammatory chemokine secretion in thyroid follicular cells (TFCs), to elucidate the role of MRP14 in HT development. Our results showed dramatically increased MRP14 expression in thyroid tissues and sera from HT patients. In addition, IL-1β significantly promoted the expression of MRP14 in TFCs, which was mediated by activation of the MAPK/NF-κB signalling pathway. More importantly, IL-1β induced the secretion of the chemokines GRO-2, CXCL9 and CCL22, which was dependent on the regulation of MRP14 in TFCs. Therefore, these findings suggested that under pro-inflammatory conditions, TFCs secreted chemokines with the help of MRP14 regulation, which might suggest a potential pathological mechanism of lymphocyte infiltration into the thyroid gland in HT.
Collapse
Affiliation(s)
- Xuan Luo
- Department of Nuclear MedicineThe Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Tingting Zheng
- Department of Nuclear MedicineThe Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Chaoming Mao
- Department of Nuclear MedicineThe Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of OncologyThe Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xin Dong
- Department of Nuclear MedicineThe Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiao Mou
- Department of Nuclear MedicineThe Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Chengcheng Xu
- Department of Nuclear MedicineThe Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Qingyan Lu
- Department of Nuclear MedicineThe Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Baocui Liu
- Department of Nuclear MedicineThe Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Shengjun Wang
- Department of Laboratory ImmunologyJiangsu University School of Medicine, Zhenjiang, China
| | - Yichuan Xiao
- Key Laboratory of Stem Cell BiologyInstitute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
78
|
β-Glucans in food modify colonic microflora by inducing antimicrobial protein, calprotectin, in a Dectin-1-induced-IL-17F-dependent manner. Mucosal Immunol 2018; 11:763-773. [PMID: 29068000 DOI: 10.1038/mi.2017.86] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 08/22/2017] [Indexed: 02/04/2023]
Abstract
Dectin-1 (gene symbol: Clec7a) is a receptor for β-glucans that play an important role for the host defense against fungi. Recently, we showed that Clec7a-/- mice are resistant against dextran sodium sulfate (DSS)-induced colitis because of regulatory T-cell population expansion in the colon. The regulatory T-cell expansion is caused by expansion of commensal Lactobacillus murinus whose growth is suppressed by an antimicrobial protein, calprotectin S100A8/A9. In this report, we showed that S100A8 was mainly produced by mouse colonic epithelial cells. S100A8 was not induced directly by Dectin-1 but by Dectin-1-induced cytokines, especially interleukin-17F (IL-17F), that were produced by several types of innate immune cells including CD11c+/CD11b+ myeloid cells in colonic lamina propria. S100A8/A9 heterodimer preferentially suppressed the growth of L. murinus that was increased in both Clec7a-/- and Il17f-/- mice. Furthermore, similar expansion of L. murinus and DSS-colitis resistance were observed in mice fed with β-glucan-free food. These observations suggest that food-derived β-glucans control the specific commensal microbiota via the Dectin-1-IL-17F-calprotectin axis to maintain the intestinal homeostasis.
Collapse
|
79
|
Preianò M, Maggisano G, Murfuni MS, Villella C, Pelaia C, Montalcini T, Lombardo N, Pelaia G, Savino R, Terracciano R. An Analytical Method for Assessing Optimal Storage Conditions of Gingival Crevicular Fluid and Disclosing a Peptide Biomarker Signature of Gingivitis by MALDI-TOF MS. Proteomics Clin Appl 2018; 12:e1800005. [DOI: 10.1002/prca.201800005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/27/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Mariaimmacolata Preianò
- Department of Health Sciences; Laboratory of Mass Spectrometry and Proteomics; University “Magna Graecia”; Catanzaro 88100 Italy
| | - Giuseppina Maggisano
- Department of Health Sciences; Laboratory of Mass Spectrometry and Proteomics; University “Magna Graecia”; Catanzaro 88100 Italy
| | - Maria Stella Murfuni
- Department of Health Sciences; Laboratory of Mass Spectrometry and Proteomics; University “Magna Graecia”; Catanzaro 88100 Italy
| | - Chiara Villella
- Department of Health Sciences; Laboratory of Mass Spectrometry and Proteomics; University “Magna Graecia”; Catanzaro 88100 Italy
| | - Corrado Pelaia
- Department of Medical and Surgical Sciences; University “Magna Graecia”; Catanzaro 88100 Italy
| | - Tiziana Montalcini
- Department of Experimental and Clinical Medicine; University “Magna Graecia”; Catanzaro 88100 Italy
| | - Nicola Lombardo
- Department of Medical and Surgical Sciences; University “Magna Graecia”; Catanzaro 88100 Italy
| | - Girolamo Pelaia
- Department of Medical and Surgical Sciences; University “Magna Graecia”; Catanzaro 88100 Italy
| | - Rocco Savino
- Department of Health Sciences; Laboratory of Mass Spectrometry and Proteomics; University “Magna Graecia”; Catanzaro 88100 Italy
| | - Rosa Terracciano
- Department of Health Sciences; Laboratory of Mass Spectrometry and Proteomics; University “Magna Graecia”; Catanzaro 88100 Italy
| |
Collapse
|
80
|
Bauer R, Udonta F, Wroblewski M, Ben-Batalla I, Santos IM, Taverna F, Kuhlencord M, Gensch V, Päsler S, Vinckier S, Brandner JM, Pantel K, Bokemeyer C, Vogl T, Roth J, Carmeliet P, Loges S. Blockade of Myeloid-Derived Suppressor Cell Expansion with All-Trans Retinoic Acid Increases the Efficacy of Antiangiogenic Therapy. Cancer Res 2018; 78:3220-3232. [DOI: 10.1158/0008-5472.can-17-3415] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 03/13/2018] [Accepted: 04/17/2018] [Indexed: 11/16/2022]
|
81
|
Nielsen UB, Bruhn LV, Ellingsen T, Stengaard-Pedersen K, Hornung N. Calprotectin in patients with chronic rheumatoid arthritis correlates with disease activity and responsiveness to methotrexate. Scandinavian Journal of Clinical and Laboratory Investigation 2017; 78:62-67. [PMID: 29228799 DOI: 10.1080/00365513.2017.1413591] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Calprotectin (myeloid-related protein 8/14) is elevated in inflammatory diseases and a correlation of serum calprotectin and disease activity in rheumatoid arthritis (RA) has been shown. In this study, we investigated plasma calprotectin as a disease marker in patients with chronic RA treated with methotrexate (MTX) monotherapy and compared plasma calprotectin with C-reactive protein (CRP) in this matter. METHODS Seventy-six patients with chronic RA were included in this open prospective study and of these 40 were included prior to initiation of MTX therapy. The patients were followed with laboratory and clinical parameters for 52-56 weeks. Plasma calprotectin was analyzed at the start of study and at various intervals. Radiographic evaluation was performed at baseline and after 17.2 months and progression in joint destruction was measured with Larsen score. The response to MTX was evaluated according to the American College of Rheumatology criteria. RESULTS Patients starting MTX treatment had significantly higher levels of plasma calprotectin compared to patients well established on MTX therapy (p = .008). Among the 40 patients naive to MTX, 25 responded to MTX therapy and serum calprotectin decreased significantly in these patients (p = .0007). The radiographic damage showed no relation to calprotectin. CONCLUSIONS Plasma calprotectin is associated with disease activity in patients with chronic RA and is more strongly correlated to MTX response compared to CRP. The role of calprotectin as a disease marker is promising and the advantages compared to CRP needs to be further investigated.
Collapse
Affiliation(s)
| | | | - Torkell Ellingsen
- c Department of Reumatology , Odense University Hospital , Odense C , Denmark
| | | | - Nete Hornung
- a Department of Clinical Biochemistry , Randers Regional Hospital , Randers NØ , Denmark
| |
Collapse
|
82
|
Study of serial serum myeloid-related protein 8/14 as a sensitive biomarker in Takayasu arteritis: a single centre study. Rheumatol Int 2017; 38:623-630. [DOI: 10.1007/s00296-017-3881-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 11/08/2017] [Indexed: 11/26/2022]
|
83
|
Brook B, Harbeson D, Ben-Othman R, Viemann D, Kollmann TR. Newborn susceptibility to infection vs. disease depends on complex in vivo interactions of host and pathogen. Semin Immunopathol 2017; 39:615-625. [PMID: 29098373 DOI: 10.1007/s00281-017-0651-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 09/01/2017] [Indexed: 02/07/2023]
Abstract
The burden of newborn infectious disease has long been recognized as the highest across the entire human life span. The precise underlying cause is unfortunately still far from clear. A substantial body of data derived mostly from in vitro experimentation indicates "lower" host immune responses in early vs. adult life and is briefly summarized within this review. However, emerging data derived mostly from in vivo experimentation reveal that the newborn host also exhibits an exuberant immune and inflammatory response following infection when compared to the adult. In this context, it is important to emphasize that "infection" does not equate "infectious disease," as for many infections it is the host response to the infection that causes disease. This simple insight readily arranges existing evidence into cause-effect relationships that explain much of the increase in clinical suffering from infection in early life. We here briefly summarize the evidence in support of this paradigm and highlight the important implications it has for efforts to ameliorate the suffering and dying from infection in early life.
Collapse
Affiliation(s)
- Byron Brook
- Department of Experimental Medicine, University of British Columbia, UBC, BCCHRI A5-175, 950 W 28th Ave, Vancouver, BC, V5Z4H4, Canada
| | - Danny Harbeson
- Department of Experimental Medicine, University of British Columbia, UBC, BCCHRI A5-175, 950 W 28th Ave, Vancouver, BC, V5Z4H4, Canada
| | - Rym Ben-Othman
- Department of Pediatrics, Division of Infectious Diseases, University of British Columbia, Vancouver, Canada
| | - Dorothee Viemann
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Tobias R Kollmann
- Department of Experimental Medicine, University of British Columbia, UBC, BCCHRI A5-175, 950 W 28th Ave, Vancouver, BC, V5Z4H4, Canada. .,Department of Pediatrics, Division of Infectious Diseases, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
84
|
Nyalwidhe JO, Grzesik WJ, Burch TC, Semeraro ML, Waseem T, Gerling IC, Mirmira RG, Morris MA, Nadler JL. Comparative quantitative proteomic analysis of disease stratified laser captured microdissected human islets identifies proteins and pathways potentially related to type 1 diabetes. PLoS One 2017; 12:e0183908. [PMID: 28877242 PMCID: PMC5587329 DOI: 10.1371/journal.pone.0183908] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 08/14/2017] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes (T1D) is a chronic inflammatory disease that is characterized by autoimmune destruction of insulin-producing pancreatic beta cells. The goal of this study was to identify novel protein signatures that distinguish Islets from patients with T1D, patients who are autoantibody positive without symptoms of diabetes, and from individuals with no evidence of disease. High resolution high mass accuracy label free quantitative mass spectrometry analysis was applied to islets isolated by laser capture microdissection from disease stratified human pancreata from the Network for Pancreatic Organ Donors with Diabetes (nPOD), these included donors without diabetes, donors with T1D-associated autoantibodies in the absence of diabetes, and donors with T1D. Thirty-nine proteins were found to be differentially regulated in autoantibody positive cases compared to the no-disease group, with 25 upregulated and 14 downregulated proteins. For the T1D cases, 63 proteins were differentially expressed, with 24 upregulated and 39 downregulated, compared to the no disease controls. We have identified functional annotated enriched gene families and multiple protein-protein interaction clusters of proteins are involved in biological and molecular processes that may have a role in T1D. The proteins that are upregulated in T1D cases include S100A9, S100A8, REG1B, REG3A and C9 amongst others. These proteins have important biological functions, such as inflammation, metabolic regulation, and autoimmunity, all of which are pathways linked to the pathogenesis of T1D. The identified proteins may be involved in T1D development and pathogenesis. Our findings of novel proteins uniquely upregulated in T1D pancreas provides impetus for further investigations focusing on their expression profiles in beta cells/ islets to evaluate their role in the disease pathogenesis. Some of these molecules may be novel therapeutic targets T1D.
Collapse
Affiliation(s)
- Julius O. Nyalwidhe
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - Wojciech J. Grzesik
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - Tanya C. Burch
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - Michele L. Semeraro
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - Tayab Waseem
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - Ivan C. Gerling
- Division of Endocrinology, Diabetes and Metabolism, The University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Raghavendra G. Mirmira
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indiana University, Indianapolis, Indiana, United States of America
| | - Margaret A. Morris
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - Jerry L. Nadler
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
- The Strelitz Diabetes Center, Eastern Virginia Medical Center, Norfolk, Virginia, United States of America
| |
Collapse
|
85
|
Lood C, Tydén H, Gullstrand B, Jönsen A, Källberg E, Mörgelin M, Kahn R, Gunnarsson I, Leanderson T, Ivars F, Svenungsson E, Bengtsson AA. Platelet-Derived S100A8/A9 and Cardiovascular Disease in Systemic Lupus Erythematosus. Arthritis Rheumatol 2017; 68:1970-80. [PMID: 26946461 DOI: 10.1002/art.39656] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 02/18/2016] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Levels of S100A8/A9, a proinflammatory and prothrombotic protein complex, are increased in several diseases, and high levels predispose to cardiovascular disease (CVD). Recently, platelet S100A8/A9 synthesis was described in mice and humans in relation to CVD. The aim of this study was to investigate the role of platelet S100A8/A9 in systemic lupus erythematosus (SLE), a disease with markedly increased cardiovascular morbidity, as well as the exact platelet distribution of the S100A8/A9 proteins. METHODS The occurrence and distribution of platelet S100A8/A9 protein were detected by enzyme-linked immunosorbent assay, electron microscopy, Western blotting, and flow cytometry in healthy controls (n = 79) and in 2 individual cohorts of SLE patients (n = 148 and n = 318, respectively) and related to cardiovascular morbidity. RESULTS We observed that human platelets expressed S100A8/A9 proteins, and that these were localized in close proximity to intracellular membranes and granules as well as on the cell surface upon activation with physiologic and pathophysiologic stimuli. Interestingly, S100A8/A9 was enriched at sites of membrane interactions, indicating a role of S100A8/A9 in cell-cell communication. S100A8/A9 levels were highly regulated by interferon-α, both in vivo and in vitro. Patients with SLE had increased platelet S100A8/A9 content compared with healthy individuals. Increased levels of platelet S100A8/A9 were associated with CVD, particularly myocardial infarction (odds ratio 4.8, 95% confidence interval 1.5-14.9, P = 0.032 [adjusted for age, sex, and smoking]). CONCLUSION Platelets contain S100A8/A9 in membrane-enclosed vesicles, enabling rapid cell surface deposition upon activation. Furthermore, platelet S100A8/A9 protein levels were increased in SLE patients, particularly in those with CVD, and may be a future therapeutic target.
Collapse
Affiliation(s)
- Christian Lood
- Lund University and Skåne University Hospital, Lund, Sweden
| | - Helena Tydén
- Lund University and Skåne University Hospital, Lund, Sweden
| | | | - Andreas Jönsen
- Lund University and Skåne University Hospital, Lund, Sweden
| | | | | | | | - Iva Gunnarsson
- Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
86
|
Pepper RJ, Draibe JB, Caplin B, Fervenza FC, Hoffman GS, Kallenberg CGM, Langford CA, Monach PA, Seo P, Spiera R, William St Clair E, Tchao NK, Stone JH, Specks U, Merkel PA, Salama AD. Association of Serum Calprotectin (S100A8/A9) Level With Disease Relapse in Proteinase 3-Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. Arthritis Rheumatol 2017; 69:185-193. [PMID: 27428710 DOI: 10.1002/art.39814] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 07/07/2016] [Indexed: 02/01/2023]
Abstract
OBJECTIVE S100A8/A9 (calprotectin) has shown promise as a biomarker for predicting relapse in antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). This study was undertaken to investigate serum S100A8/A9 level as a biomarker for predicting future relapse in a large cohort of patients with severe AAV. METHODS Serum levels of S100A8/A9 were measured at baseline and months 1, 2, and 6 following treatment initiation in 144 patients in the Rituximab in ANCA-Associated Vasculitis trial (cyclophosphamide/azathioprine versus rituximab [RTX] for induction of remission) in whom complete remission was attained. RESULTS Patients were divided into 4 groups: proteinase 3 (PR3)-ANCA with relapse (n = 37), PR3-ANCA without relapse (n = 56), myeloperoxidase (MPO)-ANCA with relapse (n = 6), and MPO-ANCA without relapse (n = 45). Serum S100A8/A9 level decreased in all groups during the first 6 months of treatment. The percentage reduction from baseline to month 2 was significantly different between patients who experienced a relapse and those who did not in the PR3-ANCA group (P = 0.046). A significantly higher risk of relapse was associated with an increase in S100A8/A9 level between baseline and month 2 (P = 0.0043) and baseline and month 6 (P = 0.0029). Subgroup analysis demonstrated that patients treated with RTX who had increased levels of S100A8/A9 were at greatest risk of future relapse (P = 0.028). CONCLUSION An increase in serum S100A8/A9 level by month 2 or 6 compared to baseline identifies a subgroup of PR3-ANCA patients treated with RTX who are at higher risk of relapse by 18 months. Since RTX is increasingly used for remission induction in PR3-ANCA-positive patients experiencing a relapse, S100A8/A9 level may assist in identifying those patients requiring more intensive or prolonged treatment.
Collapse
Affiliation(s)
- Ruth J Pepper
- University College London Centre for Nephrology, Royal Free Hospital, London, UK
| | - Juliana B Draibe
- University College London Centre for Nephrology, Royal Free Hospital, London, UK
| | - Ben Caplin
- University College London Centre for Nephrology, Royal Free Hospital, London, UK
| | | | | | | | | | | | - Philip Seo
- Johns Hopkins University, Baltimore, Maryland
| | | | | | | | | | | | | | - Alan D Salama
- University College London Centre for Nephrology, Royal Free Hospital, London, UK
| | | |
Collapse
|
87
|
Austermann J, Zenker S, Roth J. S100-alarmins: potential therapeutic targets for arthritis. Expert Opin Ther Targets 2017; 21:739-751. [PMID: 28494625 DOI: 10.1080/14728222.2017.1330411] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION In arthritis, inflammatory processes are triggered by numerous factors that are released from joint tissues, promoting joint destruction and pathological progression. During inflammation, a novel family of pro-inflammatory molecules called alarmins is released, amplifying inflammation and joint damage. Areas covered: With regard to the role of the alarmins S100A8 and S100A9 in the pathogenesis of arthritis, recent advances and the future prospects in terms of therapeutic implications are considered. Expert opinion: There is still an urgent need for novel treatment strategies addressing the local mechanisms of joint inflammation and tissue destruction, offering promising therapeutic alternatives. S100A8 and S100A9, which are the most up-regulated alarmins during arthritis, are endogenous triggers of inflammation, defining these proteins as promising targets for local suppression of arthritis. In murine models, the blockade of S100A8/S100A9 ameliorates inflammatory processes, including arthritis, and there are several lines of evidence that S100-alarmins may already be targeted in therapeutic approaches in man.
Collapse
Affiliation(s)
- Judith Austermann
- a Institute of Immunology , University of Münster , Münster , Germany
| | - Stefanie Zenker
- a Institute of Immunology , University of Münster , Münster , Germany
| | - Johannes Roth
- a Institute of Immunology , University of Münster , Münster , Germany
| |
Collapse
|
88
|
Discriminating Value of Calprotectin in Disease Activity and Progression of Nonradiographic Axial Spondyloarthritis and Ankylosing Spondylitis. DISEASE MARKERS 2017. [PMID: 28630526 PMCID: PMC5463132 DOI: 10.1155/2017/7574147] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
It has been controversial whether ankylosing spondylitis (AS) and nonradiographic axial spondyloarthritis (nr-axSpA) are separate or different phases of radiographic progression. We determined that serum calprotectin level (ng/ml) was higher in AS (15.30 ± 6.49) and nr-axSpA (17.76 ± 8.59) patients than in healthy individuals (7.40 ± 2.67). No difference was observed in calprotectin level between these two groups. Elevated calprotectin was positively correlated with ESR, CRP, BASDAI, and ASDAS as well as SPARCC scoring and had no correlation with BASFI and mSASSS. No correlation was observed between calprotectin and Wnt/β-catenin pathway markers. Serum calprotectin can be used as a marker for inflammation in both nr-axSpA and AS, while it does not contribute to the discrimination of AS and nr-axSpA. Calprotectin-mediated inflammation was not correlated with principle effectors of Wnt/β-catenin pathway, indicating that inflammation and bone fusion might be separate processes of the disease.
Collapse
|
89
|
Ma L, Sun P, Zhang JC, Zhang Q, Yao SL. Proinflammatory effects of S100A8/A9 via TLR4 and RAGE signaling pathways in BV-2 microglial cells. Int J Mol Med 2017; 40:31-38. [PMID: 28498464 PMCID: PMC5466387 DOI: 10.3892/ijmm.2017.2987] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 04/24/2017] [Indexed: 01/11/2023] Open
Abstract
S100A8/A9, a heterodimer of the two calcium-binding proteins S100A8 and S100A9, has emerged as an important proinflammatory mediator in acute and chronic inflammation. However, whether S100A8/A9 is implicated in microglial-induced neuroinflammatory response remains unclear. Here, we found that S100A8/A9 significantly increased the secretion of proinflammatory cytokines including tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in cultured BV-2 microglial cells. Inhibition of the Toll-like receptor 4 (TLR4) and the receptor for advanced glycation end-products (RAGE) with C225 and a RAGE-blocking antibody, respectively significantly reduced the secretion of TNF-α and IL-6 from S100A8/A9-stimulated BV-2 microglial cells. Furthermore, S100A8/A9 markedly enhanced the nuclear translocation of NF-κB p65 and the DNA-binding activities of NF-κB in BV-2 microglial cells, and suppression of ERK and JNK/MAPK signaling pathways by PD98059 or SP600125 significantly inhibited NF-κB activity and the release of TNF-α and IL-6 in the S100A8/A9-treated BV-2 microglial cells. Our data also showed that inhibition of NF-κB with pyrrolidine dithiocarbamate (PDTC) significantly reduced the secretion of TNF-α and IL-6 from BV-2 microglial cells treated with S100A8/A9. Taken together, our data suggest that S100A8/A9 acts directly on BV-2 microglial cells via binding to TLR4 and RAGE on the membrane and then stimulates the secretion of proinflammatory cytokines through ERK and JNK-mediated NF-κB activity in BV-2 microglial cells. Targeting S100A8/A9 may provide a novel therapeutic strategy in microglial-induced neuroinflammatory diseases.
Collapse
Affiliation(s)
- Li Ma
- Department of Anesthesiology and Intensive Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Peng Sun
- Department of Emergency, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jian-Cheng Zhang
- Department of Anesthesiology and Intensive Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Qing Zhang
- Department of Anesthesiology and Intensive Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Shang-Long Yao
- Department of Anesthesiology and Intensive Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
90
|
Wiciński M, Malinowski B, Węclewicz MM, Grześk E, Grześk G. Anti-atherogenic properties of resveratrol: 4-week resveratrol administration associated with serum concentrations of SIRT1, adiponectin, S100A8/A9 and VSMCs contractility in a rat model. Exp Ther Med 2017; 13:2071-2078. [PMID: 28565810 DOI: 10.3892/etm.2017.4180] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/06/2016] [Indexed: 12/18/2022] Open
Abstract
Resveratrol (3, 4', 5-trihydroxy-trans-stilbene) is a natural, non-flavonoid polyphenol that exerts protective properties against atherosclerosis-associated endothelial dysfunction and senescence. The present study aimed to assess the influence of resveratrol on vascular contractility and molecular factors including sirtuin-1 (SIRT1), adiponectin and calprotectin (S100A8/A9) that are considered to be important elements of atherogenesis. A total of 17 male rats were divided into a control and treatment group and administered resveratrol or a placebo. Pharmacometrics were performed on an isolated and perfused tail artery. Serum SIRT1, adiponectin and S100A8/A9 levels were quantified using an ELISA assay. The level of SIRT1 in the control and treatment groups at time 0 was 4.26 and 4.45 ng/ml, respectively. SIRT1 in the control and treatment groups following 2 weeks of treatment was 4.59 and 6.86 ng/ml, respectively (P<0.05) and following 4 weeks of treatment was 4.15 and 6.38 ng/ml, respectively (P<0.05). The level of adiponectin in the control and treatment groups at time 0 was 1.24 and 1.21 ng/ml, respectively. Following 2 weeks of treatment, the level of adiponectin in the control and treatment groups was 1.22 and 1.2 ng/ml, respectively (P>0.05) and following 4 weeks of treatment was 1.26 and 1.58 ng/ml, respectively (P<0.05). The S100A8/A9 level in control and treatment groups at time 0 was 0.39 and 0.33 ng/ml, respectively. The level of S100A8/A9 in control and treatment groups following 2 weeks of treatment was 0.37 and 0.35 ng/ml, respectively (P>0.05) and following 4 weeks of treatment was 0.34 and 0.32 ng/ml, respectively (P>0.05). EC50 values obtained for phenylephrine in resveratrol-pretreated arteries were significantly higher than controls in the presence and absence of A7-hydrochloride (P<0.05). The results of the present study indicate a significant increase in the concentration of SIRT1 and adiponectin in the resveratrol-pretreated group (P<0.05). S100A8/A9 serum concentrations remained unchanged. Reactivity of resistant arteries was significantly reduced for resveratrol-pretreated vessels and this effect was partially independent of phosphodiesterase (PDE1). Additionally, there was a synergistic interaction observed between resveratrol and the PDE1 inhibitor.
Collapse
Affiliation(s)
- Michał Wiciński
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Bartosz Malinowski
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Mateusz M Węclewicz
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Elżbieta Grześk
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Grzegorz Grześk
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| |
Collapse
|
91
|
Buyukterzi Z, Can U, Alpaydin S, Guzelant A, Karaarslan S, Kocyigit D, Gurses KM. Enhanced S100A9 and S100A12 expression in acute coronary syndrome. Biomark Med 2017; 11:229-237. [PMID: 28157385 DOI: 10.2217/bmm-2016-0253] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
AIMS In this study, we aimed to investigate whether serum S100A8, S100A9 and S100A12 levels were markers of acute coronary syndrome (ACS). MATERIALS & METHODS Patients who underwent coronary angiography and/or percutaneous coronary interventions between June 2015-October 2015 were consecutively recruited in this study and categorized three groups each containing 30 patients (normal coronary arteries, stable coronary artery disease, and acute coronary syndrome). Baseline characteristics, including co- morbidities and medications, were recorded and serum S100A8, S100A9, S100A12, and C- reactive protein levels were measured besides routine laboratory tests. RESULTS A total of 90 patients (63.00 [56.00-73.00] years, 62.89% male) have been included. None of the groups differed from each other regarding baseline characteristics (p > 0.05). S100A9 levels were elevated in ACS when compared with the normal coronary arteries (p = 0.033) and S100A12 levels were found to be elevated in ACS when compared with both patients with normal coronary arteries and stable coronary artery disease (p = 0.001). S100A12 was identified as an independent associate of ACS (p = 0.002). CONCLUSION These results suggest that S100A12 may serve as a marker of coronary plaque instability, and may have a therapeutic implication in ACS treatment.
Collapse
Affiliation(s)
- Zafer Buyukterzi
- Department of Cardiology, Konya Training & Research Hospital, University of Health Sciences, 42090 Meram Konya, Turkey
| | - Ummugulsum Can
- Department of Biochemistry, Konya Training & Research Hospital, University of Health Sciences, 42090 Meram Konya, Turkey
| | - Sertac Alpaydin
- Department of Cardiology, Konya Training & Research Hospital, University of Health Sciences, 42090 Meram Konya, Turkey
| | - Asuman Guzelant
- Department of Microbiology & Infectious Diseases, Konya Training & Research Hospital, University of Health Sciences, 42090 Meram Konya, Turkey
| | - Sukru Karaarslan
- Department of Cardiology, Ufuk University Faculty of Medicine, 06520 Balgat Ankara, Turkey
| | - Duygu Kocyigit
- Department of Cardiology, Hacettepe University Faculty of Medicine, 06100 Sihhiye Ankara, Turkey
| | - Kadri Murat Gurses
- Department of Cardiology, Konya Training & Research Hospital, University of Health Sciences, 42090 Meram Konya, Turkey
| |
Collapse
|
92
|
Muthas D, Reznichenko A, Balendran CA, Böttcher G, Clausen IG, Kärrman Mårdh C, Ottosson T, Uddin M, MacDonald TT, Danese S, Berner Hansen M. Neutrophils in ulcerative colitis: a review of selected biomarkers and their potential therapeutic implications. Scand J Gastroenterol 2017; 52:125-135. [PMID: 27610713 DOI: 10.1080/00365521.2016.1235224] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES This review article describes the role of neutrophils in mucosal injury and the resulting crypt abscesses characteristic of ulcerative colitis. We also review selected biomarkers for monitoring neutrophil presence and activity in the mucosa as well as their potential as therapeutic targets. MATERIAL We have collated and selectively reviewed data on the most prominent well-established and emerging neutrophil-related biomarkers and potential therapeutic targets (calprotectin, lactoferrin, CXCR1, CXCR2, MMP-9, NGAL, elafin, HNE, pANCAs, MPO, CD16, CD177, CD64, HNPs, SLPI and PTX3) in ulcerative colitis. RESULTS Systemic and intestinal neutrophil activity increases substantially in active ulcerative colitis, driving tissue damage and extra-intestinal manifestations. Calprotectin is a robust neutrophil and disease biomarker, and a few neutrophil-related targets are being clinically explored as therapeutic targets. CONCLUSION We propose that targeting neutrophils and their inflammatory mediators per se is an opportunity that should be explored to identify new effective medical therapies. The overall clinical goal for neutrophil-targeted therapy will be to modulate, but not completely silence, neutrophil activity, thereby abolishing the destructive inflammation with associated acute and chronic tissue damage without compromising host-defense.
Collapse
Affiliation(s)
- Daniel Muthas
- a Department of Respiratory , Inflammation and Autoimmunity, AstraZeneca R&D Gothenburg , Mölndal , Sweden
| | - Anna Reznichenko
- b Department of Cardiovascular and Metabolic Diseases , AstraZeneca R&D Gothenburg , Mölndal , Sweden
| | - Clare A Balendran
- c Department of Personalised HealthCare & Biomarkers , AstraZeneca R&D Gothenburg , Mölndal , Sweden
| | - Gerhard Böttcher
- d Department of Drug Safety and Metabolism , AstraZeneca R&D Gothenburg , Mölndal , Sweden
| | - Ib Groth Clausen
- a Department of Respiratory , Inflammation and Autoimmunity, AstraZeneca R&D Gothenburg , Mölndal , Sweden
| | - Carina Kärrman Mårdh
- a Department of Respiratory , Inflammation and Autoimmunity, AstraZeneca R&D Gothenburg , Mölndal , Sweden
| | - Tomas Ottosson
- a Department of Respiratory , Inflammation and Autoimmunity, AstraZeneca R&D Gothenburg , Mölndal , Sweden
| | - Mohib Uddin
- c Department of Personalised HealthCare & Biomarkers , AstraZeneca R&D Gothenburg , Mölndal , Sweden
| | - Thomas T MacDonald
- e Blizard Institute, Barts and the London School of Medicine and Dentistry, QMUL , London , UK
| | - Silvio Danese
- f Department of Gastroenterology , IBD Center, Humanitas Research Hospital , Milan , Italy
| | - Mark Berner Hansen
- a Department of Respiratory , Inflammation and Autoimmunity, AstraZeneca R&D Gothenburg , Mölndal , Sweden.,g Digestive Disease Center K, Bispebjerg Hospital, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
93
|
Heinemann AS, Pirr S, Fehlhaber B, Mellinger L, Burgmann J, Busse M, Ginzel M, Friesenhagen J, von Köckritz-Blickwede M, Ulas T, von Kaisenberg CS, Roth J, Vogl T, Viemann D. In neonates S100A8/S100A9 alarmins prevent the expansion of a specific inflammatory monocyte population promoting septic shock. FASEB J 2016; 31:1153-1164. [PMID: 27993995 DOI: 10.1096/fj.201601083r] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 11/28/2016] [Indexed: 12/28/2022]
Abstract
The high susceptibility of newborn infants to sepsis is ascribed to an immaturity of the neonatal immune system, but the molecular mechanisms remain unclear. Newborn monocytes massively release the alarmins S100A8/S100A9. In adults, these are major regulators of immunosuppressive myeloid-derived suppressor cells (MDSCs). We investigated whether S100A8/S100A9 cause an expansion of monocytic MDSCs (Mo-MDSCs) in neonates, thereby contributing to an immunocompromised state. Mo-MDSCs have been assigned to CD14+/human leukocyte antigen (HLA)-DR-/low/CD33+ monocytes in humans and to CD11b+/Gr-1int/Ly6G-/Ly6Chi cells in mice. We found monocytes with these phenotypes significantly expanded in their respective newborns. Functionally, however, they did not prove immunosuppressive but rather responded inflammatorily to microbial stimulation. Their expansion did not correlate with high S100A8/S100A9 levels in cord blood. Murine studies revealed an excessive expansion of CD11b+/Gr-1int/Ly6G-/Ly6Chi monocytes in S100A9-/- neonates compared to wild-type neonates. This strong baseline expansion was associated with hyperinflammatory responses during endotoxemia and fatal septic courses. Treating S100A9-/- neonates directly after birth with S100A8/S100A9 alarmins prevented excessive expansion of this inflammatory monocyte population and death from septic shock. Our data suggest that a specific population of inflammatory monocytes promotes fatal courses of sepsis in neonates if its expansion is not regulated by S100A8/S100A9 alarmins.-Heinemann, A. S., Pirr, S., Fehlhaber, B., Mellinger, L., Burgmann, J., Busse, M., Ginzel, M., Friesenhagen, J., von Köckritz-Blickwede, M., Ulas, T., von Kaisenberg, C. S., Roth, J., Vogl, T., Viemann, D. In neonates S100A8/S100A9 alarmins prevent the expansion of a specific inflammatory monocyte population promoting septic shock.
Collapse
Affiliation(s)
- Anna S Heinemann
- Department of Pediatric Pneumology, Allergy, and Neonatology, Hannover Medical School, Hannover, Germany
| | - Sabine Pirr
- Department of Pediatric Pneumology, Allergy, and Neonatology, Hannover Medical School, Hannover, Germany
| | - Beate Fehlhaber
- Department of Pediatric Pneumology, Allergy, and Neonatology, Hannover Medical School, Hannover, Germany
| | - Lara Mellinger
- Department of Pediatric Pneumology, Allergy, and Neonatology, Hannover Medical School, Hannover, Germany
| | - Johanna Burgmann
- Department of Pediatric Pneumology, Allergy, and Neonatology, Hannover Medical School, Hannover, Germany
| | - Mandy Busse
- Department of Pediatric Pneumology, Allergy, and Neonatology, Hannover Medical School, Hannover, Germany
| | - Marco Ginzel
- Department of Pediatric Pneumology, Allergy, and Neonatology, Hannover Medical School, Hannover, Germany
| | - Judith Friesenhagen
- Department of Pediatric Pneumology, Allergy, and Neonatology, Hannover Medical School, Hannover, Germany
| | - Maren von Köckritz-Blickwede
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Hannover, Germany.,Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Thomas Ulas
- Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | | | - Johannes Roth
- Institute of Immunology, University of Münster, Münster, Germany.,Interdisciplinary Centre for Clinical Research, University of Münster, Münster, Germany; and
| | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany.,Interdisciplinary Centre for Clinical Research, University of Münster, Münster, Germany; and
| | - Dorothee Viemann
- Department of Pediatric Pneumology, Allergy, and Neonatology, Hannover Medical School, Hannover, Germany;
| |
Collapse
|
94
|
Neutrophils and the S100A9 protein critically regulate granuloma formation. Blood Adv 2016; 1:184-192. [PMID: 29296934 DOI: 10.1182/bloodadvances.2016000497] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 10/27/2016] [Indexed: 12/22/2022] Open
Abstract
Macrophages have the potential to undergo cellular transformation into epithelioid cells, and their concentric accumulation in tissues results in the development of granulomas. Although epithelioid cells are an essential and dominant component of granulomas, other cell types have also been detected, which may contribute to the establishment of well-organized granulomas, as observed in human granulomatous diseases. We herein demonstrated that neutrophils may mediate these functions. By taking advantage of the guinea pig pulmonary granuloma model, we obtained a rat monoclonal antibody with unique reactivity to granuloma cells. This antibody, termed G213, reacted with clusters of neutrophils located in the central area of granulomas, and a biochemical analysis identified the G213-reactive antigen as S100A9, a calcium-binding protein of the S100 family, which was expressed abundantly in neutrophils. Consistent with the multifaceted functions attributed to S100A9, including its role in neutrophil extravasation and macrophage activation, the blockade of S100A9 functions with the specific inhibitor, tasquinimod, impaired the formation of organized granulomas with neutrophil cores. These results demonstrate the critical role of neutrophils and the S100A9 protein in granuloma formation. Because intragranuloma S100A9+ neutrophils were also detected in humans, these results indicate the potential of tasquinimod, a new anticancer drug candidate, for manipulating human granulomatous diseases.
Collapse
|
95
|
Li S, Xu F, Li H, Zhang J, Zhong A, Huang B, Lai M. S100A8 + stroma cells predict a good prognosis and inhibit aggressiveness in colorectal carcinoma. Oncoimmunology 2016; 6:e1260213. [PMID: 28197382 DOI: 10.1080/2162402x.2016.1260213] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/04/2016] [Accepted: 11/07/2016] [Indexed: 01/07/2023] Open
Abstract
Gene microarray and bioinformatic analysis showed that S100A8 was more abundant in the stroma surrounding tumor buddings (TBs) than in the stroma surrounding primary tumor cells in colorectal carcinomas. Here, S100A8+ cells in 419 colorectal carcinoma samples were stained by immunohistochemistry and counted using Image-pro plus 6.0. TBs were also counted and biomarkers associated with the epithelial-mesenchymal transition and apoptosis were assessed by immunohistochemistry. We evaluated the association between S100A8+ cells and clinico-pathological variables as well as survival. Migration and invasion as well as biomarkers of the epithelial-mesenchymal transition and apoptosis were tested in CRC cells, treated with graded concentrations of recombinant human S100A8 protein. We found that the density of S100A8+ cells in the tumor invasive front (S100A8+TIF) clearly distinguished patients with 5-y survival from those who did not survive (p = 0.01). The S100A8+-associated tumor budding (SATB) index determined by the S100A8+TIF and TB was an independent predictor of overall survival (p = 0.001) other than the S100A8+TIF or TB alone. Migration and invasion properties of CRC cells were inhibited by recombinant human S100A8 treatment. The particular S100A8+ cells in the stroma were associated with important biomarkers of the epithelial-mesenchymal transition (E-cadherin and SNAIL) and apoptosis (BCL2). In conclusion, S100A8+ cells in the stroma predict a good prognosis in colorectal carcinoma. An index combining S100A8+ cells and TB independently predicts survival. Recombinant human S100A8 inhibited CRC cell migration and invasion, which was involved in epithelial-mesenchymal transition (E-cadherin and SNAIL) and apoptosis (BCL2).
Collapse
Affiliation(s)
- Si Li
- Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China
| | - Fangying Xu
- Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China
| | - Hui Li
- Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China
| | - Jing Zhang
- Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China
| | - Anjing Zhong
- Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China
| | - Bin Huang
- Department of Pathology, The First Peoples Hospital of Xiaoshan , Hangzhou, Xiaoshan, China
| | - Maode Lai
- Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China
| |
Collapse
|
96
|
Exploring experimental cerebral malaria pathogenesis through the characterisation of host-derived plasma microparticle protein content. Sci Rep 2016; 6:37871. [PMID: 27917875 PMCID: PMC5137300 DOI: 10.1038/srep37871] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 10/28/2016] [Indexed: 01/09/2023] Open
Abstract
Cerebral malaria (CM) is a severe complication of Plasmodium falciparum infection responsible for thousands of deaths in children in sub-Saharan Africa. CM pathogenesis remains incompletely understood but a number of effectors have been proposed, including plasma microparticles (MP). MP numbers are increased in CM patients’ circulation and, in the mouse model, they can be localised within inflamed vessels, suggesting their involvement in vascular damage. In the present work we define, for the first time, the protein cargo of MP during experimental cerebral malaria (ECM) with the overarching hypothesis that this characterisation could help understand CM pathogenesis. Using qualitative and quantitative high-throughput proteomics we compared MP proteins from non-infected and P. berghei ANKA-infected mice. More than 360 proteins were identified, 60 of which were differentially abundant, as determined by quantitative comparison using TMTTM isobaric labelling. Network analyses showed that ECM MP carry proteins implicated in molecular mechanisms relevant to CM pathogenesis, including endothelial activation. Among these proteins, the strict association of carbonic anhydrase I and S100A8 with ECM was verified by western blot on MP from DBA/1 and C57BL/6 mice. These results demonstrate that MP protein cargo represents a novel ECM pathogenic trait to consider in the understanding of CM pathogenesis.
Collapse
|
97
|
Abu El-Asrar AM, Alam K, Siddiquei MM, Van den Eynde K, Mohammad G, De Hertogh G, Opdenakker G. Myeloid-Related Protein-14/MRP-14/S100A9/Calgranulin B is Associated with Inflammation in Proliferative Diabetic Retinopathy. Ocul Immunol Inflamm 2016; 26:615-624. [PMID: 27849448 DOI: 10.1080/09273948.2016.1245759] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE To investigate the expression of the leukocyte proteins myeloid-related protein (MRP)-8 and MRP-14 in proliferative diabetic retinopathy (PDR) and the effect of MRP-8/MRP-14 (calprotectin) heterodimer on induction of proinflammatory factors in human retinal microvascular endothelial cells (HRMEC). METHODS Epiretinal membranes from 20 patients with PDR and 10 patients with proliferative vitreoretinopathy (PVR), vitreous fluid samples from PDR and non-diabetic subjects and HRMEC were studied by immunohistochemistry and Western blot analysis. RESULTS MRP-14 expression was localized in endothelial cells, leukocytes and myofibroblasts in all PDR membranes. MRP-8 expression was limited to intravascular leukocytes in 42% of the studied membranes. In PVR membranes, MRP-14 was expressed in leukocytes and myofibroblasts, whereas MRP-8 immunoreactivity was limited to leukocytes. MRP-14 was significantly upregulated in vitreous from PDR patients. MRP-8/MRP-14 (calprotectin) increased expression of intercellular adhesion molecule-1, but attenuated vascular cell adhesion molecule-1 expression in HRMEC. CONCLUSIONS Increased MRP-14 levels are associated with inflammation in PDR.
Collapse
Affiliation(s)
- Ahmed M Abu El-Asrar
- a Department of Ophthalmology , College of Medicine, King Saud University , Riyadh , Saudi Arabia.,b Dr Nasser Al-Rashid Research Chair in Ophthalmology , Riyadh , Saudi Arabia
| | - Kaiser Alam
- a Department of Ophthalmology , College of Medicine, King Saud University , Riyadh , Saudi Arabia
| | - Mohammad M Siddiquei
- a Department of Ophthalmology , College of Medicine, King Saud University , Riyadh , Saudi Arabia
| | - Kathleen Van den Eynde
- c Laboratory of Histochemistry and Cytochemistry, University of Leuven , KU Leuven , Leuven , Belgium
| | - Ghulam Mohammad
- a Department of Ophthalmology , College of Medicine, King Saud University , Riyadh , Saudi Arabia
| | - Gert De Hertogh
- c Laboratory of Histochemistry and Cytochemistry, University of Leuven , KU Leuven , Leuven , Belgium
| | - Ghislain Opdenakker
- d Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology and Immunology , University of Leuven , KU Leuven , Leuven , Belgium
| |
Collapse
|
98
|
Lee HW, Jie HB, Bollyky PL, Sarracino D, Kim TS, Wilson BS. Role of dendritic cell maturation factors produced by human invariant NKT cells in immune tolerance. J Leukoc Biol 2016; 101:989-1003. [PMID: 27837018 DOI: 10.1189/jlb.1a0416-164rrr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 10/04/2016] [Accepted: 10/27/2016] [Indexed: 01/12/2023] Open
Abstract
In this study, we used the culture supernatant of iNKT cells to identify human myeloid DC maturation factors produced by human CD4+ iNKT cells. S100A8 had a strong maturation effect. Notably, the recombinant S100A8 protein displayed properties of DC maturation functioning, and the induction of DC differentiation by both the purified and the recombinant protein were blocked by anti-S100A8 and anti-TLR-4 mAbs. DC differentiation induced by anti-major histocompatibility complex class II/CD1d Ab, S100A8, or both was qualitatively indistinguishable from that induced by the coculture of DCs and iNKT cells or via culture supplementation with supernatants from activated CD4+ iNKT cells. S100A8 also induced CD4+/CD25+/Foxp3+ Treg cells from naïve T cells. S100A8 may contribute to DC differentiation by elevating transcription factors or activating transcription factor-2, heat shock factor-1, or both, in mature DCs. S100A8 is a novel candidate iNKT cell-dependent DC maturation factor.
Collapse
Affiliation(s)
- Hyeong-Woo Lee
- Departments of Tropical Medicine and Parasitology, Inha University School of Medicine, Incheon, Republic of Korea.,Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida
| | - Hyun Bae Jie
- OncoMed Pharmaceuticals, Inc., Redwood City, California
| | - Paul L Bollyky
- Division of Infectious Diseases, Stanford University Medical Center, Stanford, California; and
| | - David Sarracino
- Thermo Fisher Scientific Biomarkers Research Initiatives in Mass Spectrometry (BRIMS) Center, Cambridge, Massachusetts
| | - Tong-Soo Kim
- Departments of Tropical Medicine and Parasitology, Inha University School of Medicine, Incheon, Republic of Korea;
| | - Brian S Wilson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida;
| |
Collapse
|
99
|
Treatment with Quinoline-3-carboxamide does not successfully prevent immune-mediated glomerulonephritis in mice. Nefrologia 2016; 36:687-693. [DOI: 10.1016/j.nefro.2016.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 02/29/2016] [Accepted: 03/16/2016] [Indexed: 11/19/2022] Open
|
100
|
Landers-Ramos RQ, Sapp RM, VandeWater E, Macko J, Robinson S, Wang Y, Chin ER, Spangenburg EE, Prior SJ, Hagberg JM. Investigating the extremes of the continuum of paracrine functions in CD34-/CD31+ CACs across diverse populations. Am J Physiol Heart Circ Physiol 2016; 312:H162-H172. [PMID: 27793853 DOI: 10.1152/ajpheart.00342.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 09/30/2016] [Accepted: 10/14/2016] [Indexed: 11/22/2022]
Abstract
Paracrine function of circulating angiogenic cells (CACs) is thought to contribute to vascular maintenance. We previously identified S100A8 and S100A9 secreted from physically inactive individuals' CD34-/CD31+ CACs as negative regulators of capillary-like network formation. The purpose of this study was to investigate further the extremes of the continuum of CAC paracrine actions using two distinctly different groups representing "healthy" and "impaired" CAC function. We aimed to determine how capillary-like network formation in human umbilical vein endothelial cells (HUVECs) is affected by S100A8 and S100A9 in concentrations secreted by CACs from different ends of the health spectrum. CD34-/CD31+ CACs were isolated and cultured from 10 impaired function individuals defined as older (50-89 yr), non-ST-elevation myocardial infarction patients and 10 healthy individuals defined as younger (18-35 yr), healthy individuals, and conditioned media (CM) was generated. CM from the impaired function group's CACs significantly diminished network formation compared with CM from the healthy group (P < 0.05). We identified elevations in S100A8, S100A9, and S100A8/A9 in the CM from the impaired function group (P < 0.05). Pretreatment of HUVECs with inhibitors to a known S100A8 and S100A9 receptor, Toll-like receptor 4 (TLR4), but not receptor for advanced glycation end products, improved HUVEC network formation (P < 0.05) compared with CM alone in the impaired function conditions. Exposure of HUVECs to the TLR4 signaling inhibitor also blocked recombinant S100A8- and S100A9-mediated reductions in network formation. Collectively, the results suggest that the mechanisms behind impaired CAC CD34-/CD31+ CM-mediated reductions in capillary-like network formation involve secretion of S100A8 and S100A9 and binding of these proteins to TLR4 receptors on HUVECs. NEW & NOTEWORTHY S100A8 and S100A9 proteins in concentrations secreted by CD34-/CD31+ circulating angiogenic cells (CACs) with impaired function reduce endothelial cell capillary-like network formation. These effects appear to be mediated by Toll-like receptor 4 and are absent with S100A8 and S100A9 in concentrations secreted by healthy CD34-/CD31+ CACs.
Collapse
Affiliation(s)
- Rian Q Landers-Ramos
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Ryan M Sapp
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Emily VandeWater
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Jennifer Macko
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Shawn Robinson
- Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Yan Wang
- Proteomics Core Facility, College of Computer, Mathematics, and Natural Sciences, University of Maryland, College Park, Maryland; and
| | - Eva R Chin
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Espen E Spangenburg
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Steven J Prior
- University of Maryland School of Medicine and Baltimore Veterans Affairs Geriatric Research, Education and Clinical Center, Baltimore, Maryland
| | - James M Hagberg
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland;
| |
Collapse
|