51
|
Proteomic Studies of Blood and Vascular Wall in Atherosclerosis. Int J Mol Sci 2021; 22:ijms222413267. [PMID: 34948066 PMCID: PMC8707794 DOI: 10.3390/ijms222413267] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 12/12/2022] Open
Abstract
The review is devoted to the analysis of literature data related to the role of proteomic studies in the study of atherosclerotic cardiovascular diseases. Diagnosis of patients with atherosclerotic plaques before clinical manifestations is an arduous task. The review presents the results of research on the new proteomic potential biomarkers of coronary heart disease, coronary atherosclerosis, acute coronary syndrome, myocardial infarction, carotid artery atherosclerosis. Also, the analysis of literature data on proteomic studies of the vascular wall was carried out. To assess the involvement of proteins in the pathological process of atherosclerosis, it is important to investigate the specific relationships between proteins in the arteries, expression and concentration of proteins. The development of proteomic technologies has made it possible to analyse the number of proteins associated with the development of the disease. Analysis of the proteomic profile of the vascular wall in atherosclerosis can help to detect possible diagnostically significant protein structures or potential biomarkers of the disease and develop novel approaches to the diagnosis of atherosclerosis and its complications.
Collapse
|
52
|
Sun Y, Wang Z, Wang C, Tang Z, Zhao H. Psycho-cardiology therapeutic effects of Shuangxinfang in rats with depression-behavior post acute myocardial infarction: Focus on protein S100A9 from proteomics. Biomed Pharmacother 2021; 144:112303. [PMID: 34673424 DOI: 10.1016/j.biopha.2021.112303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Depressive disorders induced by acute myocardial infarction (AMI) play a pivotal role in the deterioration of cardiac function, and Shuangxinfang (Psycho-cardiology Formula, PCF) was reported to alleviate heart function damage and improve depression-like behavior, but the complex mechanism in such process has not been clarified. METHODS AMI models were established and PCF was administered in rats. Subjects were then assessed in open field test (OFT) and forced swimming test (FST) recapitulating symptoms of depressive disorder. Afterward, pharmacoproteomic profiling of the hippocampus and peri-infarct border zone (BZ) was performed using a label-free liquid chromatography-tandem mass spectrometry (LC-MS/MS) technique, to identify contributing proteins and pathways responsible for myocardial ischemia and behavioral allostasis. Bioinformatics analysis was processed for further investigation, while western blotting was employed for testing dominating proteins to validate proteomic results. RESULTS Rats in the AMI group showed depression-like behavior in OFT and FST, which was improved by PCF. There were 131 differentially expressed proteins (DEPs) in BZ and 64 proteins in the hippocampus being detected and quantified shared by the sham group, the AMI group, and the PCF group. Subsequently, pertinent pathways and molecular functions were further identified. Altered molecules were discovered to be enriched in the apoptotic process, innate immune response, and NF-κB transcription factor activity in BZ, as well as chemical synaptic transmission, axon, collagen binding, cell adhesion, response to carbohydrate, laminin binding, and cellular response to nitric oxide in the hippocampus. Groups of signal transducers were also able to select multiple pathways, including innate immunity and arginine biosynthesis in the heart, also integrin signaling in the brain. DEPs were intersected from the myocardium and hippocampus to screen out the protein S100A9, which was up-regulated in the AMI group compared with the sham, and showed a down-regulation trend after treatment with PCF. CONCLUSION Taken together, we present a comprehensive proteomics analysis of rat models with depression post-AMI. Reviewing the literatures concerned, it's hypothesized that macrophage/microglia inflammation mediated by S100A9 might be the pivotal pathogenic process of psycho-cardiology disease, as well as potential mechanisms for the treatment of PCF.
Collapse
Affiliation(s)
- Yize Sun
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zheyi Wang
- Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Chunguo Wang
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhuoran Tang
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Haibin Zhao
- The DongFang Hospital of Beijing University of Chinese Medicine, Beijing, 100078, China.
| |
Collapse
|
53
|
Kim RY, Sunkara KP, Bracke KR, Jarnicki AG, Donovan C, Hsu AC, Ieni A, Beckett EL, Galvão I, Wijnant S, Ricciardolo FL, Di Stefano A, Haw TJ, Liu G, Ferguson AL, Palendira U, Wark PA, Conickx G, Mestdagh P, Brusselle GG, Caramori G, Foster PS, Horvat JC, Hansbro PM. A microRNA-21-mediated SATB1/S100A9/NF-κB axis promotes chronic obstructive pulmonary disease pathogenesis. Sci Transl Med 2021; 13:eaav7223. [PMID: 34818056 DOI: 10.1126/scitranslmed.aav7223] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Richard Y Kim
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, New South Wales 2007, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia
| | - Krishna P Sunkara
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia.,Graduate School of Health, Discipline of Pharmacy, University of Technology Sydney, Sydney, New South Wales 2007, Australia.,Intensive Care Unit, John Hunter Hospital, Newcastle, New South Wales 2308, Australia
| | - Ken R Bracke
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent 9000, Belgium
| | - Andrew G Jarnicki
- Department of Biochemistry and Pharmacology, University of Melbourne, Victoria 3010, Australia
| | - Chantal Donovan
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, New South Wales 2007, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia
| | - Alan C Hsu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia
| | - Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Section of Anatomic Pathology, University of Messina, Messina 98100, Italy
| | - Emma L Beckett
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia
| | - Izabela Galvão
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Sara Wijnant
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent 9000, Belgium
| | - Fabio Lm Ricciardolo
- Rare Lung Disease Unit, Department of Clinical and Biological Sciences, University of Torino, San Luigi Gonzaga University Hospital Orbassano, Torino 10043, Italy
| | - Antonino Di Stefano
- Istituti Clinici Scientifici Maugeri, IRCCS, SpA Società Benefit, Divisione di Pneumologia e Laboratorio di Citoimmunopatologia dell'Apparato Cardio Respiratorio, Veruno, Novara 28100, Italy
| | - Tatt Jhong Haw
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia
| | - Gang Liu
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Angela L Ferguson
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales 2006, Australia.,Centenary Institute and University of Technology Sydney, Sydney, New South Wales 2006, Australia
| | - Umamainthan Palendira
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Peter A Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia
| | - Griet Conickx
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent 9000, Belgium.,Ablynx N.V., a Sanofi company, Ghent 9052, Belgium
| | - Pieter Mestdagh
- Center for Medical Genetics and Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| | - Guy G Brusselle
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent 9000, Belgium
| | - Gaetano Caramori
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina 98100, Italy
| | - Paul S Foster
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, New South Wales 2007, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia
| |
Collapse
|
54
|
Hudson J, Farkas L. Epigenetic Regulation of Endothelial Dysfunction and Inflammation in Pulmonary Arterial Hypertension. Int J Mol Sci 2021; 22:ijms222212098. [PMID: 34829978 PMCID: PMC8617605 DOI: 10.3390/ijms222212098] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/05/2021] [Accepted: 11/06/2021] [Indexed: 12/13/2022] Open
Abstract
Once perceived as a disorder treated by vasodilation, pulmonary artery hypertension (PAH) has emerged as a pulmonary vascular disease with severe endothelial cell dysfunction. In the absence of a cure, many studies seek to understand the detailed mechanisms of EC regulation to potentially create more therapeutic options for PAH. Endothelial dysfunction is characterized by complex phenotypic changes including unchecked proliferation, apoptosis-resistance, enhanced inflammatory signaling and metabolic reprogramming. Recent studies have highlighted the role of epigenetic modifications leading to pro-inflammatory response pathways, endothelial dysfunction, and the progression of PAH. This review summarizes the existing literature on epigenetic mechanisms such as DNA methylation, histone modifications, and non-coding RNAs, which can lead to aberrant endothelial function. Our goal is to develop a conceptual framework for immune dysregulation and epigenetic changes in endothelial cells in the context of PAH. These studies as well as others may lead to advances in therapeutics to treat this devastating disease.
Collapse
|
55
|
Wei D, Li R, Si T, He H, Wu W. Screening and bioinformatics analysis of key biomarkers in acute myocardial infarction. Pteridines 2021. [DOI: 10.1515/pteridines-2020-0031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Abstract
Acute myocardial infarction (AMI) is the most severe manifestation of coronary artery disease. Considerable efforts have been made to elucidate its etiology and pathology, but the genetic factors that play a decisive role in the occurrence of AMI are still unclear. To determine the molecular mechanism of the occurrence and development of AMI, four microarray datasets, namely, GSE29111, GSE48060, GSE66360, and GSE97320, were downloaded from the Gene Expression Omnibus (GEO) database. We analyzed the four GEO datasets to obtain the differential expression genes (DEGs) of patients with AMI and patients with non-AMI and then performed gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, and Protein-protein interaction (PPI) network analysis. A total of 41 DEGs were identified, including 39 upregulated genes and 2 downregulated genes. The enriched functions and pathways of the DEGs included the inflammatory response, neutrophil chemotaxis, immune response, extracellular space, positive regulation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) transcription factor activity, response to lipopolysaccharide, receptor for advanced glycation end products (RAGE) receptor binding, innate immune response, defense response to bacterium, and receptor activity. The cytoHubba plug-in in Cytoscape was used to select the most significant hub gene from the PPI network. Ten hub genes were identified, and GO enrichment analysis revealed that these genes were mainly enriched in inflammatory response, neutrophil chemotaxis, immune response, RAGE receptor binding, and extracellular region. In conclusion, this study integrated four datasets and used bioinformatics methods to analyze the gene chips of AMI samples and control samples and identified DEGs that may be involved in the occurrence and development of AMI. The study provides reliable molecular biomarkers for AMI screening, diagnosis, and prognosis.
Collapse
Affiliation(s)
- Dongmei Wei
- Department of Cardiovasology, Liuzhou Traditional Chinese Medical Hospital , Liuzhou , Guangxi Province, 545001 , People’s Republic of China
| | - Rui Li
- Guangzhou University of Chinese Medicine , Guangzhou , Guangdong Province, 510405 , People’s Republic of China
| | - Tao Si
- Guangzhou University of Chinese Medicine , Guangzhou , Guangdong Province, 510405 , People’s Republic of China
| | - Hankang He
- Department of Cardiovasology, Liuzhou Traditional Chinese Medical Hospital , Liuzhou , Guangxi Province, 545001 , People’s Republic of China
| | - Wei Wu
- Guangzhou University of Chinese Medicine , Guangzhou , Guangdong Province, 510405 , People’s Republic of China
| |
Collapse
|
56
|
Mihaila AC, Ciortan L, Macarie RD, Vadana M, Cecoltan S, Preda MB, Hudita A, Gan AM, Jakobsson G, Tucureanu MM, Barbu E, Balanescu S, Simionescu M, Schiopu A, Butoi E. Transcriptional Profiling and Functional Analysis of N1/N2 Neutrophils Reveal an Immunomodulatory Effect of S100A9-Blockade on the Pro-Inflammatory N1 Subpopulation. Front Immunol 2021; 12:708770. [PMID: 34447377 PMCID: PMC8384118 DOI: 10.3389/fimmu.2021.708770] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/23/2021] [Indexed: 12/21/2022] Open
Abstract
Neutrophils have been classically viewed as a homogenous population. Recently, neutrophils were phenotypically classified into pro-inflammatory N1 and anti-inflammatory N2 sub-populations, but the functional differences between the two subtypes are not completely understood. We aimed to investigate the phenotypic and functional differences between N1 and N2 neutrophils, and to identify the potential contribution of the S100A9 alarmin in neutrophil polarization. We describe distinct transcriptomic profiles and functional differences between N1 and N2 neutrophils. Compared to N2, the N1 neutrophils exhibited: i) higher levels of ROS and oxidative burst, ii) increased activity of MPO and MMP-9, and iii) enhanced chemotactic response. N1 neutrophils were also characterized by elevated expression of NADPH oxidase subunits, as well as activation of the signaling molecules ERK and the p65 subunit of NF-kB. Moreover, we found that the S100A9 alarmin promotes the chemotactic and enzymatic activity of N1 neutrophils. S100A9 inhibition with a specific small-molecule blocker, reduced CCL2, CCL3 and CCL5 chemokine expression and decreased MPO and MMP-9 activity, by interfering with the NF-kB signaling pathway. Together, these findings reveal that N1 neutrophils are pro-inflammatory effectors of the innate immune response. Pharmacological blockade of S100A9 dampens the function of the pro-inflammatory N1 phenotype, promoting the alarmin as a novel target for therapeutic intervention in inflammatory diseases.
Collapse
Affiliation(s)
- Andreea C Mihaila
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Letitia Ciortan
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Razvan D Macarie
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Mihaela Vadana
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Sergiu Cecoltan
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Mihai Bogdan Preda
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Ariana Hudita
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Ana-Maria Gan
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Gabriel Jakobsson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Monica M Tucureanu
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Elena Barbu
- Departament of Cardiology, Elias Emergency Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Serban Balanescu
- Departament of Cardiology, Elias Emergency Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Maya Simionescu
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Alexandru Schiopu
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden.,Department of Pathophysiology, University of Medicine, Pharmacy, Sciences and Technology of Targu-Mures, Targu-Mures, Romania
| | - Elena Butoi
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| |
Collapse
|
57
|
Rodrigues RM, He Y, Hwang S, Bertola A, Mackowiak B, Ahmed YA, Seo W, Ma J, Wang X, Park SH, Guan Y, Fu Y, Vanhaecke T, Feng D, Gao B. E-Selectin-Dependent Inflammation and Lipolysis in Adipose Tissue Exacerbate Steatosis-to-NASH Progression via S100A8/9. Cell Mol Gastroenterol Hepatol 2021; 13:151-171. [PMID: 34390865 PMCID: PMC8593619 DOI: 10.1016/j.jcmgh.2021.08.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/04/2021] [Accepted: 08/04/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Nonalcoholic steatohepatitis (NASH) is a leading cause of chronic liver disease, characterized by steatosis and hallmark liver neutrophil infiltration. NASH also is associated with adipose tissue inflammation, but the role of adipose tissue inflammation in NASH pathogenesis remains obscure. The aim of this study was to investigate the interplay between neutrophil recruitment in adipose tissue and the progression of NASH. METHODS A mouse model of NASH was obtained by high-fat diet (HFD) feeding plus adenovirus-Cxcl1 overexpression (HFD+AdCxcl1). Genetic deletion of E-selectin (Sele) and treatment with an S100A9 inhibitor (Paquinimod) were investigated using this model. RESULTS By analyzing transcriptomic data sets of adipose tissue from NASH patients, we found that E-selectin, a key adhesion molecule for neutrophils, is the highest up-regulated gene among neutrophil recruitment-related factors in adipose tissue of NASH patients compared with those in patients with simple steatosis. A marked up-regulation of Sele in adipose tissue also was observed in HFD+AdCxcl1 mice. The HFD+AdCxcl1-induced NASH phenotype was ameliorated in Sele knockout mice and was accompanied by reduced lipolysis and inflammation in adipose tissue, which resulted in decreased serum free fatty acids and proinflammatory adipokines. S100A8/A9, a major proinflammatory protein secreted by neutrophils, was highly increased in adipose tissue of HFD+AdCxcl1 mice. This increase was blunted in the Sele knockout mice. Therapeutically, treatment with the S100A9 inhibitor Paquinimod reduced lipolysis, inflammation, and adipokine production, ameliorating the NASH phenotype in mice. CONCLUSIONS E-selectin plays an important role in inducing neutrophil recruitment in adipose tissue, which subsequently promotes inflammation and lipolysis via the production of S100A8/A9, thereby exacerbating the steatosis-to-NASH progression. Targeting adipose tissue inflammation therefore may represent a potential novel therapy for treatment of NASH.
Collapse
Affiliation(s)
- Robim M. Rodrigues
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland,Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yong He
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Seonghwan Hwang
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Adeline Bertola
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Bryan Mackowiak
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Yeni Ait Ahmed
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Wonhyo Seo
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Jing Ma
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Xiaolin Wang
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Seol Hee Park
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Yukun Guan
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Yaojie Fu
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Tamara Vanhaecke
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland,Correspondence Address correspondence to: Bin Gao, MD, PhD, Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Bethesda, Maryland 20892. fax: (301) 480-0257.
| |
Collapse
|
58
|
Matsunaga Y, Hashimoto Y, Ishiko A. Stratum corneum levels of calprotectin proteins S100A8/A9 correlate with disease activity in psoriasis patients. J Dermatol 2021; 48:1518-1525. [PMID: 34165193 DOI: 10.1111/1346-8138.16032] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 05/08/2021] [Accepted: 05/28/2021] [Indexed: 01/09/2023]
Abstract
Psoriasis is an intractable inflammatory skin disorder characterized by scaly erythema and plaques. The Psoriasis Area and Severity Index (PASI) is widely used to score disease severity, but evaluation is subjective, and an objective biomarker would be useful. The stratum corneum (SC), which can be non-invasively harvested, may reflect psoriasis-associated changes in epidermal keratinocytes, such as the upregulation of the calprotectin proteins S100A8 and S100A9. The aim of this study was to examine the availability of S100A8/A9 protein levels in SC as a biomarker of psoriasis disease activity. Fifty-three patients with psoriasis, 30 with psoriasis vulgaris (PsV), and 23 with psoriatic arthritis (PsA) participated. SC cells from lesional and non-lesional skin were collected by tape-stripping. S100A8/A9 levels in serum and in SC were quantified by enzyme-linked immunosorbent assay and compared with PASI score before and after treatment initiation or switching. Atopic dermatitis (AD) patients and disease-free individuals were used as controls. Expression of S100A8/A9 in SC of lesional skin of psoriasis patients was significantly higher than in non-lesional skin or AD skin. There was no significant difference of SC S100A8/A9 levels between PsV and PsA patients. The S100A8/A9 levels in SC of psoriasis patients were significantly positively correlated with the PASI score. When patients' skin lesions cleared (PASI clear) in response to treatment, expression of S100A8/A9 in SC was no longer detectable. S100A8/A9 protein levels in SC may be available as an objective, non-invasive biomarker of psoriasis activity to complement PASI scoring.
Collapse
Affiliation(s)
- Yukiko Matsunaga
- Department of Dermatology, Toho University Graduate School of Medicine, Tokyo, Japan
| | - Yuki Hashimoto
- Department of Dermatology, Toho University School of Medicine, Tokyo, Japan
| | - Akira Ishiko
- Department of Dermatology, Toho University School of Medicine, Tokyo, Japan
| |
Collapse
|
59
|
Singh H, Rai V, Agrawal DK. Discerning the promising binding sites of S100/calgranulins and their therapeutic potential in atherosclerosis. Expert Opin Ther Pat 2021; 31:1045-1057. [PMID: 34056993 DOI: 10.1080/13543776.2021.1937122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Atherosclerosis is a chronic inflammatory disease in which the members of S100 family proteins (calgranulins) bind with their receptors, particularly receptor for advanced glycation end products (RAGE) and toll-like receptor-4 (TLR-4) and play a key role in the pathogenesis and progression of disease. Thus, these proteins could be considered as potential biomarkers and therapeutic targets in the treatment of atherosclerotic inflammation. AREAS COVERED This review summarizes the pathology of S100A8, S100A9, and S100A12 in the development of atherosclerosis and reveals key structural features of these proteins which are potentially critical in their pathological effects. This article focuses on the translational significance of antagonizing these proteins by using small molecules in patent literature, clinical and preclinical studies and also discusses future approaches that could be employed to block these proteins in the treatment of atherosclerosis. EXPERT OPINION Based on the critical role of S100/calgranulins in the regulation of atherosclerosis, these proteins are potential targets to develop better therapeutic options in the treatment of inflammatory diseases. However, further research is still needed to clarify their exact molecular mechanism by analyzing their detailed structural features that can expedite future research to develop novel therapeutics against these proteins to treat atherosclerotic inflammation.
Collapse
Affiliation(s)
- Harbinder Singh
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Vikrant Rai
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|
60
|
Yang Y, Shen L, Xu M, Chen L, Lu W, Wang W. Serum calprotectin as a prognostic predictor in severe traumatic brain injury. Clin Chim Acta 2021; 520:101-107. [PMID: 34102135 DOI: 10.1016/j.cca.2021.06.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/24/2021] [Accepted: 06/04/2021] [Indexed: 01/13/2023]
Abstract
BACKGROUND Calprotectin plays an important role during inflammation. We intended to explore the prognostic value of serum calprotectin levels in patients with severe traumatic brain injury (sTBI). METHODS In this prospective cohort study of 149 sTBI patients, we determined the relationship between serum calprotectin levels and 90-day overall survival plus poor outcome (Glasgow outcome scale score of 1-3) after sTBI, and analyzed its associations with Rotterdam computerized tomography (CT) scores, Glasgow coma scale (GCS) scores and two markers of inflammatory reaction including serum C-reactive protein levels and blood leucocyte count. RESULTS Serum calprotectin levels were significantly correlated with Rotterdam CT scores, GCS scores, serum C-reactive protein levels and blood leucocyte count. Patients with poor outcome at 90 days displayed higher serum calprotectin levels than the other remainders. Serum calprotectin appeared as an independent predictor for 90-day overall survival and poor outcome. Under receiver operating characteristic curve, serum calprotectin levels exhibited an efficient discrimination capacity for 90-day poor outcome. CONCLUSIONS Serum calprotectin levels are significantly correlated with inflammation, trauma severity and poor outcome at 90 days in sTBI patients, suggesting that serum calprotectin may be a biomarker for providing complementary prognostic information to identify patients at risk of poor outcome after sTBI.
Collapse
Affiliation(s)
- Yan Yang
- Department of Critical Care Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, 215300, China
| | - Likui Shen
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou 266021, China
| | - Min Xu
- Department of Neurosurgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan 215300, Jiangsu Province, China
| | - Long Chen
- Department of Neurosurgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan 215300, Jiangsu Province, China
| | - Wei Lu
- Department of Neurosurgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan 215300, Jiangsu Province, China
| | - Wenhua Wang
- Department of Neurosurgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan 215300, Jiangsu Province, China.
| |
Collapse
|
61
|
Meng L, Tang Q, Zhao J, Wang Z, Wei L, Wei Q, Yin L, Luo S, Song J. S100A9 Derived From Myeloma Associated Myeloid Cells Promotes TNFSF13B/TNFRSF13B-Dependent Proliferation and Survival of Myeloma Cells. Front Oncol 2021; 11:691705. [PMID: 34150664 PMCID: PMC8210673 DOI: 10.3389/fonc.2021.691705] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/10/2021] [Indexed: 01/01/2023] Open
Abstract
Multiple myeloma (MM) is a lethal hematological malignancy characterized by abundant myeloid cells in the microenvironment that fuel tumor progression. But the mechanism by which myeloid cells support myeloma cells has not been fully explored. We aimed to examine their effect on bone marrow cells of MM patients by scRNA-seq transcriptome analysis and reveal a high-resolution gene profile of myeloma cells and myeloma-associated myeloid cells. Based on correlation analysis of integrated scRNA-seq and bulk RNA-seq datasets from patients, we confirmed that myeloid-derived S100A9 was involved in TNFSF13B-dependent myeloma cell proliferation and survival. In the animal experiments, S100A9 was found to be critical for MM cell proliferation and survival via TNFSF13B production by myeloid cells, neutrophils, and macrophages. In-vitro analysis of patient primary myeloma cells further demonstrated that enhanced TNFSF13B signaling triggered the canonical NF-κB pathway to boost tumor cell proliferation. All these results suggest that myeloid-derived S100A9 is required for TNFSF13B/TNFRSF13B-dependent cell-fate specification, which provides fresh insights into MM progression.
Collapse
Affiliation(s)
- Lingzhang Meng
- Department of Radiation Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Center for Systemic Inflammation Research (CSIR), School of Preclinical Medicine, Youjiang Medical University for Nationalities, Baise, China
| | - Qiang Tang
- Department of Burn Plastic and Wound Repair Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Jingjie Zhao
- Life Science and Clinical Research Center, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Zechen Wang
- Center for Systemic Inflammation Research (CSIR), School of Preclinical Medicine, Youjiang Medical University for Nationalities, Baise, China
| | - Liuzhi Wei
- Center for Systemic Inflammation Research (CSIR), School of Preclinical Medicine, Youjiang Medical University for Nationalities, Baise, China.,College of Pharmacy, Youjiang Medical University for Nationalities, Baise, China
| | - Qiuju Wei
- Center for Systemic Inflammation Research (CSIR), School of Preclinical Medicine, Youjiang Medical University for Nationalities, Baise, China.,College of Pharmacy, Youjiang Medical University for Nationalities, Baise, China
| | - Lianfei Yin
- School of Imaging, Youjiang Medical University for Nationalities, Baise, China
| | - Shiguan Luo
- Department of Cardiovascular Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Jian Song
- Department of Radiation Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Center for Systemic Inflammation Research (CSIR), School of Preclinical Medicine, Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
62
|
Fetz AE, Bowlin GL. Neutrophil Extracellular Traps: Inflammation and Biomaterial Preconditioning for Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:437-450. [PMID: 33736452 DOI: 10.1089/ten.teb.2021.0013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tissue injury initiates a tissue repair program, characterized by acute inflammation and recruitment of immune cells, dominated by neutrophils. Neutrophils prevent infection in the injured tissue through multiple effector functions, including the production of reactive oxygen species, the release of granules, the phagocytosis of invaders, and the extrusion of neutrophil extracellular traps (NETs). However, these canonical protective mechanisms can also have detrimental effects both in the context of infection and in response to sterile injuries. Of particular interest to biomaterials and tissue engineering is the release of NETs, which are extracellular structures composed of decondensed chromatin and various toxic nuclear and granular components. These structures and their dysregulated release can cause collateral tissue damage, uncontrolled inflammation, and fibrosis and prevent the neutrophil from exerting its prohealing functions. This review discusses our knowledge of NETs, including their composition and morphology, signaling pathways, inhibitors, and contribution to inflammatory pathologies, as well as their role in the resolution of inflammation. In addition, we summarize what is known about the release of NETs as a preconditioning event in the response to biomaterials and highlight future considerations to target the neutrophil response and enhance biomaterial-guided tissue repair and regeneration. Impact statement Neutrophil extracellular trap (NET) release is an active process programmed into the neutrophil's molecular machinery to prevent infection. However, the release of NETs on biomaterials appears to be a significant preconditioning event that influences the potential for tissue healing with largely detrimental consequences. Given their contribution to inflammatory pathologies, this review highlights the role of NETs in the response to biomaterials. Together, the studies discussed in this review suggest that biomaterials should be designed to regulate NET release to avoid maladaptive immune responses and improve the therapeutic potential of tissue-engineered biomaterials and their applications in the clinical setting.
Collapse
Affiliation(s)
- Allison E Fetz
- Department of Biomedical Engineering, University of Memphis, Memphis, Tennessee, USA
| | - Gary L Bowlin
- Department of Biomedical Engineering, University of Memphis, Memphis, Tennessee, USA
| |
Collapse
|
63
|
Zhang C, Qin L, Sun B, Wu Y, Zhong F, Wu L, Liu T. Transcriptome analysis of the effect of diosgenin on autoimmune thyroiditis in a rat model. Sci Rep 2021; 11:6401. [PMID: 33737640 PMCID: PMC7973441 DOI: 10.1038/s41598-021-85822-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/04/2021] [Indexed: 01/11/2023] Open
Abstract
In a mouse model of Graves' disease (GD), diosgenin has been shown to have a therapeutic effect on GD by alleviating goitre. However, research on the effect of diosgenin on autoimmune thyroiditis (AIT) is lacking. In this study, transcriptomics was used to comprehensively analyse the protective effect of diosgenin against AIT in rats and the possible mechanism. The results showed that in the diosgenin-intervention group, compared to the model group, the expression of serum triiodothyronine, thyroxine, free triiodothyronine, and free thyroxine was decreased and that of thyroid-stimulating hormone was increased; these changes were accompanied by the downregulation of thyroglobulin, TSH receptor antibody and thyroid peroxidase expression in serum. Furthermore, transcriptome detection, RT-qPCR and immunohistochemistry verification revealed that in thyroid tissue, the relative mRNA and protein expression of cyclic adenosine 3',5'-monophosphate (cAMP), protein kinase A (PKA) and cAMP response element-binding protein (Creb) were increased and the mRNA expression of S100 calcium-binding protein A9 (S100A9) was decreased in the diosgenin groups. In summary, diosgenin alleviates the development of AIT, possibly via the activation of the cAMP/PKA/Creb pathway and downregulation of S100A9 gene expression.
Collapse
Affiliation(s)
- Chengfei Zhang
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Lingling Qin
- Technology Department, Beijing University of Chinese Medicine, Beijing, China
| | - Boju Sun
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - You Wu
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Fengying Zhong
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Lili Wu
- Key Laboratory of TCM Health Cultivation of Beijing, Beijing University of Chinese Medicine, Beijing, China
| | - Tonghua Liu
- Key Laboratory of TCM Health Cultivation of Beijing, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
64
|
Hwang S, Yun H, Moon S, Cho YE, Gao B. Role of Neutrophils in the Pathogenesis of Nonalcoholic Steatohepatitis. Front Endocrinol (Lausanne) 2021; 12:751802. [PMID: 34707573 PMCID: PMC8542869 DOI: 10.3389/fendo.2021.751802] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/23/2021] [Indexed: 12/18/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) includes a spectrum of liver disorders, from fatty liver to nonalcoholic steatohepatitis (NASH), cirrhosis, and hepatocellular carcinoma. Compared with fatty liver, NASH is characterized by increased liver injury and inflammation, in which liver-infiltrating immune cells, with neutrophil infiltration as a hallmark of NASH, play a critical role in promoting the progression of fatty liver to NASH. Neutrophils are the first responders to injury and infection in various tissues, establishing the first line of defense through multiple mechanisms such as phagocytosis, cytokine secretion, reactive oxygen species production, and neutrophil extracellular trap formation; however, their roles in the pathogenesis of NASH remain obscure. The current review summarizes the roles of neutrophils that facilitate the progression of fatty liver to NASH and their involvement in inflammation resolution during NASH pathogenesis. The notion that neutrophils are potential therapeutic targets for the treatment of NASH is also discussed.
Collapse
Affiliation(s)
- Seonghwan Hwang
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, South Korea
| | - Hwayoung Yun
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, South Korea
| | - Sungwon Moon
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, South Korea
| | - Ye Eun Cho
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, South Korea
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
65
|
Cai Z, Xie Q, Hu T, Yao Q, Zhao J, Wu Q, Tang Q. S100A8/A9 in Myocardial Infarction: A Promising Biomarker and Therapeutic Target. Front Cell Dev Biol 2020; 8:603902. [PMID: 33282877 PMCID: PMC7688918 DOI: 10.3389/fcell.2020.603902] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/21/2020] [Indexed: 12/21/2022] Open
Abstract
Myocardial infarction (MI), the main cause of cardiovascular-related deaths worldwide, has long been a hot topic because of its threat to public health. S100A8/A9 has recently attracted an increasing amount of interest as a crucial alarmin that regulates the pathogenesis of cardiovascular disease after its release from myeloid cells. However, the role of S100A8/A9 in the etiology of MI is not well understood. Here, we elaborate on the critical roles and potential mechanisms of S100A8/A9 driving the pathogenesis of MI. First, cellular source of S100A8/A9 in infarcted heart is discussed. Then we highlight the effect of S100A8/A9 heterodimer in the early inflammatory period and the late reparative period of MI as well as myocardial ischemia/reperfusion (I/R) injury. Moreover, the predictive value of S100A8/A9 for the risk of recurrence of cardiovascular events is elucidated. Therefore, this review focuses on the molecular mechanisms of S100A8/A9 in MI pathogenesis to provide a promising biomarker and therapeutic target for MI.
Collapse
Affiliation(s)
- ZhuLan Cai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Qingwen Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Tongtong Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Qi Yao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jinhua Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Qingqing Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| |
Collapse
|
66
|
Lee YG, Hong J, Lee PH, Lee J, Park SW, Kim D, Jang AS. Serum Calprotectin Is a Potential Marker in Patients with Asthma. J Korean Med Sci 2020; 35:e362. [PMID: 33169556 PMCID: PMC7653171 DOI: 10.3346/jkms.2020.35.e362] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Calprotectin is the major cytosolic protein in neutrophil granulocytes. Although asthma is known to cause eosinophilic inflammation, some patients with asthma have non-eosinophilic inflammation, which is characterized by local neutrophilic inflammation. The aim of this study was to assess calprotectin expression levels in a mouse model of asthma, and to observe the relationship of serum calprotectin level and clinical variables in patients with asthma. METHODS Mice were sensitized and challenged with 10 μg and 20 μg of Aspergillus fumigatus, respectively; mice treated with saline were used as a control. The levels of calprotectin were determined using enzyme-linked immunosorbent assay, immunoblotting, and immunohistochemical analysis. The serum levels of calprotectin were also assessed in patients with asthma. The relationship between calprotectin and clinicopathological characteristics was determined. RESULTS Calprotectin, S100A8, and S100A9 expression was elevated in the mouse lungs, calprotectin levels were higher in the serum of patients with asthma (n = 33) compared with those of healthy individuals (n = 28). Calprotectin levels correlated with forced expiratory volume in one second/forced vital capacity (r = -0.215, P = 0.043), smoke amount (r = 0.413, P = 0.017), body mass index (r = -0.445, P = 0.000), and blood neutrophil percentage (r = 0.300, P = 0.004) in patients with asthma. CONCLUSION Our data suggest that calprotectin could potentially be used as a biomarker for asthma.
Collapse
Affiliation(s)
- Yun Gi Lee
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Jisu Hong
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Pureun Haneul Lee
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Junehyuk Lee
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Sung Woo Park
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - DoJin Kim
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - An Soo Jang
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea.
| |
Collapse
|
67
|
Sreejit G, Abdel Latif A, Murphy AJ, Nagareddy PR. Emerging roles of neutrophil-borne S100A8/A9 in cardiovascular inflammation. Pharmacol Res 2020; 161:105212. [PMID: 32991974 DOI: 10.1016/j.phrs.2020.105212] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/11/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
Elevated neutrophil count is associated with higher risk of major adverse cardiac events including myocardial infarction and early development of heart failure. Neutrophils contribute to cardiac damage through a number of mechanisms, including attraction of other immune cells and release of inflammatory mediators. Recently, a number of independent studies have reported a causal role for neutrophil-derived alarmins (i.e. S100A8/A9) in inducing inflammation and cardiac injury following myocardial infarction (MI). Furthermore, a positive correlation between serum S100A8/A9 levels and major adverse cardiac events (MACE) in MI patients was also observed implying that targeting neutrophils or their inflammatory cargo could be beneficial in reducing heart failure. However, contradictory to this idea, neutrophils and neutrophil-derived S100A8/A9 also seem to play a vital role in the resolution of inflammation. Thus, a better understanding of how neutrophils balance these seemingly contrasting functions would allow us to develop effective therapies that preserve the inflammation-resolving function while restricting the damage caused by inflammation. In this review, we specifically discuss the mechanisms behind neutrophil-derived S100A8/A9 in promoting inflammation and resolution in the context of MI. We also provide a perspective on how neutrophils could be potentially targeted to ameliorate cardiac inflammation and the ensuing damage.
Collapse
Affiliation(s)
- Gopalkrishna Sreejit
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ahmed Abdel Latif
- Division of Cardiovascular Medicine, Department of Medicine, University of Kentucky, Lexington, KY, USA
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, Division of Immunometabolism, Melbourne, Australia
| | - Prabhakara R Nagareddy
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
68
|
Gao H, Xu J, He L, Meng H, Hou J. Calprotectin levels in gingival crevicular fluid and serum of patients with chronic periodontitis and type 2 diabetes mellitus before and after initial periodontal therapy. J Periodontal Res 2020; 56:121-130. [PMID: 32936946 DOI: 10.1111/jre.12800] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/11/2020] [Accepted: 08/19/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND This study is aimed to compare the total amount of calprotectin in gingival crevicular fluid (GCF) and the concentration of calprotectin in serum among the patients with type 2 diabetes mellitus and chronic periodontitis (DM-P), the patients with chronic periodontitis (CP) and the healthy controls, as well as the variation of these indicators before and three months after the initial periodontal therapy for the DM-P patients. METHODS 35 patients with DM-P patients, 32 patients with CP patients, and 43 healthy controls were recruited. Calprotectin levels in serum and GCF, periodontal parameters, fasting blood glucose (FBG), and HbA1c were measured at baseline for all the groups and three months after the initial periodontal therapy for the DM-P patients. RESULTS At baseline, the calprotectin levels in GCF and serum were the highest in DM-P, followed by CP, and the lowest in healthy controls. GCF calprotectin was significantly and positively correlated with serum calprotectin and probing depth (PD), while serum calprotectin had a significant positive correlation with GCF calprotectin and HbA1c. Periodontal parameters, HbA1c, and serum and GCF calprotectin became significantly reduced after the initial periodontal treatment. The reduction of serum calprotectin was consistent with that of HbA1c, while the decrease of GCF calprotectin was in agreement with that of PD, attachment loss (AL), and bleeding on probing (BOP). CONCLUSIONS The levels of calprotectin in serum and GCF in the DM-P patients are significantly higher than those in CP patients and healthy controls, which significantly reduced 3 months after the initial periodontal therapy. Furthermore, it suggests diabetic patients might exhibit more pronounced inflammation periodontally and systemically.
Collapse
Affiliation(s)
- Hongyu Gao
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China.,Department of Periodontology, School and Hospital of Stomatology, Tianjin Medical University, Tianjin, China
| | - Jingling Xu
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China.,Department of Stomatology, Peking University International Hospital, Beijing, China
| | - Lu He
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Huanxin Meng
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Jianxia Hou
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
69
|
Liu Y, Zhu Y, Jia W, Sun D, Zhao L, Zhang C, Wang C, Lyu Q, Chen Y, Chen G, Bo Y, Xing Y. Association of the Total White Blood Cell, Neutrophils, and Monocytes Count With the Presence, Severity, and Types of Carotid Atherosclerotic Plaque. Front Med (Lausanne) 2020; 7:313. [PMID: 32793608 PMCID: PMC7385072 DOI: 10.3389/fmed.2020.00313] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/29/2020] [Indexed: 01/07/2023] Open
Abstract
Background: Previous studies have indicated that white blood cells (WBCs) might contribute to the development of atherosclerosis. However, the associations of WBCs and WBC subgroups with carotid atherosclerotic plaque (CAP) have not been compared. Methods: A cross-sectional study including 3,569 healthy Chinese adults was conducted between January 2016 and December 2018 in Zhengzhou, China, to explore the associations of WBC and WBC subtypes with the presence, severity, and types of CAPs. Fasting peripheral venous blood was collected for measurement of the total WBC and WBC subtype counts. The size, composition, and types of CAPs in the common carotid artery, the internal carotid artery, and the external carotid artery were measured bilaterally using B-mode ultrasound. Results: The total WBC, neutrophil, and monocyte counts showed significant associations with the presence of CAPs in men, but not in women, with the adjusted odds ratios (95% CI) in the highest (compared to the lowest) quartile 1.99 (1.33-2.97), 1.65 (1.10-2.47), and 2.17 (1.41-3.18) (P trend = 0.004, P trend = 0.004, and P trend < 0.001), respectively. The three leukocyte counts were also significantly associated with the severity of CAPs, as judged by the count of CAPs, maximal internal carotid plaque thickness, and the plaque score (all P < 0.01, P trend < 0.05). Compared with individuals without CAPs, those with echolucent plaques had significantly increased total WBC and neutrophil counts, whereas those with polytype plaques had a significantly increased monocyte count. Conclusion: WBC, neutrophil, and monocyte counts were significantly associated with the presence, severity, and types of CAPs in a healthy Chinese population.
Collapse
Affiliation(s)
- Yanhua Liu
- Department of Nutrition, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yongjian Zhu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenrui Jia
- Department of Physical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dan Sun
- Department of Physical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Zhao
- Department of Physical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chen Zhang
- Department of Physical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Cuicui Wang
- Department of Physical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Quanjun Lyu
- Department of Nutrition, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuming Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Gaiyun Chen
- Department of Nutrition, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yacong Bo
- Department of Nutrition, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yurong Xing
- Department of Physical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
70
|
Lorenzin M, Ometto F, Ortolan A, Felicetti M, Favero M, Doria A, Ramonda R. An update on serum biomarkers to assess axial spondyloarthritis and to guide treatment decision. Ther Adv Musculoskelet Dis 2020; 12:1759720X20934277. [PMID: 32636944 PMCID: PMC7315656 DOI: 10.1177/1759720x20934277] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/22/2020] [Indexed: 12/17/2022] Open
Abstract
Axial spondyloarthritis (axSpA) is a group of debilitating, chronic, rheumatic conditions characterized by inflammation and new bone formation, mainly involving the spine and the sacroiliac joints. The lack of biomarkers in axSpA is well known. Despite significant treatment advances in recent years thanks to the introduction of drugs with a new mode of action, such as new biologic and targeted synthetic disease-modifying antirheumatic drugs, no relevant improvement in the identification of disease biomarkers has been achieved. Common parameters, such as erythrocyte sedimentation rate and C-reactive protein, which are routinely used to measure systemic inflammation, are the sole markers available to date and are not adequate to assess disease activity in all patients. The aim of this study is to review the most promising serum biomarkers that may help treatment decision in axSpA via a proper assessment of disease activity and identification of negative prognostic factors.
Collapse
Affiliation(s)
- Mariagrazia Lorenzin
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Padova, Italy
| | - Francesca Ometto
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Padova, Italy
| | - Augusta Ortolan
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Padova, Italy
| | - Mara Felicetti
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Padova, Italy
| | - Marta Favero
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Padova, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Padova, Italy
| | - Roberta Ramonda
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Via Giustiniani 2, Padova, 35128, Italy
| |
Collapse
|
71
|
Mossel DM, Moganti K, Riabov V, Weiss C, Kopf S, Cordero J, Dobreva G, Rots MG, Klüter H, Harmsen MC, Kzhyshkowska J. Epigenetic Regulation of S100A9 and S100A12 Expression in Monocyte-Macrophage System in Hyperglycemic Conditions. Front Immunol 2020; 11:1071. [PMID: 32582175 PMCID: PMC7280556 DOI: 10.3389/fimmu.2020.01071] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
The number of diabetic patients in Europe and world-wide is growing. Diabetes confers a 2-fold higher risk for vascular disease. Lack of insulin production (Type 1 diabetes, T1D) or lack of insulin responsiveness (Type 2 diabetes, T2D) causes systemic metabolic changes such as hyperglycemia (HG) which contribute to the pathology of diabetes. Monocytes and macrophages are key innate immune cells that control inflammatory reactions associated with diabetic vascular complications. Inflammatory programming of macrophages is regulated and maintained by epigenetic mechanisms, in particular histone modifications. The aim of our study was to identify the epigenetic mechanisms involved in the hyperglycemia-mediated macrophage activation. Using Affymetrix microarray profiling and RT-qPCR we identified that hyperglycemia increased the expression of S100A9 and S100A12 in primary human macrophages. Expression of S100A12 was sustained after glucose levels were normalized. Glucose augmented the response of macrophages to Toll-like receptor (TLR)-ligands Palmatic acid (PA) and Lipopolysaccharide (LPS) i.e., pro-inflammatory stimulation. The abundance of activating histone Histone 3 Lysine 4 methylation marks (H3K4me1, H3K4me3) and general acetylation on histone 3 (AceH3) with the promoters of these genes was analyzed by chromatin immunoprecipitation. Hyperglycemia increased acetylation of histones bound to the promoters of S100A9 and S100A12 in M1 macrophages. In contrast, hyperglycemia caused a reduction in total H3 which correlated with the increased expression of both S100 genes. The inhibition of histone methyltransferases SET domain-containing protein (SET)7/9 and SET and MYND domain-containing protein (SMYD)3 showed that these specifically regulated S100A12 expression. We conclude that hyperglycemia upregulates expression of S100A9, S100A12 via epigenetic regulation and induces an activating histone code on the respective gene promoters in M1 macrophages. Mechanistically, this regulation relies on action of histone methyltransferases SMYD3 and SET7/9. The results define an important role for epigenetic regulation in macrophage mediated inflammation in diabetic conditions.
Collapse
Affiliation(s)
- Dieuwertje M Mossel
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany
| | - Kondaiah Moganti
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany.,Department of Dermatology, University of Münster, Münster, Germany
| | - Vladimir Riabov
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany
| | - Christel Weiss
- Department of Medical Statistics, Biomathematics and Information Processing, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan Kopf
- Department of Medicine I: Endocrinology and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
| | - Julio Cordero
- Anatomy and Developmental Biology, CBTM, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gergana Dobreva
- Anatomy and Developmental Biology, CBTM, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Marianne G Rots
- Department Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Harald Klüter
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany.,German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
| | - Martin C Harmsen
- Department Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Julia Kzhyshkowska
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany.,German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
| |
Collapse
|
72
|
Zuurbier CJ, Abbate A, Cabrera-Fuentes HA, Cohen MV, Collino M, De Kleijn DPV, Downey JM, Pagliaro P, Preissner KT, Takahashi M, Davidson SM. Innate immunity as a target for acute cardioprotection. Cardiovasc Res 2020; 115:1131-1142. [PMID: 30576455 DOI: 10.1093/cvr/cvy304] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/07/2018] [Accepted: 12/14/2018] [Indexed: 12/18/2022] Open
Abstract
Acute obstruction of a coronary artery causes myocardial ischaemia and if prolonged, may result in an ST-segment elevation myocardial infarction (STEMI). First-line treatment involves rapid reperfusion. However, a highly dynamic and co-ordinated inflammatory response is rapidly mounted to repair and remove the injured cells which, paradoxically, can further exacerbate myocardial injury. Furthermore, although cardiac remodelling may initially preserve some function to the heart, it can lead over time to adverse remodelling and eventually heart failure. Since the size of the infarct corresponds to the subsequent risk of developing heart failure, it is important to find ways to limit initial infarct development. In this review, we focus on the role of the innate immune system in the acute response to ischaemia-reperfusion (IR) and specifically its contribution to cell death and myocardial infarction. Numerous danger-associated molecular patterns are released from dying cells in the myocardium, which can stimulate pattern recognition receptors including toll like receptors and NOD-like receptors (NLRs) in resident cardiac and immune cells. Activation of the NLRP3 inflammasome, caspase 1, and pyroptosis may ensue, particularly when the myocardium has been previously aggravated by the presence of comorbidities. Evidence will be discussed that suggests agents targeting innate immunity may be a promising means of protecting the hearts of STEMI patients against acute IR injury. However, the dosing and timing of such agents should be carefully determined because innate immunity pathways may also be involved in cardioprotection. This article is part of a Cardiovascular Research Spotlight Issue entitled 'Cardioprotection Beyond the Cardiomyocyte', and emerged as part of the discussions of the European Union (EU)-CARDIOPROTECTION Cooperation in Science and Technology (COST) Action, CA16225.
Collapse
Affiliation(s)
- Coert J Zuurbier
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands
| | - Antonio Abbate
- VCU Pauley Heart Center and Wright Center for Clinical and Translational Research, Richmond, VA, USA
| | - Hector A Cabrera-Fuentes
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore.,Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Centro de Biotecnología-FEMSA, Monterrey, Nuevo León, México.,Department of Microbiology, Kazan Federal University, Kazan, Russian Federation.,Institute of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Michael V Cohen
- Department of Medicine, University of South Alabama College of Medicine, Mobile, AL, USA.,Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL, USA
| | - Massimo Collino
- Department of Drug Science and Technology, University of Turin, Torino, Italy
| | - Dominique P V De Kleijn
- Department of Vascular Surgery, UMC Utrecht, Utrecht University, Utrecht, the Netherlands.,Netherlands Heart Institute, Utrecht, the Netherlands
| | - James M Downey
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL, USA
| | - Pasquale Pagliaro
- Department of Biological and Clinical Sciences, University of Turin, Torino, Italy.,National Institute for Cardiovascular Research, Bologna, Italy
| | - Klaus T Preissner
- Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Masafumi Takahashi
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, UK
| |
Collapse
|
73
|
Marinković G, Koenis DS, de Camp L, Jablonowski R, Graber N, de Waard V, de Vries CJ, Goncalves I, Nilsson J, Jovinge S, Schiopu A. S100A9 Links Inflammation and Repair in Myocardial Infarction. Circ Res 2020; 127:664-676. [PMID: 32434457 DOI: 10.1161/circresaha.120.315865] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
RATIONALE The alarmin S100A9 has been identified as a potential therapeutic target in myocardial infarction. Short-term S100A9 blockade during the inflammatory phase post-myocardial infarction inhibits systemic and cardiac inflammation and improves cardiac function long term. OBJECTIVE To evaluate the impact of S100A9 blockade on postischemic cardiac repair. METHODS AND RESULTS We assessed cardiac function, hematopoietic response, and myeloid phagocyte dynamics in WT (wild type) C57BL/6 mice with permanent coronary artery ligation, treated with the specific S100A9 blocker ABR-238901 for 7 or 21 days. In contrast to the beneficial effects of short-term therapy, extended S100A9 blockade led to progressive deterioration of cardiac function and left ventricle dilation. The treatment reduced the proliferation of Lin-Sca-1+c-Kit+ hematopoietic stem and progenitor cells in the bone marrow and the production of proreparatory CD150+CD48-CCR2+ hematopoietic stem cells. Monocyte trafficking from the spleen to the myocardium and subsequent phenotype switching to reparatory Ly6CloMerTKhi macrophages was also impaired, leading to inefficient efferocytosis, accumulation of apoptotic cardiomyocytes, and a larger myocardial scar. The transcription factor Nur77 (Nr4a1 [nuclear receptor subfamily 4 group A member 1]) mediates the transition from inflammatory Ly6Chi monocytes to reparatory Ly6Clo macrophages. S100A9 upregulated the levels and activity of Nur77 in monocytes and macrophages in vitro and in Ly6Chi/int monocytes in vivo, and S100A9 blockade antagonized these effects. Finally, the presence of reparatory macrophages in the myocardium was also impaired in S100A9-/- mice with permanent myocardial ischemia, leading to depressed cardiac function long term. CONCLUSIONS We show that S100A9 plays an important role in both the inflammatory and the reparatory immune responses to myocardial infarction. Long-term S100A9 blockade negatively impacts cardiac recovery and counterbalances the beneficial effects of short-term therapy. These results define a therapeutic window targeting the inflammatory phase for optimal effects of S100A9 blockade as potential immunomodulatory treatment in acute myocardial infarction.
Collapse
Affiliation(s)
- Goran Marinković
- From the Department of Clinical Sciences Malmö, Lund University, Sweden (G.M., I.G., J.N., A.S.)
| | - Duco Steven Koenis
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, the Netherlands (D.S.K., V.d.W., C.J.d.V.)
| | - Lisa de Camp
- DeVos Cardiovascular Research Program, Van Andel Institute, Grand Rapids, MI (L.d.C., N.G., S.J.)
| | | | - Naomi Graber
- DeVos Cardiovascular Research Program, Van Andel Institute, Grand Rapids, MI (L.d.C., N.G., S.J.)
| | - Vivian de Waard
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, the Netherlands (D.S.K., V.d.W., C.J.d.V.)
| | - Carlie Jacoba de Vries
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, the Netherlands (D.S.K., V.d.W., C.J.d.V.)
| | - Isabel Goncalves
- From the Department of Clinical Sciences Malmö, Lund University, Sweden (G.M., I.G., J.N., A.S.).,Department of Cardiology, Skane University Hospital, Sweden (I.G.)
| | - Jan Nilsson
- From the Department of Clinical Sciences Malmö, Lund University, Sweden (G.M., I.G., J.N., A.S.)
| | - Stefan Jovinge
- DeVos Cardiovascular Research Program, Van Andel Institute, Grand Rapids, MI (L.d.C., N.G., S.J.).,DeVos Cardiovascular Research Program, Fredrik Meijer Heart and Vascular Institute, Spectrum Health, Grand Rapids, MI (S.J.).,Cardiovascular Institute, Stanford University, CA (S.J.)
| | - Alexandru Schiopu
- From the Department of Clinical Sciences Malmö, Lund University, Sweden (G.M., I.G., J.N., A.S.).,University of Medicine, Pharmacy, Sciences and Technology of Targu-Mures, Romania (A.S.).,Department of Internal Medicine, Skane University Hospital, Sweden (A.S.)
| |
Collapse
|
74
|
Steffens S, Van Linthout S, Sluijter JPG, Tocchetti CG, Thum T, Madonna R. Stimulating pro-reparative immune responses to prevent adverse cardiac remodelling: consensus document from the joint 2019 meeting of the ESC Working Groups of cellular biology of the heart and myocardial function. Cardiovasc Res 2020; 116:1850-1862. [DOI: 10.1093/cvr/cvaa137] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/31/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022] Open
Abstract
Abstract
Cardiac injury may have multiple causes, including ischaemic, non-ischaemic, autoimmune, and infectious triggers. Independent of the underlying pathophysiology, cardiac tissue damage induces an inflammatory response to initiate repair processes. Immune cells are recruited to the heart to remove dead cardiomyocytes, which is essential for cardiac healing. Insufficient clearance of dying cardiomyocytes after myocardial infarction (MI) has been shown to promote unfavourable cardiac remodelling, which may result in heart failure (HF). Although immune cells are integral key players of cardiac healing, an unbalanced or unresolved immune reaction aggravates tissue damage that triggers maladaptive remodelling and HF. Neutrophils and macrophages are involved in both, inflammatory as well as reparative processes. Stimulating the resolution of cardiac inflammation seems to be an attractive therapeutic strategy to prevent adverse remodelling. Along with numerous experimental studies, the promising outcomes from recent clinical trials testing canakinumab or colchicine in patients with MI are boosting the interest in novel therapies targeting inflammation in cardiovascular disease patients. The aim of this review is to discuss recent experimental studies that provide new insights into the signalling pathways and local regulators within the cardiac microenvironment promoting the resolution of inflammation and tissue regeneration. We will cover ischaemia- and non-ischaemic-induced as well as infection-related cardiac remodelling and address potential targets to prevent adverse cardiac remodelling.
Collapse
Affiliation(s)
- Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité, University Medicine Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Berlin, Germany
| | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Regenerative Medicine Center, Circulatory Health Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences and Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy
| | - Thomas Thum
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Via Paradisa, Pisa 56124, Italy
| |
Collapse
|
75
|
Wang C, Kou Y, Han Y, Li X. Early Serum Calprotectin (S100A8/A9) Predicts Delayed Cerebral Ischemia and Outcomes after Aneurysmal Subarachnoid Hemorrhage. J Stroke Cerebrovasc Dis 2020; 29:104770. [DOI: 10.1016/j.jstrokecerebrovasdis.2020.104770] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 01/17/2020] [Accepted: 02/14/2020] [Indexed: 12/20/2022] Open
|
76
|
Harman JL, Loes AN, Warren GD, Heaphy MC, Lampi KJ, Harms MJ. Evolution of multifunctionality through a pleiotropic substitution in the innate immune protein S100A9. eLife 2020; 9:e54100. [PMID: 32255429 PMCID: PMC7213983 DOI: 10.7554/elife.54100] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/03/2020] [Indexed: 12/16/2022] Open
Abstract
Multifunctional proteins are evolutionary puzzles: how do proteins evolve to satisfy multiple functional constraints? S100A9 is one such multifunctional protein. It potently amplifies inflammation via Toll-like receptor four and is antimicrobial as part of a heterocomplex with S100A8. These two functions are seemingly regulated by proteolysis: S100A9 is readily degraded, while S100A8/S100A9 is resistant. We take an evolutionary biochemical approach to show that S100A9 evolved both functions and lost proteolytic resistance from a weakly proinflammatory, proteolytically resistant amniote ancestor. We identify a historical substitution that has pleiotropic effects on S100A9 proinflammatory activity and proteolytic resistance but has little effect on S100A8/S100A9 antimicrobial activity. We thus propose that mammals evolved S100A8/S100A9 antimicrobial and S100A9 proinflammatory activities concomitantly with a proteolytic 'timer' to selectively regulate S100A9. This highlights how the same mutation can have pleiotropic effects on one functional state of a protein but not another, thus facilitating the evolution of multifunctionality.
Collapse
Affiliation(s)
- Joseph L Harman
- Department of Chemistry and Biochemistry, University of OregonEugeneUnited States
- Institute of Molecular Biology, University of OregonEugeneUnited States
| | - Andrea N Loes
- Department of Chemistry and Biochemistry, University of OregonEugeneUnited States
- Institute of Molecular Biology, University of OregonEugeneUnited States
| | - Gus D Warren
- Department of Chemistry and Biochemistry, University of OregonEugeneUnited States
- Institute of Molecular Biology, University of OregonEugeneUnited States
| | - Maureen C Heaphy
- Department of Chemistry and Biochemistry, University of OregonEugeneUnited States
- Institute of Molecular Biology, University of OregonEugeneUnited States
| | | | - Michael J Harms
- Department of Chemistry and Biochemistry, University of OregonEugeneUnited States
- Institute of Molecular Biology, University of OregonEugeneUnited States
| |
Collapse
|
77
|
Nieman DC, Groen AJ, Pugachev A, Simonson AJ, Polley K, James K, El-Khodor BF, Varadharaj S, Hernández-Armenta C. Proteomics-Based Detection of Immune Dysfunction in an Elite Adventure Athlete Trekking Across the Antarctica. Proteomes 2020; 8:proteomes8010004. [PMID: 32138228 PMCID: PMC7151708 DOI: 10.3390/proteomes8010004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 02/29/2020] [Accepted: 03/01/2020] [Indexed: 12/31/2022] Open
Abstract
Proteomics monitoring of an elite adventure athlete (age 33 years) was conducted over a 28-week period that culminated in the successful, solo, unassisted, and unsupported two month trek across the Antarctica (1500 km). Training distress was monitored weekly using a 19-item, validated training distress scale (TDS). Weekly dried blood spot (DBS) specimens were collected via fingerprick blood drops onto standard blood spot cards. DBS proteins were measured with nano-electrospray ionization liquid chromatography tandem mass spectrometry (nanoLC-MS/MS) in data-independent acquisition (DIA) mode, and 712 proteins were identified and quantified. The 28-week period was divided into time segments based on TDS scores, and a contrast analysis between weeks five and eight (low TDS) and between weeks 20 and 23 (high TDS, last month of Antarctica trek) showed that 31 proteins (n = 20 immune related) were upregulated and 35 (n = 17 immune related) were downregulated. Protein-protein interaction (PPI) networks supported a dichotomous immune response. Gene ontology (GO) biological process terms for the upregulated immune proteins showed an increase in regulation of the immune system process, especially inflammation, complement activation, and leukocyte mediated immunity. At the same time, GO terms for the downregulated immune-related proteins indicated a decrease in several aspects of the overall immune system process including neutrophil degranulation and the antimicrobial humoral response. These proteomics data support a dysfunctional immune response in an elite adventure athlete during a sustained period of mental and physical distress while trekking solo across the Antarctica.
Collapse
Affiliation(s)
- David C. Nieman
- North Carolina Research Campus, Appalachian State University, Kannapolis, NC 28081, USA;
- Correspondence: ; Tel.: +1-828-773-0056
| | - Arnoud J. Groen
- ProteiQ Biosciences GmbH, 10967 Berlin, Germany; (A.J.G.); (A.P.); (C.H.-A.)
| | - Artyom Pugachev
- ProteiQ Biosciences GmbH, 10967 Berlin, Germany; (A.J.G.); (A.P.); (C.H.-A.)
| | - Andrew J. Simonson
- North Carolina Research Campus, Appalachian State University, Kannapolis, NC 28081, USA;
| | - Kristine Polley
- Standard Process Nutrition Innovation, Kannapolis, NC 28081, USA; (K.P.); (K.J.); (B.F.E.-K.); (S.V.)
| | - Karma James
- Standard Process Nutrition Innovation, Kannapolis, NC 28081, USA; (K.P.); (K.J.); (B.F.E.-K.); (S.V.)
| | - Bassem F. El-Khodor
- Standard Process Nutrition Innovation, Kannapolis, NC 28081, USA; (K.P.); (K.J.); (B.F.E.-K.); (S.V.)
| | - Saradhadevi Varadharaj
- Standard Process Nutrition Innovation, Kannapolis, NC 28081, USA; (K.P.); (K.J.); (B.F.E.-K.); (S.V.)
| | | |
Collapse
|
78
|
Brusletto BS, Løberg EM, Hellerud BC, Goverud IL, Berg JP, Olstad OK, Gopinathan U, Brandtzaeg P, Øvstebø R. Extensive Changes in Transcriptomic "Fingerprints" and Immunological Cells in the Large Organs of Patients Dying of Acute Septic Shock and Multiple Organ Failure Caused by Neisseria meningitidis. Front Cell Infect Microbiol 2020; 10:42. [PMID: 32154187 PMCID: PMC7045056 DOI: 10.3389/fcimb.2020.00042] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/22/2020] [Indexed: 12/16/2022] Open
Abstract
Background: Patients developing meningococcal septic shock reveal levels of Neisseria meningitidis (106-108/mL) and endotoxin (101-103 EU/mL) in the circulation and organs, leading to acute cardiovascular, pulmonary and renal failure, coagulopathy and a high case fatality rate within 24 h. Objective: To investigate transcriptional profiles in heart, lungs, kidneys, liver, and spleen and immunostain key inflammatory cells and proteins in post mortem formalin-fixed, paraffin-embedded (FFPE) tissue samples from meningococcal septic shock patients. Patients and Methods: Total RNA was isolated from FFPE and fresh frozen (FF) tissue samples from five patients and two controls (acute non-infectious death). Differential expression of genes was detected using Affymetrix microarray analysis. Lung and heart tissue samples were immunostained for T-and B cells, macrophages, neutrophils and the inflammatory markers PAI-1 and MCP-1. Inflammatory mediators were quantified in lysates from FF tissues. Results: The transcriptional profiles showed a complex pattern of protein-coding and non-coding RNAs with significant regulation of pathways associated with organismal death, cell death and survival, leukocyte migration, cellular movement, proliferation of cells, cell-to-cell signaling, immune cell trafficking, and inflammatory responses in an organ-specific clustering manner. The canonical pathways including acute phase response-, EIF2-, TREM1-, IL-6-, HMBG1-, PPAR signaling, and LXR/RXR activation were associated with acute heart, pulmonary, and renal failure. Fewer genes were regulated in the liver and particularly in the spleen. The main upstream regulators were TNF, IL-1β, IL-6, RICTOR, miR-6739-3p, and CD3. Increased numbers of inflammatory cells (CD68+, MPO+, CD3+, and CD20+) were found in lungs and heart. PAI-1 inhibiting fibrinolysis and MCP-1 attracting leukocyte were found significantly present in the septic tissue samples compared to the controls. Conclusions: FFPE tissue samples can be suitable for gene expression studies as well as immunostaining of specific cells or molecules. The most pronounced gene expression patterns were found in the organs with highest levels of Neisseria meningitidis DNA. Thousands of protein-coding and non-coding RNA transcripts were altered in lungs, heart and kidneys. We identified specific biomarker panels both protein-coding and non-coding RNA transcripts, which differed from organ to organ. Involvement of many genes and pathways add up and the combined effect induce organ failure.
Collapse
Affiliation(s)
- Berit Sletbakk Brusletto
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Else Marit Løberg
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Pathology, Oslo University Hospital, Oslo, Norway
| | | | - Ingeborg Løstegaard Goverud
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Jens Petter Berg
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Unni Gopinathan
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Petter Brandtzaeg
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Pediatrics, Oslo University Hospital, Oslo, Norway
| | - Reidun Øvstebø
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
79
|
Funeshima N, Tanikawa N, Yaginuma H, Watanabe H, Iwata H, Kuwayama T, Hamano S, Shirasuna K. Adverse reproductive effects of S100A9 on bovine sperm and early embryonic development in vitro. PLoS One 2020; 15:e0227885. [PMID: 31945120 PMCID: PMC6964853 DOI: 10.1371/journal.pone.0227885] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 12/31/2019] [Indexed: 02/05/2023] Open
Abstract
The phenomenon of aging arises from multiple, complex interactions causing dysfunction in cells and organs. In particular, fertility drastically decreases with age. Previously, we have demonstrated that the functional characteristics of the bovine oviduct and uterus change with the age-dependent upregulation of inflammation and noted that S100A9 triggers inflammatory responses in oviduct epithelial cells. In the present study, we investigated the hypothesis that S100A9 affects reproductive events to aspect such as sperm function, fertilization, and the development of the embryo in cows. To investigate the effect of S100A9 on bovine sperm, we incubated sperms in vitro with S100A9 for 5 h and observed significantly decreased sperm motility and viability. During in vitro fertilization, S100A9 treatment for 5 h did not affect the rate of fertilization, time of first division of embryos, or embryo development to blastocyst stage. Treatment of 2-cell stage embryos with S100A9 for 5 h significantly reduced the proportion of cells undergoing normal division (4-8 cell embryos) and embryo development to the blastocyst stage. In experiment involving 24 h treatment of 2-cell embryos, the development of all embryos stopped at the 2-cell stage in the S100A9-treated group. In blastocyst-stage embryos, S100A9 treatment significantly stimulated the expression of endoplasmic reticulum (ER) and the mRNA expression of ER stress markers, and activated caspase-3 with subsequent nuclear fragmentation. Pre-treatment with an ER stress inhibitor significantly suppressed caspase-3 activation by the S100A9 treatment, suggesting that S100A9 induces blastocyst dysfunction by apoptosis (via caspase-3 activation) depending on ER stress. These results indicate that direct exposure to S100A9 exerted adverse effects on sperm function and embryo development. These findings suggest that excessive dose of S100A9 may have an adverse effect to the reproductive machinery by inducing inflammation and tissue dysfunction.
Collapse
Affiliation(s)
- Natsumi Funeshima
- Department of Animal Science, Tokyo University of Agriculture, Atsugi, Kanagawa, Japan
| | - Nao Tanikawa
- Department of Animal Science, Tokyo University of Agriculture, Atsugi, Kanagawa, Japan
| | - Hikari Yaginuma
- Animal Bio-Technology Center, Livestock Improvement Association of Japan Inc., Tokyo, Japan
| | - Hiroyuki Watanabe
- Department of Life and Food Sciences, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Hisataka Iwata
- Department of Animal Science, Tokyo University of Agriculture, Atsugi, Kanagawa, Japan
| | - Takehito Kuwayama
- Department of Animal Science, Tokyo University of Agriculture, Atsugi, Kanagawa, Japan
| | - Seizo Hamano
- Animal Bio-Technology Center, Livestock Improvement Association of Japan Inc., Tokyo, Japan
- Maebashi Institute of Animal Science, Livestock Improvement Association of Japan Inc., Gunma, Japan
| | - Koumei Shirasuna
- Department of Animal Science, Tokyo University of Agriculture, Atsugi, Kanagawa, Japan
| |
Collapse
|
80
|
Riuzzi F, Chiappalupi S, Arcuri C, Giambanco I, Sorci G, Donato R. S100 proteins in obesity: liaisons dangereuses. Cell Mol Life Sci 2020; 77:129-147. [PMID: 31363816 PMCID: PMC11104817 DOI: 10.1007/s00018-019-03257-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/19/2019] [Accepted: 07/24/2019] [Indexed: 02/07/2023]
Abstract
Obesity is an endemic pathophysiological condition and a comorbidity associated with hypercholesterolemia, hypertension, cardiovascular disease, type 2 diabetes mellitus, and cancer. The adipose tissue of obese subjects shows hypertrophic adipocytes, adipocyte hyperplasia, and chronic low-grade inflammation. S100 proteins are Ca2+-binding proteins exclusively expressed in vertebrates in a cell-specific manner. They have been implicated in the regulation of a variety of functions acting as intracellular Ca2+ sensors transducing the Ca2+ signal and extracellular factors affecting cellular activity via ligation of a battery of membrane receptors. Certain S100 proteins, namely S100A4, the S100A8/S100A9 heterodimer and S100B, have been implicated in the pathophysiology of obesity-promoting macrophage-based inflammation via toll-like receptor 4 and/or receptor for advanced glycation end-products ligation. Also, serum levels of S100A4, S100A8/S100A9, S100A12, and S100B correlate with insulin resistance/type 2 diabetes, metabolic risk score, and fat cell size. Yet, secreted S100B appears to exert neurotrophic effects on sympathetic fibers in brown adipose tissue contributing to the larger sympathetic innervation of this latter relative to white adipose tissue. In the present review we first briefly introduce S100 proteins and then critically examine their role(s) in adipose tissue and obesity.
Collapse
Affiliation(s)
- Francesca Riuzzi
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy
| | - Sara Chiappalupi
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy
| | - Cataldo Arcuri
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
| | - Ileana Giambanco
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
| | - Guglielmo Sorci
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy
- Centro Universitario di Ricerca sulla Genomica Funzionale, University of Perugia, 06132, Perugia, Italy
| | - Rosario Donato
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy.
| |
Collapse
|
81
|
Bach M, Moon J, Moore R, Pan T, Nelson JL, Lood C. A Neutrophil Activation Biomarker Panel in Prognosis and Monitoring of Patients With Rheumatoid Arthritis. Arthritis Rheumatol 2020; 72:47-56. [PMID: 31353807 PMCID: PMC6935396 DOI: 10.1002/art.41062] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/25/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Exaggerated neutrophil activation and formation of neutrophil extracellular traps (NETs) are linked to inflammation and autoimmunity, including rheumatoid arthritis (RA). However, whether NETs are present in the circulation of RA patients and contribute to inflammation and disease progression has not been carefully addressed. We undertook this study to assess markers of neutrophil activation and NET formation in plasma samples, investigating whether they add clinical value in improving the determination of prognosis and monitoring in RA patients. METHODS Markers of neutrophil activation (calprotectin) and cell death (NETs) were analyzed, using enzyme-linked immunosorbent assay, in serum and plasma obtained from patients in 3 cross-sectional RA cohorts and sex-matched healthy controls. A longitudinal inception cohort (n = 247), seen for a median follow-up of 8 years, was used for predictive analyses. RESULTS Markers of neutrophil activation and cell death were increased in RA patients compared to healthy individuals (P < 0.0001). Calprotectin levels correlated with the Clinical Disease Activity Index (r = 0.53, P < 0.0001) and could be used to distinguish between patients with disease in remission and those with active disease, an observation not seen when examining C-reactive protein levels. A biomarker panel consisting of anti-citrullinated protein antibody and calprotectin could predict erosive disease (odds ratio [OR] 7.5, P < 0.0001) and joint space narrowing (OR 4.9, P = 0.001). NET levels were associated with markers of inflammation (P = 0.0002). Furthermore, NETs and a "neutrophil activation signature" biomarker panel had good predictive value in identifying patients who were developing extraarticular nodules (OR 5.6, P = 0.006). CONCLUSION Neutrophils undergo marked activation and cell death in RA. Neutrophil biomarkers can provide added clinical value in the monitoring and prognosis of RA patients and may allow for early preventive treatment intervention.
Collapse
Affiliation(s)
- Mary Bach
- Division of Rheumatology, Department of Medicine,
University of Washington, Seattle, WA, USA
| | | | - Richard Moore
- Division of Rheumatology, Department of Medicine,
University of Washington, Seattle, WA, USA
| | - Tiffany Pan
- Fred Hutchinson Cancer Research Center, Seattle, WA,
USA
| | - J. Lee Nelson
- Division of Rheumatology, Department of Medicine,
University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Seattle, WA,
USA
| | - Christian Lood
- Division of Rheumatology, Department of Medicine,
University of Washington, Seattle, WA, USA
| |
Collapse
|
82
|
Wang X, Guan M, Zhang X, Ma T, Wu M, Li Y, Chen X, Zheng Y. The Association Between S100A8/A9 and the Development of Very Late Stent Thrombosis in Patients With Acute Myocardial Infarction. Clin Appl Thromb Hemost 2020; 26:1076029620943295. [PMID: 32734774 PMCID: PMC7401045 DOI: 10.1177/1076029620943295] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/18/2020] [Accepted: 06/28/2020] [Indexed: 02/05/2023] Open
Abstract
Very late stent thrombosis (VLST) is a rare but serious complication following percutaneous coronary intervention (PCI). S100A8/A9 plays an important role in thrombosis through modulating the inflammatory response. This observational study aimed to reveal the association between S100A8/A9 and VLST. Continuous blood samples were collected from patients at both the time of index PCI for acute myocardial infarction (AMI) and the time of PCI for VLST (VLST group) or follow-up coronary angiography (AMI group). In all, 56 patients were selected in each group from a cohort of 8476 patients and other 112 individuals who underwent health checkups (normal control [NC] group) were selected as controls. Serum levels of S100A8/A9 and high sensitivity C-reactive protein (hs-CRP) were tested and compared. The mean level of S100A8/A9 was 3754.4 ± 1688.9 ng/mL during index PCI and increased to 5517.8 ± 2650.9 ng/mL at the time of VLST; in the AMI group, S100A8/A9 level was 2434.9 ± 1243.4 ng/mL during index PCI and decreased to 1568.2 ± 772.1 ng/mL during follow-up, similar to that detected in the NC group (1618.2 ± 641.4 ng/mL). Of note, S100A8/A9 levels showed significant increases during VLST when compared to its own levels during index PCI, which was different from the changes of hs-CRP. Higher serum levels of S100A8/A9 are associated with the development of VLST.
Collapse
Affiliation(s)
- Xiang Wang
- Department of Cardiovascular Disease Center, The First Hospital of Jilin University, Jilin University, Jilin, People’s Republic of China
| | - Meng Guan
- Department of Oncology, The First Hospital of Jilin University, Jilin University, Jilin, People’s Republic of China
| | - Xiuhang Zhang
- Department of Burn Surgery, The First Hospital of Jilin University, Jilin University, Jilin, People’s Republic of China
| | - Taiyuan Ma
- Department of Burn Surgery, The First Hospital of Jilin University, Jilin University, Jilin, People’s Republic of China
| | - Muli Wu
- Department of Cardiovascular Center, The First Affiliated Hospital of Shantou University Medical College, Shantou University, Guangdong, People’s Republic of China
| | - Yulin Li
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Xinxin Chen
- Department of Burn Surgery, The First Hospital of Jilin University, Jilin University, Jilin, People’s Republic of China
- Xinxin Chen and Yang Zheng, No. 1 Xinmin Ave, Changchun, Jilin 130021, People’s Republic of China. Emails: ;
| | - Yang Zheng
- Department of Cardiovascular Disease Center, The First Hospital of Jilin University, Jilin University, Jilin, People’s Republic of China
- Xinxin Chen and Yang Zheng, No. 1 Xinmin Ave, Changchun, Jilin 130021, People’s Republic of China. Emails: ;
| |
Collapse
|
83
|
Diklić M, Mitrović-Ajtić O, Subotički T, Djikić D, Kovačić M, Bjelica S, Beleslin-Čokić B, Tošić M, Leković D, Gotić M, Santibanez JF, Čokić VP. IL6 inhibition of inflammatory S100A8/9 proteins is NF-κB mediated in essential thrombocythemia. Cell Biochem Funct 2019; 38:362-372. [PMID: 31885098 DOI: 10.1002/cbf.3482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/24/2019] [Accepted: 12/15/2019] [Indexed: 11/07/2022]
Abstract
This study has been performed to determine the mechanism of activation of the myeloid related S100A proteins by inflammatory cytokines in myeloproliferative neoplasm (MPN). Besides microarray analysis of MPN-derived CD34+ cells, we analysed the pro-inflammatory IL6 and anti-inflammatory IL10 dependence of NF-κB, PI3K-AKT, and JAK-STAT signalling during induction of S100A proteins in mononuclear cells of MPN, by immunoblotting and flow cytometry. We observed the reduced gene expression linked to NF-κB and inflammation signalling in MPN-derived CD34+ cells. Both IL6 and IL10 reduced S100A8 and 100A9 protein levels mediated via NF-κB and PI3K signalling, respectively, in mononuclear cells of essential thrombocythemia (ET). We also determined the increased percentage of S100A8 and S100A9 positive granulocytes in ET and primary myelofibrosis, upgraded by the JAK2V617F mutant allele burden. S100A8/9 heterodimer induced JAK1/2-dependent mitotic arrest of the ET-derived granulocytes. SIGNIFICANCE OF THE STUDY: We demonstrated that inflammation reduced the myeloid related S100A8/9 proteins by negative feedback mechanism in ET. S100A8/9 can be a diagnostic marker of inflammation in MPN, supported by the concomitant NF-κB and JAK1/2 signalling inhibition in regulation of myeloproliferation and therapy of MPN.
Collapse
Affiliation(s)
- Miloš Diklić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Olivera Mitrović-Ajtić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Tijana Subotički
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Dragoslava Djikić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Marijana Kovačić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Sunčica Bjelica
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Bojana Beleslin-Čokić
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, Genetic Laboratory, Clinical Center of Serbia, Belgrade, Serbia
| | - Milica Tošić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Danijela Leković
- Clinic of Hematology, Clinical Center of Serbia, Belgrade, Serbia.,School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Mirjana Gotić
- Clinic of Hematology, Clinical Center of Serbia, Belgrade, Serbia.,School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Juan F Santibanez
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Vladan P Čokić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| |
Collapse
|
84
|
Immune-Mediated Inflammation in Vulnerable Atherosclerotic Plaques. Molecules 2019; 24:molecules24173072. [PMID: 31450823 PMCID: PMC6749340 DOI: 10.3390/molecules24173072] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 01/16/2023] Open
Abstract
Atherosclerosis is a chronic long-lasting vascular disease leading to myocardial infarction and stroke. Vulnerable atherosclerotic (AS) plaques are responsible for these life-threatening clinical endpoints. To more successfully work against atherosclerosis, improvements in early diagnosis and treatment of AS plaque lesions are required. Vulnerable AS plaques are frequently undetectable by conventional imaging because they are non-stenotic. Although blood biomarkers like lipids, C-reactive protein, interleukin-6, troponins, and natriuretic peptides are in pathological ranges, these markers are insufficient in detecting the critical perpetuation of AS anteceding endpoints. Thus, chances to treat the patient in a preventive way are wasted. It is now time to solve this dilemma because clear results indicate a benefit of anti-inflammatory therapy per se without modification of blood lipids (CANTOS Trial, NCT01327846). This fact identifies modulation of immune-mediated inflammation as a new promising point of action for the eradication of fatal atherosclerotic endpoints.
Collapse
|
85
|
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: 12] [Impact Index Per Article: 2.4] [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
|
86
|
Daseke MJ, Valerio FM, Kalusche WJ, Ma Y, DeLeon-Pennell KY, Lindsey ML. Neutrophil proteome shifts over the myocardial infarction time continuum. Basic Res Cardiol 2019; 114:37. [PMID: 31418072 PMCID: PMC6695384 DOI: 10.1007/s00395-019-0746-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 08/06/2019] [Indexed: 12/16/2022]
Abstract
In response to myocardial infarction (MI), neutrophils (PMNs) are early responders that initiate the inflammatory reaction. Because macrophages and fibroblasts show polarization states after MI, we hypothesized PMNs also undergo phenotypic changes over the MI time course. The objective of the current study was to map the continuum of polarization phenotypes in cardiac neutrophils over the first week of MI. C57BL/6J male mice (3–6 months old) underwent permanent coronary artery ligation to induce MI, and PMNs were isolated from the infarct region at days 1, 3, 5, and 7 after MI. Day 0 served as a no MI negative control. Aptamer proteomics was performed on biological replicates (n = 10–12) for each time point. Day (D)1 MI neutrophils had a high degranulation profile with increased matrix metalloproteinase (MMP) activity. D3 MI neutrophil profiles showed upregulation of apoptosis and induction of extracellular matrix (ECM) organization. D5 MI neutrophils further increased their ECM reorganization profile. D7 MI neutrophils had a reparative signature that included expression of fibronectin, galectin-3, and fibrinogen to contribute to scar formation by stimulating ECM reorganization. Of note, fibronectin was a key modulator of degranulation, as it amplified MMP-9 release in the presence of an inflammatory stimulus. Our results indicate that neutrophils selectively degranulate over the MI time course, reflective of both their intrinsic protein profiles as well as the ECM environment in which they reside. MMPs, cathepsins, and ECM proteins were prominent neutrophil degranulation indicators.
Collapse
Affiliation(s)
- Michael J Daseke
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Fritz M Valerio
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - William J Kalusche
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Yonggang Ma
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, 33612, USA
| | - Kristine Y DeLeon-Pennell
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA.,Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Merry L Lindsey
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, 985850 Nebraska Medical Center, Omaha, NE, 68198-5850, USA. .,Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, USA.
| |
Collapse
|
87
|
Bossel Ben-Moshe N, Hen-Avivi S, Levitin N, Yehezkel D, Oosting M, Joosten LAB, Netea MG, Avraham R. Predicting bacterial infection outcomes using single cell RNA-sequencing analysis of human immune cells. Nat Commun 2019; 10:3266. [PMID: 31332193 PMCID: PMC6646406 DOI: 10.1038/s41467-019-11257-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 07/03/2019] [Indexed: 12/20/2022] Open
Abstract
Complex interactions between different host immune cell types can determine the outcome of pathogen infections. Advances in single cell RNA-sequencing (scRNA-seq) allow probing of these immune interactions, such as cell-type compositions, which are then interpreted by deconvolution algorithms using bulk RNA-seq measurements. However, not all aspects of immune surveillance are represented by current algorithms. Here, using scRNA-seq of human peripheral blood cells infected with Salmonella, we develop a deconvolution algorithm for inferring cell-type specific infection responses from bulk measurements. We apply our dynamic deconvolution algorithm to a cohort of healthy individuals challenged ex vivo with Salmonella, and to three cohorts of tuberculosis patients during different stages of disease. We reveal cell-type specific immune responses associated not only with ex vivo infection phenotype but also with clinical disease stage. We propose that our approach provides a predictive power to identify risk for disease, and human infection outcomes. Complex interactions between different host immune cell types can determine the outcome of pathogen infections. Here, Avraham and colleagues present a deconvolution algorithm that uses single-cell RNA and bulk RNA sequencing measurements of pathogen-infected cells to predict disease risk outcomes.
Collapse
Affiliation(s)
- Noa Bossel Ben-Moshe
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Shelly Hen-Avivi
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Natalia Levitin
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Dror Yehezkel
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Marije Oosting
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525, HP, Nijmegen, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525, HP, Nijmegen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525, HP, Nijmegen, the Netherlands.,Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115, Bonn, Germany
| | - Roi Avraham
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel.
| |
Collapse
|
88
|
Lin CR, Bahmed K, Tomar D, Marchetti N, Criner GJ, Bolla S, Wilson MA, Madesh M, Kosmider B. The relationship between DJ-1 and S100A8 in human primary alveolar type II cells in emphysema. Am J Physiol Lung Cell Mol Physiol 2019; 317:L791-L804. [PMID: 31313618 DOI: 10.1152/ajplung.00494.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pulmonary emphysema is characterized by alveolar type II (ATII) cell death, destruction of alveolar wall septa, and irreversible airflow limitation. Cigarette smoke induces oxidative stress and is the main risk factor for this disease development. ATII cells isolated from nonsmokers, smokers, and patients with emphysema were used for this study. ATII cell apoptosis in individuals with this disease was detected. DJ-1 and S100A8 have cytoprotective functions against oxidative stress-induced cell injury. Reduced DJ-1 and S100A8 interaction was found in ATII cells in patients with emphysema. The molecular function of S100A8 was determined by an analysis of the oxidation status of its cysteine residues using chemoselective probes. Decreased S100A8 sulfination was observed in emphysema patients. In addition, its lower levels correlated with higher cell apoptosis induced by cigarette smoke extract in vitro. Cysteine at position 106 within DJ-1 is a central redox-sensitive residue. DJ-1 C106A mutant construct abolished the cytoprotective activity of DJ-1 against cell injury induced by cigarette smoke extract. Furthermore, a molecular and complementary relationship between DJ-1 and S100A8 was detected using gain- and loss-of-function studies. DJ-1 knockdown sensitized cells to apoptosis induced by cigarette smoke extract, and S100A8 overexpression provided cytoprotection in the absence of DJ-1. DJ-1 knockout mice were more susceptible to ATII cell apoptosis induced by cigarette smoke compared with wild-type mice. Our results indicate that the impairment of DJ-1 and S100A8 function may contribute to cigarette smoke-induced ATII cell injury and emphysema pathogenesis.
Collapse
Affiliation(s)
- Chih-Ru Lin
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania.,Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania
| | - Karim Bahmed
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania.,Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania
| | - Dhanendra Tomar
- Medical Genetics and Molecular Biochemistry, Temple University, Philadelphia, Pennsylvania
| | - Nathaniel Marchetti
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania.,Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania
| | - Gerard J Criner
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania.,Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania
| | - Sudhir Bolla
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania
| | - Mark A Wilson
- Redox Biology Center and Department of Biochemistry, University of Nebraska, Lincoln, Nebraska
| | - Muniswamy Madesh
- Medical Genetics and Molecular Biochemistry, Temple University, Philadelphia, Pennsylvania
| | - Beata Kosmider
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania.,Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania.,Department of Physiology, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
89
|
Nakamura Y, Iwata H, Kuwayama T, Shirasuna K. S100A8, which increases with age, induces cellular senescence-like changes in bovine oviduct epithelial cells. Am J Reprod Immunol 2019; 82:e13163. [PMID: 31237976 DOI: 10.1111/aji.13163] [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: 04/19/2019] [Revised: 05/30/2019] [Accepted: 06/14/2019] [Indexed: 12/11/2022] Open
Abstract
PROBLEM The oviduct is an essential component in reproduction and oviduct epithelial cells (OECs) secrete various types of cytokine. However, mechanisms of aging and inflammation of OECs are unknown. We previously reported the age-dependent functional changes of bovine OECs such that aged OECs expressed higher levels of inflammatory cytokines. We selected S100A8 and S100A9 as molecules expressed more highly in aged OECs, as candidates to induce age-related changes, and investigated using bovine OECs. METHOD OF STUDY The OECs were isolated from bovine oviductal tissues (Aged, more than 120 months; Young, between 30 and 50 months) and cultured. RESULTS Aged OECs exhibited higher senescence-associated (SA)-β-gal staining (a biomarker of cellular senescence) and mRNA expression of SA-inflammatory cytokines than young OECs. Cellular senescence occurred in both young and aged OECs upon passaging the cells. Treatment with S100A8, but not S100A9, resulted in the induction of cellular senescence in bovine OECs. Both S100A8 and S100A9 stimulated the secretion of the inflammatory cytokine IL-8 from bovine OECs. S100A8-induced IL-8 secretion was dependent on receptor RAGE, AP-1 activation, and reactive oxygen species production. In addition, S100A8 reduced the content of collagen while inducing the expression of matrix metalloproteinases, suggesting the induction of dysregulation of the extracellular matrix in OECs. CONCLUSION We suggest that bovine OECs recognize an excessive increase in age-associated DAMPs, such as S100A8 and S100A9, and that these signals may contribute to chronic oviductal inflammation, resulting in infertility associated with oviductal dysfunction.
Collapse
Affiliation(s)
- Yuki Nakamura
- Laboratory of Animal Reproduction, Department of Agriculture, Tokyo University of Agriculture, Atsugi, Japan
| | - Hisataka Iwata
- Laboratory of Animal Reproduction, Department of Agriculture, Tokyo University of Agriculture, Atsugi, Japan
| | - Takehito Kuwayama
- Laboratory of Animal Reproduction, Department of Agriculture, Tokyo University of Agriculture, Atsugi, Japan
| | - Koumei Shirasuna
- Laboratory of Animal Reproduction, Department of Agriculture, Tokyo University of Agriculture, Atsugi, Japan
| |
Collapse
|
90
|
The cardioprotective effects of icariin on the isoprenaline-induced takotsubo-like rat model: Involvement of reactive oxygen species and the TLR4/NF-κB signaling pathway. Int Immunopharmacol 2019; 74:105733. [PMID: 31288151 DOI: 10.1016/j.intimp.2019.105733] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 06/26/2019] [Accepted: 06/29/2019] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Takotsubo syndrome (TS) is an acute cardiac syndrome that mimics acute coronary syndrome (ACS) but lacks coronary obstruction and is associated with sudden physical or psychiatric episodes. Several hypotheses have been proposed for the TS mechanism, but the precise cause of this syndrome remains poorly known. Recent studies noted TS patients with acute endogenous catecholamine discharge, which could trigger an oxidative stress response and inflammatory action. METHODS A single dose of the selective β-adrenergic agonist isoprenaline (ISO) was used to induce a takotsubo-like (TS-like) model. Different icariin or metoprolol doses were supplied as cardioprotective agents by intragastric administration (IG), and lipopolysaccharides (LPS) were assessed to investigate the possible mechanism of action of icariin. Transthoracic echocardiography was used to study cardiac function and morphology. The amounts of intracellular lipids and myocardial fibrosis, which represent the degree of cardiac impairment, were assessed by histological analysis. Real-time polymerase chain reaction (RT-PCR) was performed to analyze a variety of anti-oxidant elements and inflammatory factors, and Western blotting was conducted to analyze the expression of signaling pathway proteins involved in the development of TS. RESULTS The TS-like incidence in rats was lowest with icariin precondition at 2-h post-ISO administration, and both the left ventricular ejection fraction (LVEF) and ejection volume per minute were higher than those of the other groups. However, LPS administration increased the incidence of TS and aggravated cardiac impairment. Moreover, ISO significantly increased the levels of both reactive oxygen species (ROS) and TLR4/NF-κB signaling pathway proteins compared to those of the Sha-group, whereas icariin remarkably decreased the ROS levels and increased anti-oxidant element expression while reducing pro-inflammatory factor secretion and suppressing TLR4/NF-κB signaling pathway protein expression. However, the cardioprotective effect of icariin was significantly weakened by combining treatment with LPS. CONCLUSION Icariin prevented ISO-induced TS-like cardiac dysfunction in rats. The effects were induced mainly through maintenance of the dynamic balance of the ROS system, promotion of anti-oxidant element activity, and suppression of TLR4/NF-κB signaling pathway protein expression. Furthermore, the ability of icariin to increase anti-inflammatory and reduce pro-inflammatory factor secretion may be involved in the protective process.
Collapse
|
91
|
Hey YY, O'Neill TJ, O'Neill HC. A novel myeloid cell in murine spleen defined through gene profiling. J Cell Mol Med 2019; 23:5128-5143. [PMID: 31210415 PMCID: PMC6653018 DOI: 10.1111/jcmm.14382] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 04/04/2019] [Accepted: 04/17/2019] [Indexed: 12/17/2022] Open
Abstract
A novel myeloid antigen presenting cell can be generated through in vitro haematopoiesis in long‐term splenic stromal cocultures. The in vivo equivalent subset was recently identified as phenotypically and functionally distinct from the spleen subsets of macrophages, conventional (c) dendritic cells (DC), resident monocytes, inflammatory monocytes and eosinophils. This novel subset which is myeloid on the basis of cell surface phenotype, but dendritic‐like on the basis of cell surface marker expression and antigen presenting function, has been tentatively labelled “L‐DC.” Transcriptome analysis has now been employed to determine the lineage relationship of this cell type with known splenic cDC and monocyte subsets. Principal components analysis showed separation of “L‐DC” and monocytes from cDC subsets in the second principal component. Hierarchical clustering then indicated a close lineage relationship between this novel subset, resident monocytes and inflammatory monocytes. Resident monocytes were the most closely aligned, with no genes specifically expressed by the novel subset. This subset, however, showed upregulation of genes reflecting both dendritic and myeloid lineages, with strong upregulation of several genes, particularly CD300e. While resident monocytes were found to be dependent on Toll‐like receptor signalling for development and were reduced in number in Myd88‐/‐ and Trif‐/‐ mutant mice, both the novel subset and inflammatory monocytes were unaffected. Here, we describe a novel myeloid cell type closely aligned with resident monocytes in terms of lineage but distinct in terms of development and functional capacity.
Collapse
Affiliation(s)
- Ying-Ying Hey
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QLD, Australia
| | | | - Helen C O'Neill
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QLD, Australia
| |
Collapse
|
92
|
Li Y, Kong F, Jin C, Hu E, Shao Q, Liu J, He D, Xiao X. The expression of S100A8/S100A9 is inducible and regulated by the Hippo/YAP pathway in squamous cell carcinomas. BMC Cancer 2019; 19:597. [PMID: 31208368 PMCID: PMC6580480 DOI: 10.1186/s12885-019-5784-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 05/31/2019] [Indexed: 12/15/2022] Open
Abstract
Background S100A8 and S100A9, two heterodimer-forming members of the S100 family, aberrantly express in a variety of cancer types. However, little is known about the mechanism that regulates S100A8/S100A9 co-expression in cancer cells. Methods The expression level of S100A8/S100A9 measured in three squamous cell carcinomas (SCC) cell lines and their corresponding xenografts, as well as in 257 SCC tissues. The correlation between S100A8/S100A9, Hippo pathway and F-actin cytoskeleton were evaluated using western blot, qPCR, ChIP and Immunofluorescence staining tests. IncuCyte ZOOM long time live cell image monitoring system, qPCR and Flow Cytometry measured the effects of S100A8/S100A9 and YAP on cell proliferation, cell differentiation and apoptosis. Results Here, we report that through activation of the Hippo pathway, suspension and dense culture significantly induce S100A8/S100A9 co-expression and co-localization in SCC cells. Furthermore, these expressional characteristics of S100A8/S100A9 also observed in the xenografts derived from the corresponding SCC cells. Importantly, Co-expression of S100A8/S100A9 detected in 257 SCC specimens derived from five types of SCC tissues. Activation of the Hippo pathway by overexpression of Lats1, knockdown of YAP, as well as disruption of F-actin indeed obviously results in S100A8/S100A9 co-expression in attached SCC cells. Conversely, inhibition of the Hippo pathway leads to S100A8/S100A9 co-expression in a manner opposite of cell suspension and dense. In addition, we found that TEAD1 is required for YAP-induced S100A8/S100A9-expressions. The functional studies provide evidence that knockdown of S100A8/S100A9 together significantly inhibit cell proliferation but promote squamous differentiation and apoptosis. Conclusions Our findings demonstrate for the first time that the expression of S100A8/S100A9 is inducible by changes of cell shape and density through activation of the Hippo pathway in SCC cells. Induced S100A8/S100A9 promoted cell proliferation, inhibit cell differentiation and apoptosis. Electronic supplementary material The online version of this article (10.1186/s12885-019-5784-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yunguang Li
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, 19th, Beijing, 100875, China
| | - Fei Kong
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, 19th, Beijing, 100875, China
| | - Chang Jin
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, 19th, Beijing, 100875, China
| | - Enze Hu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, 19th, Beijing, 100875, China
| | - Qirui Shao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, 19th, Beijing, 100875, China
| | - Jin Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, 19th, Beijing, 100875, China
| | - Dacheng He
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, 19th, Beijing, 100875, China
| | - Xueyuan Xiao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, 19th, Beijing, 100875, China.
| |
Collapse
|
93
|
Flynn MC, Pernes G, Lee MKS, Nagareddy PR, Murphy AJ. Monocytes, Macrophages, and Metabolic Disease in Atherosclerosis. Front Pharmacol 2019; 10:666. [PMID: 31249530 PMCID: PMC6584106 DOI: 10.3389/fphar.2019.00666] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 05/23/2019] [Indexed: 12/20/2022] Open
Abstract
Atherosclerotic cardiovascular disease (CVD) is a lipid-driven chronic inflammatory disease, in which macrophages are responsible for taking up these lipids and driving disease progression. Over the years, we and others have uncovered key pathways that regulate macrophage number/function and identified how metabolic disorders such as diabetes and obesity, which are common risk factors for CVD, exacerbate these pathways. This ultimately accelerates the progression of atherosclerosis and hinders atherosclerotic regression. In this review, we discuss the different types of macrophages, from monocyte-derived macrophages, local macrophage proliferation, to macrophage-like vascular smooth muscle cells, that contribute to atherosclerosis as well as myeloid-derived suppressor cells that may have anti-atherogenic effects. We will also discuss how diabetes and obesity influence plaque macrophage accumulation and monocyte production (myelopoiesis) to promote atherogenesis as well as an exciting therapeutic target, S100A8/A9, which mediates myelopoiesis in response to both diabetes and obesity, shown to be effective in reducing atherosclerosis in pre-clinical models of diabetes.
Collapse
Affiliation(s)
- Michelle C Flynn
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Gerard Pernes
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Man Kit Sam Lee
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Prabhakara R Nagareddy
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andrew J Murphy
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| |
Collapse
|
94
|
Vadgama N, Lamont D, Hardy J, Nasir J, Lovering RC. Distinct proteomic profiles in monozygotic twins discordant for ischaemic stroke. Mol Cell Biochem 2019; 456:157-165. [PMID: 30694515 DOI: 10.1007/s11010-019-03501-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/19/2019] [Indexed: 01/13/2023]
Abstract
Stroke is a common disorder with significant morbidity and mortality, and complex aetiology involving both environmental and genetic risk factors. Although some of the major risk factors for stoke, such as smoking and hypertension, are well-documented, the underlying genetic and detailed molecular mechanisms remain elusive. Exploring the relevant biochemical pathways may contribute to the clinical diagnosis of stroke and shed light on its aetiology. A comparative proteomic analysis of blood serum of a pair of monozygotic (MZ) twins discordant for ischaemic stroke (IS) was performed using a label-free quantitative proteomics approach. To overcome the limit of reproducibility in the serum preparation, two separate runs were performed, each consisting of three technical replicates per sample. Biological processes associated with proteins differentially expressed between the twins were explored with gene ontology (GO) classification using the functional analysis tool g:Profiler. ANOVA test performed in Progenesis LC-MS identified 179 (run 1) and 209 (run 2) proteins as differentially expressed between the affected and unaffected twin (p < 0.05). Furthermore, the level of serum fibulin 1, an extracellular matrix protein associated with arterial stiffness, was on average 13.37-fold higher in the affected twin. Each dataset was then analysed independently, and the proteins were classified according to GO terms. The categories overrepresented in the affected twin predominantly corresponded to stroke-relevant processes, including wound healing, blood coagulation and haemostasis, with a high proportion of the proteins overexpressed in the affected twin associated with these terms. By contrast, in the unaffected twin diagnosed with atopic dermatitis, there were increased levels of keratin proteins and GO terms associated with skin development. The identification of cellular pathways enriched in IS as well as the upregulation of fibulin 1 sheds new light on the underlying disease-causing mechanisms at the molecular level. Our findings of distinct proteomic signatures associated with IS and atopic dermatitis suggest proteomic profiling could be used as a general approach for improved diagnostic, prognostic and therapeutic strategies.
Collapse
Affiliation(s)
- Nirmal Vadgama
- Institute of Neurology, University College London, London, UK
- Cell Biology and Genetics Research Centre, St. George's University of London, London, UK
| | - Douglas Lamont
- College of Life Sciences, University of Dundee, Dundee, UK
| | - John Hardy
- Institute of Neurology, University College London, London, UK
| | - Jamal Nasir
- Cell Biology and Genetics Research Centre, St. George's University of London, London, UK.
- Molecular Biosciences Research Group, Faculty of Health & Society, University of Northampton, Northampton, UK.
| | - Ruth C Lovering
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, London, UK
| |
Collapse
|
95
|
Hrabar J, Trumbić Ž, Bočina I, Bušelić I, Vrbatović A, Mladineo I. Interplay between proinflammatory cytokines, miRNA, and tissue lesions in Anisakis-infected Sprague-Dawley rats. PLoS Negl Trop Dis 2019; 13:e0007397. [PMID: 31091271 PMCID: PMC6538193 DOI: 10.1371/journal.pntd.0007397] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/28/2019] [Accepted: 04/16/2019] [Indexed: 12/29/2022] Open
Abstract
Background Anisakiasis is an emerging public health problem, caused by Anisakis spp. nematode larvae. Anisakiasis presents as variable and unspecific gastrointestinal and/or allergic clinical symptoms, which accounts for the high rate of misdiagnosed cases. Methodology/Principal findings The aim of this study was to characterize the early cellular (6–72 h p.i.) and molecular (6 h p.i.) immune response and general underlying regulatory mechanism in Anisakis infected rats. Each Sprague-Dawley rat was infected with 10 Anisakis spp. larvae by gastric intubation. Tissues with visible lesions were processed for: i) classic histopathology (HE), immunofluorescence (CD3, iNOS, S100A8/A9), and transmission electron microscopy (TEM); ii) target genes (Il1b, Il6, Il18, Ccl3, Icam1, Mmp9) and microRNA (Rat Immunopathology MIRN-104ZF plate, Quiagen) expression analysis; and iii) global DNA methylation. Histopathology revealed that Anisakis larval migration caused moderate to extensive hemorrhages in submucosal and epimysial/perimysial connective tissue. In stomach and muscle, moderate to abundant mixed inflammatory infiltrate was present, dominated by neutrophils and macrophages, while only mild infiltration was seen in intestine. Lesions were characterized by the presence of CD3+, iNOS+, and S100A8/A9+ cells. The greatest number of iNOS+ and S100A8/A9+ cells was seen in muscle. Il6, Il1b, and Ccl3 showed particularly strong expression in stomach and visceral adipose tissues, but the order of expression differed between tissues. In total, three miRNAs were differentially expressed, two in stomach (miRNA-451 and miRNA-223) and two in intestine (miRNA-451 and miRNA-672). No changes in global DNA methylation were observed in infected tissues relative to controls. Conclusions/Significance Anisakis infection induces strong immune responses in infected rats with marked induction of specific proinflammatory cytokines and miRNA expression. Deciphering the functional role of these cytokines and miRNAs will help in understanding the anisakiasis pathology and controversies surrounding Anisakis infection in humans. Anisakiasis is a zoonotic disease (infection transmitted between animals and humans) contracted by consumption of raw or undercooked seafood contaminated with Anisakis spp. nematode larvae. Anisakiasis usually presents with variable and unspecific gastrointestinal and/or allergic symptoms, which accounts for the high rate of misdiagnosed cases. Due to changes in dietary habits, such as eating raw or undercooked seafood, anisakiasis is considered an emerging public health problem. Despite the increase in number of reported cases worldwide, mechanisms of immune response to this unspecific human pathogen are poorly known. We have shown that in experimentally infected rats, Anisakis larvae cause severe hemorrhages and necrotic changes of affected tissues in the early phase of infections. Neutrophils and macrophages were abundantly present in tissue lesions, while eosinophils, hallmark of helminth infections, were scarcely present. We have also demonstrated particularly strong expression of several inflammatory genes. Moreover, we give for the first-time insight into putative regulatory mechanism mediated via a distinct class of RNA molecules. Our study may provide new opportunities for better understanding of cellular and molecular response to Anisakis spp., aiming at development of more specific therapeutics and alleviation of pathologies associated with Anisakis spp. infection.
Collapse
Affiliation(s)
- Jerko Hrabar
- Laboratory of Aquaculture, Institute of Oceanography and Fisheries, Split, Croatia
- * E-mail:
| | - Željka Trumbić
- Department of Marine Studies, University of Split, Split, Croatia
| | - Ivana Bočina
- Faculty of Science, University of Split, Split, Croatia
| | - Ivana Bušelić
- Laboratory of Aquaculture, Institute of Oceanography and Fisheries, Split, Croatia
| | - Anamarija Vrbatović
- Laboratory of Aquaculture, Institute of Oceanography and Fisheries, Split, Croatia
| | - Ivona Mladineo
- Laboratory of Aquaculture, Institute of Oceanography and Fisheries, Split, Croatia
| |
Collapse
|
96
|
Kawano T, Shimamura M, Nakagami H, Iso T, Koriyama H, Takeda S, Baba K, Sasaki T, Sakaguchi M, Morishita R, Mochizuki H. Therapeutic Vaccine Against S100A9 (S100 Calcium-Binding Protein A9) Inhibits Thrombosis Without Increasing the Risk of Bleeding in Ischemic Stroke in Mice. Hypertension 2019; 72:1355-1364. [PMID: 30571223 DOI: 10.1161/hypertensionaha.118.11316] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Decreased adherence to daily ingestion of antiplatelet drugs is a critical issue, increasing mortality and morbidity in poststroke patients. As vaccination could be a promising approach to solving this, we designed an antiplatelet vaccine that inhibited S100A9 (S100 calcium-binding protein A9)/CD36 (cluster of differentiation 36) signaling in platelets, which was reported to be a key signal in arterial thrombosis, but not hemostasis. Immunization with this vaccine induced a sustainable increase in the anti-S100A9 antibody titer for >2 months and an additional booster immunization enhanced the antibody production further. The middle cerebral artery occlusion time was successfully prolonged in the vaccinated mice, which was comparable to that in mice treated with clopidogrel. The antithrombotic effect lasted for 84 days after the last vaccination, as well as after the booster immunization. Importantly, the bleeding time was not affected in the immunized mice. The antithrombotic effect was also observed in the common carotid artery, which was similar to that found in CD36-/- mice. Vascular injury increased the expression of S100A9 in the serum and phosphorylation of JNK (c-Jun N-terminal kinase) and VAV1 in the platelets, but these increases were inhibited in the immunized mice. Moreover, the S100A9 vaccine did not induce cell-mediated autoimmunity, as demonstrated by the enzyme-linked immunosorbent spot assay. Thus, immunization with the S100A9 vaccine resulted in long-term inhibition of thrombus formation through inhibition of increased S100A9/CD36 signaling without risk of bleeding or adverse autoimmune responses. Vaccination against S100A9 might be a novel therapy to prevent recurrent ischemic stroke.
Collapse
Affiliation(s)
- Tomohiro Kawano
- From the Department of Neurology (T.K., M.S., K.B., T.S., M.S., H.M.), Osaka University Graduate School of Medicine, Japan
- Department of Health Development and Medicine (T.K., M.S., H.N., H.K.), Osaka University Graduate School of Medicine, Japan
| | - Munehisa Shimamura
- From the Department of Neurology (T.K., M.S., K.B., T.S., M.S., H.M.), Osaka University Graduate School of Medicine, Japan
- Department of Health Development and Medicine (T.K., M.S., H.N., H.K.), Osaka University Graduate School of Medicine, Japan
| | - Hironori Nakagami
- Department of Health Development and Medicine (T.K., M.S., H.N., H.K.), Osaka University Graduate School of Medicine, Japan
| | - Tatsuya Iso
- Education and Research Support Center, Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Japan (T.I.)
| | - Hiroshi Koriyama
- Department of Health Development and Medicine (T.K., M.S., H.N., H.K.), Osaka University Graduate School of Medicine, Japan
| | - Shuko Takeda
- Department of Clinical Gene Therapy (S.T., R.M.), Osaka University Graduate School of Medicine, Japan
| | - Kosuke Baba
- From the Department of Neurology (T.K., M.S., K.B., T.S., M.S., H.M.), Osaka University Graduate School of Medicine, Japan
| | - Tsutomu Sasaki
- From the Department of Neurology (T.K., M.S., K.B., T.S., M.S., H.M.), Osaka University Graduate School of Medicine, Japan
| | - Manabu Sakaguchi
- From the Department of Neurology (T.K., M.S., K.B., T.S., M.S., H.M.), Osaka University Graduate School of Medicine, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy (S.T., R.M.), Osaka University Graduate School of Medicine, Japan
| | - Hideki Mochizuki
- From the Department of Neurology (T.K., M.S., K.B., T.S., M.S., H.M.), Osaka University Graduate School of Medicine, Japan
| |
Collapse
|
97
|
Shimamura M, Nakagami H. A vaccine targeting blood clot formation: what is the potential? Expert Rev Vaccines 2019; 18:419-421. [PMID: 30884991 DOI: 10.1080/14760584.2019.1588114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Munehisa Shimamura
- a Department of Health Development and Medicine , Osaka University Graduate School of Medicine , Suita , Osaka , Japan.,b Department of Neurology , Osaka University Graduate School of Medicine , Japan
| | - Hironori Nakagami
- a Department of Health Development and Medicine , Osaka University Graduate School of Medicine , Suita , Osaka , Japan
| |
Collapse
|
98
|
Duvetorp A, Söderman J, Assarsson M, Skarstedt M, Svensson Å, Seifert O. Observational study on Swedish plaque psoriasis patients receiving narrowband-UVB treatment show decreased S100A8/A9 protein and gene expression levels in lesional psoriasis skin but no effect on S100A8/A9 protein levels in serum. PLoS One 2019; 14:e0213344. [PMID: 30865695 PMCID: PMC6415841 DOI: 10.1371/journal.pone.0213344] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 02/12/2019] [Indexed: 11/19/2022] Open
Abstract
S100A8 and S100A9 proteins are highly upregulated in patients with psoriasis and have been proposed as potential biomarkers for psoriasis. The present study was designed to analyze the effect of narrowband ultraviolet B therapy on these proteins. S100A8, S100A9 gene expression and S100A8/A9 heterocomplex protein levels were analyzed in lesional and non-lesional skin before and after narrowband-UVB treatment in patients with chronic plaque type psoriasis. In addition, disease severity was measured by psoriasis area and severity index (PASI) and serum protein levels of S100A8/A9 were repeatedly analyzed. Narrowband-UVB treatment significantly reduced S100A8, S100A9 gene expression and S100A8/A9 protein levels in lesional skin while serum levels showed no significant change. No correlation between PASI and serum S100A8/A9 protein levels was found. These results implicate a role of S100A8/A9 in the anti-inflammatory effect of narrowband-UVB. Serum S100A8/A9 levels do not respond to treatment suggesting that serum S100A8/A9 does not originate from psoriasis skin keratinocytes. Serum S100A8/A9 levels do not correlate with PASI questioning serum S100A8/A9 as a biomarker for psoriasis skin activity. Trial Registration: DRKS 00014817.
Collapse
Affiliation(s)
- Albert Duvetorp
- Department of Dermatology and Venereology, Division of endocrinology, skin, reproductive health and ophthalmology, Skåne University Hospital, Malmö, Sweden
- Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
- * E-mail:
| | - Jan Söderman
- Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
- Laboratory Medicine, Division of Medical Diagnostics, Ryhov County Hospital, Jönköping, Sweden
| | - Malin Assarsson
- Department of Dermatology and Venereology, Division of Medical Health, Ryhov County Hospital, Jönköping, Sweden
| | - Marita Skarstedt
- Laboratory Medicine, Division of Medical Diagnostics, Ryhov County Hospital, Jönköping, Sweden
| | - Åke Svensson
- Department of Dermatology and Venereology, Division of endocrinology, skin, reproductive health and ophthalmology, Skåne University Hospital, Malmö, Sweden
| | - Oliver Seifert
- Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
- Department of Dermatology and Venereology, Division of Medical Health, Ryhov County Hospital, Jönköping, Sweden
| |
Collapse
|
99
|
Siljan WW, Holter JC, Michelsen AE, Nymo SH, Lauritzen T, Oppen K, Husebye E, Ueland T, Mollnes TE, Aukrust P, Heggelund L. Inflammatory biomarkers are associated with aetiology and predict outcomes in community-acquired pneumonia: results of a 5-year follow-up cohort study. ERJ Open Res 2019; 5:00014-2019. [PMID: 30863773 PMCID: PMC6409082 DOI: 10.1183/23120541.00014-2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 01/15/2019] [Indexed: 01/26/2023] Open
Abstract
Background Biomarkers may facilitate clinical decisions in order to guide antimicrobial treatment and prediction of prognosis in community-acquired pneumonia (CAP). We measured serum C-reactive protein, procalcitonin (PCT) and calprotectin levels, and plasma pentraxin 3 (PTX3) and presepsin levels, along with whole-blood white cell counts, at three time-points, and examined their association with microbial aetiology and adverse clinical outcomes in CAP. Methods Blood samples were obtained at hospital admission, clinical stabilisation and 6-week follow-up from 267 hospitalised adults with CAP. Adverse short-term outcome was defined as intensive care unit admission and 30-day mortality. Long-term outcome was evaluated as 5-year all-cause mortality. Results Peak levels of all biomarkers were seen at hospital admission. Increased admission levels of C-reactive protein, PCT and calprotectin were associated with bacterial aetiology of CAP, while increased admission levels of PCT, PTX3 and presepsin were associated with adverse short-term outcome. In univariate and multivariate regression models, white blood cells and calprotectin at 6-week follow-up were predictors of 5-year all-cause mortality. Conclusions Calprotectin emerges as both a potential early marker of bacterial aetiology and a predictor for 5-year all-cause mortality in CAP, whereas PCT, PTX3 and presepsin may predict short-term outcome. In 267 adults with community-acquired pneumonia, systemic calprotectin emerges as an early marker of bacterial aetiology and a predictor of 5-year mortality, whereas systemic procalcitonin, pentraxin 3 and presepsin are predictors of short-term outcomehttp://ow.ly/dz6S30nAFvn
Collapse
Affiliation(s)
- William W Siljan
- Dept of Internal Medicine, Drammen Hospital, Vestre Viken Hospital Trust, Drammen, Norway.,Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jan C Holter
- Dept of Internal Medicine, Drammen Hospital, Vestre Viken Hospital Trust, Drammen, Norway.,Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Annika E Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ståle H Nymo
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Trine Lauritzen
- Dept of Medical Biochemistry, Drammen Hospital, Vestre Viken Hospital Trust, Drammen, Norway
| | - Kjersti Oppen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Dept of Medical Biochemistry, Drammen Hospital, Vestre Viken Hospital Trust, Drammen, Norway
| | - Einar Husebye
- Dept of Internal Medicine, Drammen Hospital, Vestre Viken Hospital Trust, Drammen, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Faculty of Health Sciences, K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway
| | - Tom E Mollnes
- Faculty of Health Sciences, K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway.,Research Laboratory, Nordland Hospital, Bodø, Norway.,Dept of Immunology, Faculty of Medicine, University of Oslo, Oslo, Norway.,K.G. Jebsen Inflammatory Research Center, University of Oslo, Oslo, Norway.,Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,K.G. Jebsen Inflammatory Research Center, University of Oslo, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Lars Heggelund
- Dept of Internal Medicine, Drammen Hospital, Vestre Viken Hospital Trust, Drammen, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
100
|
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: 3.4] [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
|