1
|
Li P, Ji Y, Shen D, Liu Y, Hao Y, Yang D, Fan Y, Li W, Zhu S, Sun W, Li P, Zhang S. Integrated Analysis of Intersecting Neutrophil Signatures in Behçet's Disease and Inflammatory Bowel Disease. Int J Rheum Dis 2025; 28:e70229. [PMID: 40257285 DOI: 10.1111/1756-185x.70229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 04/22/2025]
Abstract
INTRODUCTION Behçet's disease (BD) and inflammatory bowel disease (IBD) are chronic inflammatory diseases characterized by immune system dysregulation. The critical role of neutrophils in these conditions is increasingly recognized. This study aimed to identify a shared set of neutrophils differentially expressed genes (NDEGs) to aid in the differential diagnosis of the two diseases. METHODS Bioinformatics analysis of GEO data combined with WGCNA identified 65 key NDEGs. Functional enrichment and immune infiltration analyses were conducted. RT-qPCR validated six hub NDEGs in neutrophils from IBD and BD patients. Serum CD226 levels were measured by ELISA, and a ROC curve assessed its diagnostic value. Additionally, neutrophils were stimulated with patient serum, followed by Western blot analysis. RESULTS Immune infiltration analysis showed higher blood neutrophil levels in BD than in IBD. Neutrophil sequencing identified NDEGs upregulated in BD but downregulated in IBD, linked to T-cell receptor pathways. RT-PCR confirmed elevated FYN, CD99, SKAP1, and CD226 in BD neutrophils, while KLRG1 and MATK were higher in IBD. ELISA showed increased serum CD226 in BD. Western blot revealed higher Elastase and PAD4 in BD-stimulated neutrophils, while CXCL11 was elevated in IBD-stimulated neutrophils. CONCLUSIONS Our findings suggest that BD and IBD neutrophils may have distinct functional states, potentially linked to differential T-cell interactions. These insights highlight neutrophils' diverse roles in immune dysregulation and their potential as diagnostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Pengchong Li
- State Key Laboratory of Digestive Health, Department of Gastroenterology, Beijing Digestive Disease Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing, China
| | - Yuxiao Ji
- State Key Laboratory of Digestive Health, Department of Gastroenterology, Beijing Digestive Disease Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing, China
| | - Dan Shen
- Department of Rheumatology, Institute of Geriatric Medicine, National Centre of Gerontology, Clinical Immunology Centre, Peking Union Medical College, Beijing Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuqi Liu
- Department of Rheumatology, Institute of Geriatric Medicine, National Centre of Gerontology, Clinical Immunology Centre, Peking Union Medical College, Beijing Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuanzhen Hao
- State Key Laboratory of Digestive Health, Department of Gastroenterology, Beijing Digestive Disease Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing, China
| | - Deyi Yang
- State Key Laboratory of Digestive Health, Department of Gastroenterology, Beijing Digestive Disease Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing, China
| | - Yuhui Fan
- State Key Laboratory of Digestive Health, Department of Gastroenterology, Beijing Digestive Disease Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing, China
| | - Wenkun Li
- State Key Laboratory of Digestive Health, Department of Gastroenterology, Beijing Digestive Disease Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing, China
| | - Shengtao Zhu
- State Key Laboratory of Digestive Health, Department of Gastroenterology, Beijing Digestive Disease Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing, China
| | - Wei Sun
- Department of Rheumatology, Institute of Geriatric Medicine, National Centre of Gerontology, Clinical Immunology Centre, Peking Union Medical College, Beijing Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Peng Li
- State Key Laboratory of Digestive Health, Department of Gastroenterology, Beijing Digestive Disease Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing, China
| | - Shutian Zhang
- State Key Laboratory of Digestive Health, Department of Gastroenterology, Beijing Digestive Disease Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing, China
| |
Collapse
|
2
|
Li D, Li Y, Zhu K, Yuan Y, He Z, Sun Q, Jin M. Analysis of m6A-regulated genes and subtype classification in lupus nephritis. BMC Nephrol 2024; 25:119. [PMID: 38570749 PMCID: PMC10988804 DOI: 10.1186/s12882-024-03549-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/18/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Lupus nephritis (LN) is the most common and severe clinical manifestation of systemic lupus erythematosus (SLE). N6-methyladenosine (m6A) is a reversible RNA modification and has been implicated in various biological processes. However, the roles of m6A regulators in LN are not fully demonstrated. METHODS We downloaded the kidney tissue transcriptome dataset of LN patients and normal controls from the GEO database and extracted the expression levels of m6A regulators. We constructed and compared Random Forest (RF) and Support Vector Machine (SVM) models, and subsequently selected featured genes to develop nomogram models. The m6A subtypes were identified based on significantly differentially expressed m6A regulators, and the m6A gene subtypes were identified based on m6A-associated differential genes, and the two m6A modification patterns were comprehensively evaluated. RESULTS We obtained the GSE32591 and GSE112943 datasets from the GEO database, including 78 LN samples and 36 normal control samples. We extracted the expression levels of 20 m6A regulators. By RF analysis we identified 7 characteristic m6A regulators and constructed nomogramh models with these 7 genes. We identified two m6A subtypes based on these seven important m6A regulators, and the immune cell infiltration levels of the two subtype clusters were significantly different. We identified two more m6A gene subtypes based on m6A-associated DEGs. We calculated the m6A scores using the principal component analysis (PCA) algorithm and found that the m6A scores of m6A cluster A and gene cluster A were lower than those of m6A cluster B and gene cluster B. In addition, we found that the levels of inflammatory factors were also significantly different between m6A clusters and gene clusters. CONCLUSION This study confirms that m6A regulators are involved in the LN process through different modes of action and provide new diagnostic and therapeutic targets for LN.
Collapse
Affiliation(s)
- Diangeng Li
- Department of Nephrology, Beijing-Chaoyang Hospital, Capital Medical University, 100020, Beijing, China.
| | - Yanchun Li
- Department of Nephrology, Beijing-Chaoyang Hospital, Capital Medical University, 100020, Beijing, China
| | - Kaiyi Zhu
- Department of Nephrology, Beijing-Chaoyang Hospital, Capital Medical University, 100020, Beijing, China
| | - Yuqing Yuan
- Department of Nephrology, Beijing-Chaoyang Hospital, Capital Medical University, 100020, Beijing, China
| | - Zheng He
- Department of Clinical Laboratory, Chinese PLA General Hospital, 100853, Beijing, China
| | - Qianmei Sun
- Department of Nephrology, Beijing-Chaoyang Hospital, Capital Medical University, 100020, Beijing, China.
| | - Meiling Jin
- Department of Nephrology, Beijing-Chaoyang Hospital, Capital Medical University, 100020, Beijing, China.
| |
Collapse
|
3
|
Mathur R, Deo K, Raheja A. Systemic Lupus Erythematosus in India: A Clinico-Serological Correlation. Cureus 2022; 14:e25763. [PMID: 35812543 PMCID: PMC9270079 DOI: 10.7759/cureus.25763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2022] [Indexed: 11/05/2022] Open
|
4
|
Brunekreef TE, Reteig LC, Limper M, Haitjema S, Dias J, Mathsson-Alm L, van Laar JM, Otten HG. Microarray analysis of autoantibodies can identify future Systemic Lupus Erythematosus patients. Hum Immunol 2022; 83:509-514. [DOI: 10.1016/j.humimm.2022.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/25/2022] [Accepted: 03/31/2022] [Indexed: 11/04/2022]
|
5
|
Al-Mughales JA. Anti-Nuclear Antibodies Patterns in Patients With Systemic Lupus Erythematosus and Their Correlation With Other Diagnostic Immunological Parameters. Front Immunol 2022; 13:850759. [PMID: 35359932 PMCID: PMC8964090 DOI: 10.3389/fimmu.2022.850759] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 02/18/2022] [Indexed: 11/20/2022] Open
Abstract
Background Antinuclear antibodies (ANA) are major immunodiagnostic tools in systemic lupus erythematosus (SLE); however, their clinical and pathogenic roles are not yet elucidated and are a subject of controversy. Objectives The aim of the study is to explore the pathogenic significance of ANA patterns among SLE patients, by analyzing their association with ANA titers, complement levels and other pathogenic immune markers, namely, anti-double-stranded DNA (anti-dsDNA), complements C3 and C4, rheumatoid factor (RF), anticardiolipin antibodies IgG (ACL IgG) and IgM (ACL IgM), Beta-2 Glycoprotein 1 Antibodies (β2-GP) IgG (β2-IgM) and IgM (β2-IgM), and lupus anticoagulant (LA). Method A comparative cross-sectional study was conducted among 495 SLE patients, who were diagnosed and classified by consultant rheumatologists according to the new European League Against Rheumatism (EULAR)/American College of Rheumatology (ACR) 2019 criteria. SLE immunodiagnostic profiles were analyzed including the following parameters: ANA antibody titers and staining patterns, anti-dsDNA, C3 and C4 levels, aCL, and anti-β2-GP and LA. Result The most frequently observed ANA patterns were the speckled (52.1%) and homogeneous (35.2%) patterns, while other patterns were rare representing less than 7% of the patients each. ANA titers were highest in patients with mixed pattern followed by the speckled pattern. Of all the investigated patterns, the peripheral pattern showed the most pathogenic immune profile, namely, highest levels of anti-dsDNA, lowest levels of C4, and highest levels of aCL and β2-GP IgG and IgM. Conclusion This retrospective study showed that speckled followed by homogeneous ANA patterns were predominant accounting for 52.1 and 35.2% of the patients. The ANA pattern showed several associations with other immune markers that are documented to have significant clinical implications in SLE. Peripheral, mixed, and speckled patterns were associated with higher profiles of immune markers indicative of a potential prognostic value of these patterns in SLE.
Collapse
Affiliation(s)
- Jamil A. Al-Mughales
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Clinical Laboratory Medicine, Diagnostic Immunology Division, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| |
Collapse
|
6
|
Kashiwagi H, Morishima N, Obuse S, Isoshima T, Akimoto J, Ito Y. SARS-CoV-2 Proteins Microarray by Photoimmobilization for Serodiagnosis of the Antibodies. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2021. [DOI: 10.1246/bcsj.20210215] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Hiroharu Kashiwagi
- Emergent Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
- R-NanoBio Co., Ltd., Wako-RIKEN Incubation Plaza, 3-13 Minami, Wako, Saitama 351-0104, Japan
| | - Nobuhiro Morishima
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Sei Obuse
- Emergent Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
- R-NanoBio Co., Ltd., Wako-RIKEN Incubation Plaza, 3-13 Minami, Wako, Saitama 351-0104, Japan
| | - Takashi Isoshima
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Jun Akimoto
- Emergent Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yoshihiro Ito
- Emergent Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
- R-NanoBio Co., Ltd., Wako-RIKEN Incubation Plaza, 3-13 Minami, Wako, Saitama 351-0104, Japan
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
7
|
Li S, Song G, Bai Y, Song N, Zhao J, Liu J, Hu C. Applications of Protein Microarrays in Biomarker Discovery for Autoimmune Diseases. Front Immunol 2021; 12:645632. [PMID: 34012435 PMCID: PMC8126629 DOI: 10.3389/fimmu.2021.645632] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/13/2021] [Indexed: 01/18/2023] Open
Abstract
Dysregulated autoantibodies and cytokines were deemed to provide important cues for potential illnesses, such as various carcinomas and autoimmune diseases. Increasing biotechnological approaches have been applied to screen and identify the specific alterations of these biomolecules as distinctive biomarkers in diseases, especially autoimmune diseases. As a versatile and robust platform, protein microarray technology allows researchers to easily profile dysregulated autoantibodies and cytokines associated with autoimmune diseases using various biological specimens, mainly serum samples. Here, we summarize the applications of protein microarrays in biomarker discovery for autoimmune diseases. In addition, the key issues in the process of using this approach are presented for improving future studies.
Collapse
Affiliation(s)
- Siting Li
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,Department of Rheumatology, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Guang Song
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yina Bai
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,Department of Rheumatology, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Ning Song
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,Department of Rheumatology, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Jiuliang Zhao
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,Department of Rheumatology, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Jian Liu
- Department of Rheumatology, Aerospace Center Hospital, Aerospace, Clinical Medical College, Peking University, Beijing, China
| | - Chaojun Hu
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,Department of Rheumatology, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| |
Collapse
|
8
|
Manotas MC, Calderón JC, López-Kleine L, Suárez-Obando F, Moreno OM, Rojas A. Identification of common differentially expressed genes in Turner (45,X) and Klinefelter (47,XXY) syndromes using bioinformatics analysis. Mol Genet Genomic Med 2020; 8:e1503. [PMID: 32959501 PMCID: PMC7667333 DOI: 10.1002/mgg3.1503] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 08/14/2020] [Accepted: 08/30/2020] [Indexed: 12/12/2022] Open
Abstract
Background Analysis of patients with chromosomal abnormalities, including Turner syndrome and Klinefelter syndrome, has highlighted the importance of X‐linked gene dosage as a contributing factor for disease susceptibility. Escape from X‐inactivation and X‐linked imprinting can result in transcriptional differences between normal men and women as well as in patients with sex chromosome abnormalities. Objective To identify differentially expressed genes among patients with Turner (45,X) and Klinefelter (46,XXY) syndrome using bioinformatics analysis. Methodology Two gene expression data sets of Turner (45,X) and Klinefelter syndrome (47,XXY) were obtained from the Gene Omnibus Expression (GEO) database of the National Center for Biotechnology Information (NCBI). Statistical analysis was performed using R Bioconductor libraries. Differentially expressed genes (DEGs) were determined using significance analysis of microarray (SAM). The functional annotation of the DEGs was performed with DAVID v6.8 (The Database for Annotation, Visualization, and Integrated Discovery). Results There are no genes over‐expressed simultaneously in both diseases. However, when crossing the list of under‐expressed genes for 45,X cells and the list of over‐expressed genes for 47,XXY cells, there are 16 common genes: SLC25A6, AKAP17A, ASMTL, KDM5C, KDM6A, ATRX, CSF2RA, DHRSX, CD99, ZBED1, EIF1AX, MVB12B, SMC1A, P2RY8, DOCK7, DDX3X, eight of which are involved in the regulation of gene expression by epigenetic mechanisms, regulation of splicing processes and protein synthesis. Conclusion Of the 16 identified as under‐expressed in 45,X cells and over‐expressed in 47,XXY cells, 14 are located in X chromosome and 2 in autosomal chromosome; 8 of these genes are involved in the regulation of gene expression: 5 genes are related to epigenetic mechanisms, 2 in regulation of splicing processes, and 1 in the protein synthesis process. Our results are limited by it being the product of a bioinformatic analysis from mRNA isolated from whole blood, this makes necessary further exploration of the relationships between these genes and Turner syndrome and Klinefelter syndrome in the future.
Collapse
Affiliation(s)
- María Carolina Manotas
- Institute of Human Genetics. Faculty of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Juan Camilo Calderón
- Department of Statistics, Faculty of Science, Universidad Nacional de Colombia, Ciudad Universitaria, Bogotá, Colombia
| | - Liliana López-Kleine
- Department of Statistics, Faculty of Science, Universidad Nacional de Colombia, Ciudad Universitaria, Bogotá, Colombia
| | - Fernando Suárez-Obando
- Institute of Human Genetics. Faculty of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Olga M Moreno
- Institute of Human Genetics. Faculty of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Adriana Rojas
- Institute of Human Genetics. Faculty of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
9
|
Song W, Tang D, Chen D, Zheng F, Huang S, Xu Y, Yu H, He J, Hong X, Yin L, Liu D, Dai W, Dai Y. Advances in applying of multi-omics approaches in the research of systemic lupus erythematosus. Int Rev Immunol 2020; 39:163-173. [PMID: 32138562 DOI: 10.1080/08830185.2020.1736058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Wencong Song
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Donge Tang
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Deheng Chen
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Fengping Zheng
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Shaoying Huang
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Yong Xu
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Haiyan Yu
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Jingquan He
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Xiaoping Hong
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Lianghong Yin
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dongzhou Liu
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Weier Dai
- College of Natural Science, University of Texas at Austin, Austin, TX, USA
| | - Yong Dai
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
10
|
Radin M, Sciascia S, Cecchi I, Mengozzi G, Roccatello D. The potential role of SLE-key test in identifying patients with Systemic Lupus Erythematosus: Results from a prospective, real-world experience. Eur J Intern Med 2019; 68:e12-e14. [PMID: 31378436 DOI: 10.1016/j.ejim.2019.07.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 11/15/2022]
Affiliation(s)
- Massimo Radin
- Center of Research of Immunopathology and Rare Diseases, Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, S. Giovanni Bosco Hospital, Department of Clinical and Biological Sciences, University of Turin, Italy; School of Specialization of Clinical Pathology, Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Savino Sciascia
- Center of Research of Immunopathology and Rare Diseases, Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, S. Giovanni Bosco Hospital, Department of Clinical and Biological Sciences, University of Turin, Italy; Nephrology and Dialysis, Department of Clinical and Biological Sciences, S. Giovanni Bosco Hospital, University of Turin, Italy.
| | - Irene Cecchi
- Center of Research of Immunopathology and Rare Diseases, Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, S. Giovanni Bosco Hospital, Department of Clinical and Biological Sciences, University of Turin, Italy; School of Specialization of Clinical Pathology, Department of Clinical and Biological Sciences, University of Turin, Italy
| | | | - Dario Roccatello
- Center of Research of Immunopathology and Rare Diseases, Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, S. Giovanni Bosco Hospital, Department of Clinical and Biological Sciences, University of Turin, Italy; Nephrology and Dialysis, Department of Clinical and Biological Sciences, S. Giovanni Bosco Hospital, University of Turin, Italy
| |
Collapse
|
11
|
Blomberg J, Rizwan M, Böhlin-Wiener A, Elfaitouri A, Julin P, Zachrisson O, Rosén A, Gottfries CG. Antibodies to Human Herpesviruses in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome Patients. Front Immunol 2019; 10:1946. [PMID: 31475007 PMCID: PMC6702656 DOI: 10.3389/fimmu.2019.01946] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 08/01/2019] [Indexed: 11/13/2022] Open
Abstract
Myalgic encephalomyelitis, also referred to as chronic fatigue syndrome (ME/CFS) is a debilitating disease characterized by myalgia and a sometimes severe limitation of physical activity and cognition. It is exacerbated by physical and mental activity. Its cause is unknown, but frequently starts with an infection. The eliciting infection (commonly infectious mononucleosis or an upper respiratory infection) can be more or less well diagnosed. Among the human herpesviruses (HHV-1-8), HHV-4 (Epstein-Barr virus; EBV), HHV-6 (including HHV-6A and HHV-6B), and HHV-7, have been implicated in the pathogenesis of ME/CFS. It was therefore logical to search for serological evidence of past herpesvirus infection/reactivation in several cohorts of ME/CFS patients (all diagnosed using the Canada criteria). Control samples were from Swedish blood donors. We used whole purified virus, recombinant proteins, and synthetic peptides as antigens in a suspension multiplex immunoassay (SMIA) for immunoglobulin G (IgG). The study on herpesviral peptides based on antigenicity with human sera yielded novel epitope information. Overall, IgG anti-herpes-viral reactivities of ME/CFS patients and controls did not show significant differences. However, the high precision and internally controlled format allowed us to observe minor relative differences between antibody reactivities of some herpesviral antigens in ME/CFS versus controls. ME/CFS samples reacted somewhat differently from controls with whole virus HHV-1 antigens and recombinant EBV EBNA6 and EA antigens. We conclude that ME/CFS samples had similar levels of IgG reactivity as blood donor samples with HHV-1-7 antigens. The subtle serological differences should not be over-interpreted, but they may indicate that the immune system of some ME/CFS patients interact with the ubiquitous herpesviruses in a way different from that of healthy controls.
Collapse
Affiliation(s)
- Jonas Blomberg
- Section of Clinical Microbiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Muhammad Rizwan
- Section of Clinical Microbiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Agnes Böhlin-Wiener
- Section of Clinical Microbiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Amal Elfaitouri
- Department of Infectious Disease and Tropical Medicine, Faculty of Public Health, Benghazi University, Benghazi, Libya
| | - Per Julin
- Neurological Rehabilitation Clinic, Stora Sköndal, Sköndal, Sweden.,Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | | | - Anders Rosén
- Division of Cell Biology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | | |
Collapse
|
12
|
Mazzoleni A, Mallet JM, Rovero P, Papini AM. Glycoreplica peptides to investigate molecular mechanisms of immune-mediated physiological versus pathological conditions. Arch Biochem Biophys 2019; 663:44-53. [PMID: 30594643 DOI: 10.1016/j.abb.2018.12.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/20/2018] [Accepted: 12/26/2018] [Indexed: 12/17/2022]
Abstract
Investigation of the role of saccharides and glycoconjugates in mechanisms of immune-mediated physiological and pathological conditions is a hot topic. In fact, in many autoimmune diseases cross-reactivity between sugar moieties exposed on exogenous pathogens and self-molecules has long been hinted. Several peptides have been reported as mimetics of glycans specifically interacting with sugar-binding antibodies. The seek for these glycoreplica peptides is instrumental in characterizing antigen mimicry pathways and their involvement in triggering autoimmunity. Therefore, peptides mimicking glycan-protein interactions are valuable molecular tools to overcome the difficulties of oligosaccharide preparations. The clinical impact of peptide-based probes for autoimmune diseases diagnosis and follow-up is emerging only recently as just the tip of the iceberg of an overlooked potential. Here we provide a brief overview of the relevance of the structural and functional aspects of peptide probes and their mimicry effect in autoimmunity mechanisms for promising applications in diagnostics and therapeutics.
Collapse
Affiliation(s)
- Antonio Mazzoleni
- Laboratory of Peptide and Protein Chemistry and Biology, Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 13, 50019, Sesto Fiorentino, Italy; Laboratoire des Biomolécules, UMR 7203, Département de chimie, École Normale Supérieure, PSL Research University, Sorbonne Universités, UPMC Université Paris 06, CNRS, 24 rue Lhomond, 75005, Paris, France
| | - Jean-Maurice Mallet
- Laboratoire des Biomolécules, UMR 7203, Département de chimie, École Normale Supérieure, PSL Research University, Sorbonne Universités, UPMC Université Paris 06, CNRS, 24 rue Lhomond, 75005, Paris, France
| | - Paolo Rovero
- Laboratory of Peptide and Protein Chemistry and Biology, Department of Neurosciences, Psychology, Drug Research and Child Health - Section of Pharmaceutical Sciences and Nutraceuticals, University of Florence, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Italy
| | - Anna Maria Papini
- Laboratory of Peptide and Protein Chemistry and Biology, Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 13, 50019, Sesto Fiorentino, Italy; Platform of Peptide and Protein Chemistry and Biology - PeptLab@UCP and Laboratory of Chemical Biology EA4505, Université Paris-Seine, 5 Mail Gay-Lussac, 95031, Cergy-Pontoise CEDEX, France.
| |
Collapse
|
13
|
Lakota K, Švec T, Kveder T, Sodin-Šemrl S, Žigon P, Ambrožič A, Ogrič M, Markez S, Božič B, Tomšič M, Čučnik S. Autoantibodies against dsDNA measured with nonradioactive Farr assay—an alternative for routine laboratories. Clin Rheumatol 2018; 38:353-359. [DOI: 10.1007/s10067-018-4271-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/30/2018] [Accepted: 08/20/2018] [Indexed: 10/28/2022]
|
14
|
Putterman C, Pisetsky DS, Petri M, Caricchio R, Wu AHB, Sanz I, Oates JC, Wallace S, Sorek R, Gerwien R, Safer P, Jakobi-Brook K, Cohen IR. The SLE-key test serological signature: new insights into the course of lupus. Rheumatology (Oxford) 2018; 57:1632-1640. [PMID: 29873771 PMCID: PMC6105919 DOI: 10.1093/rheumatology/key149] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 04/24/2018] [Indexed: 01/13/2023] Open
Abstract
Objective We previously described the multiplex autoantibody SLE-key Rule-Out test, which detects a signature of autoantibody reactivity that distinguishes healthy subjects from SLE patients with 94% sensitivity, 75% specificity and 93% negative predictive value; thus, an individual manifesting a positive Rule-Out test score is unlikely to have SLE (e.g. lupus is excluded). The objective of this current study was to evaluate the stability of the lupus-associated signature over time. Methods We used banked serum samples from healthy subjects (n = 51) and lupus patients (n = 50 individual samples and n = 181 paired samples, for a total of n = 412 serum samples). The samples were drawn at different times after diagnosis to analyse the impact on the SLE-key Rule-Out test of time elapsed since diagnosis and any changes in disease activity (as reflected by the SLEDAI score). Results The SLE signature remains stable for the first 10 years after diagnosis; in this time frame, <10% of patients manifested a positive Rule-Out score and the SLE-key Rule-Out score was independent of the underlying disease activity as reflected by the SLEDAI score. After ⩾10 years, ∼30% of lupus subjects scored as SLE Ruled-Out; the proportion of patients manifesting this status was even greater in the subset of individuals with a SLEDAI score of 0. Conclusion These findings raise the possibility that a significant number of SLE patients manifest a change in their serological signature over time, and that such a signature change may signify an evolution in the immunological features of their disease relevant to patient management.
Collapse
Affiliation(s)
- Chaim Putterman
- The Division of Rheumatology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY
| | - David S Pisetsky
- Durham VA Medical Center, Duke University Medical Center and Medical Research Service, Durham, NC
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Alan H B Wu
- Department of Laboratory Medicine, San Francisco General Hospital, San Francisco, CA
| | - Ignacio Sanz
- Division of Rheumatology, Emory University School of Medicine, Atlanta, GA
| | - Jim C Oates
- Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC
| | | | | | | | | | | | - Irun R Cohen
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
15
|
Fagúndez P, Brañas G, Cairoli E, Laíz J, Tosar JP. An electrochemical biosensor for rapid detection of anti-dsDNA antibodies in absolute scale. Analyst 2018; 143:3874-3882. [DOI: 10.1039/c8an00020d] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The authors report an electrochemical biosensor enabling fast (30 min) detection of anti-DNA antibodies in serum with reduced manipulation steps.
Collapse
Affiliation(s)
- Pablo Fagúndez
- Analytical Biochemistry Unit
- Nuclear Research Center
- Faculty of Science
- Universidad de la República
- Montevideo 11400
| | - Gustavo Brañas
- Analytical Biochemistry Unit
- Nuclear Research Center
- Faculty of Science
- Universidad de la República
- Montevideo 11400
| | - Ernesto Cairoli
- Systemic Autoimmune Diseases Unit
- Clínica Médica C
- Hospital de Clínicas
- Universidad de la República
- Montevideo 11600
| | - Justo Laíz
- Analytical Biochemistry Unit
- Nuclear Research Center
- Faculty of Science
- Universidad de la República
- Montevideo 11400
| | - Juan Pablo Tosar
- Analytical Biochemistry Unit
- Nuclear Research Center
- Faculty of Science
- Universidad de la República
- Montevideo 11400
| |
Collapse
|
16
|
Wu C, Zhao Y, Lin Y, Yang X, Yan M, Min Y, Pan Z, Xia S, Shao Q. Bioinformatics analysis of differentially expressed gene profiles associated with systemic lupus erythematosus. Mol Med Rep 2017; 17:3591-3598. [PMID: 29257335 PMCID: PMC5802164 DOI: 10.3892/mmr.2017.8293] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/17/2017] [Indexed: 12/20/2022] Open
Abstract
DNA microarray and high-throughput sequencing have been widely used to identify the differentially expressed genes (DEGs) in systemic lupus erythematosus (SLE). However, the big data from gene microarrays are also challenging to work with in terms of analysis and processing. The presents study combined data from the microarray expression profile (GSE65391) and bioinformatics analysis to identify the key genes and cellular pathways in SLE. Gene ontology (GO) and cellular pathway enrichment analyses of DEGs were performed to investigate significantly enriched pathways. A protein-protein interaction network was constructed to determine the key genes in the occurrence and development of SLE. A total of 310 DEGs were identified in SLE, including 193 upregulated genes and 117 downregulated genes. GO analysis revealed that the most significant biological process of DEGs was immune system process. Kyoto Encyclopedia of Genes and Genome pathway analysis showed that these DEGs were enriched in signaling pathways associated with the immune system, including the RIG-I-like receptor signaling pathway, intestinal immune network for IgA production, antigen processing and presentation and the toll-like receptor signaling pathway. The current study screened the top 10 genes with higher degrees as hub genes, which included 2′-5′-oligoadenylate synthetase 1, MX dynamin like GTPase 2, interferon induced protein with tetratricopeptide repeats 1, interferon regulatory factor 7, interferon induced with helicase C domain 1, signal transducer and activator of transcription 1, ISG15 ubiquitin-like modifier, DExD/H-box helicase 58, interferon induced protein with tetratricopeptide repeats 3 and 2′-5′-oligoadenylate synthetase 2. Module analysis revealed that these hub genes were also involved in the RIG-I-like receptor signaling, cytosolic DNA-sensing, toll-like receptor signaling and ribosome biogenesis pathways. In addition, these hub genes, from different probe sets, exhibited significant co-expressed tendency in multi-experiment microarray datasets (P<0.01). In conclusion, these key genes and cellular pathways may improve the current understanding of the underlying mechanism of development of SLE. These key genes may be potential biomarkers of diagnosis, therapy and prognosis for SLE.
Collapse
Affiliation(s)
- Chengjiang Wu
- Department of Immunology, Key Laboratory for Laboratory Medicine of Jiangsu Province, Jiangsu University Medical School, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yangjing Zhao
- Department of Immunology, Key Laboratory for Laboratory Medicine of Jiangsu Province, Jiangsu University Medical School, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yu Lin
- Center for Computational Science, University of Miami, Coral Gables, FL 33146, USA
| | - Xinxin Yang
- Department of Immunology, Key Laboratory for Laboratory Medicine of Jiangsu Province, Jiangsu University Medical School, Zhenjiang, Jiangsu 212013, P.R. China
| | - Meina Yan
- Department of Immunology, Key Laboratory for Laboratory Medicine of Jiangsu Province, Jiangsu University Medical School, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yujiao Min
- Department of Immunology, Key Laboratory for Laboratory Medicine of Jiangsu Province, Jiangsu University Medical School, Zhenjiang, Jiangsu 212013, P.R. China
| | - Zihui Pan
- Department of Immunology, Key Laboratory for Laboratory Medicine of Jiangsu Province, Jiangsu University Medical School, Zhenjiang, Jiangsu 212013, P.R. China
| | - Sheng Xia
- Department of Immunology, Key Laboratory for Laboratory Medicine of Jiangsu Province, Jiangsu University Medical School, Zhenjiang, Jiangsu 212013, P.R. China
| | - Qixiang Shao
- Department of Immunology, Key Laboratory for Laboratory Medicine of Jiangsu Province, Jiangsu University Medical School, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
17
|
van den Broek T, Madi A, Delemarre EM, Schadenberg AWL, Tesselaar K, Borghans JAM, Nierkens S, Redegeld FA, Otten HG, Rossetti M, Albani S, Sorek R, Cohen IR, Jansen NJG, van Wijk F. Human neonatal thymectomy induces altered B-cell responses and autoreactivity. Eur J Immunol 2017; 47:1970-1981. [PMID: 28691750 PMCID: PMC5697610 DOI: 10.1002/eji.201746971] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/06/2017] [Accepted: 06/07/2017] [Indexed: 01/19/2023]
Abstract
An association between T‐cell lymphopenia and autoimmunity has long been proposed, but it remains to be elucidated whether T‐cell lymphopenia affects B‐cell responses to autoantigens. Human neonatal thymectomy (Tx) results in a decrease in T‐cell numbers and we used this model to study the development of autoreactivity. Two cohorts of neonatally thymectomized individuals were examined, a cohort of young (1–5 years post‐Tx, n = 10–27) and older children (>10 years, n = 26), and compared to healthy age‐matched controls. T‐cell and B‐cell subsets were assessed and autoantibody profiling performed. Early post‐Tx, a decrease in T‐cell numbers (2.75 × 109/L vs. 0.71 × 109/L) and an increased proportion of memory T cells (19.72 vs. 57.43%) were observed. The presence of autoantibodies was correlated with an increased proportion of memory T cells in thymectomized children. No differences were seen in percentages of different B‐cell subsets between the groups. The autoantigen microarray showed a skewed autoantibody response after Tx. In the cohort of older individuals, autoantibodies were present in 62% of the thymectomized children, while they were found in only 33% of the healthy controls. Overall, our data suggest that neonatal Tx skews the autoantibody profile. Preferential expansion and preservation of Treg (regulatory T) cell stability and function, may contribute to preventing autoimmune disease development after Tx.
Collapse
Affiliation(s)
- Theo van den Broek
- Laboratory of Translational Immunology, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - Asaf Madi
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Eveline M Delemarre
- Laboratory of Translational Immunology, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - Alvin W L Schadenberg
- Laboratory of Translational Immunology, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht, The Netherlands.,Department of Pediatric Intensive Care, Bristol Royal Hospital for Children, Bristol, UK
| | - Kiki Tesselaar
- Laboratory of Translational Immunology, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - José A M Borghans
- Laboratory of Translational Immunology, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - Stefan Nierkens
- Laboratory of Translational Immunology, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - Frank A Redegeld
- Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Henny G Otten
- Laboratory of Translational Immunology, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - Maura Rossetti
- Duke-National University of Singapore Graduate Medical School, Singapore.,SingHealth Translational Immunology and Inflammation Centre, SingHealth, Singapore
| | - Salvatore Albani
- Duke-National University of Singapore Graduate Medical School, Singapore.,SingHealth Translational Immunology and Inflammation Centre, SingHealth, Singapore
| | | | - Irun R Cohen
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Nicolaas J G Jansen
- Department of Pediatric Intensive Care, University Medical Centre Utrecht, Utrecht, The Netherlands.,Department of Pediatric Cardiothoracic Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Femke van Wijk
- Laboratory of Translational Immunology, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht, The Netherlands
| |
Collapse
|
18
|
Olsen NJ, Choi MY, Fritzler MJ. Emerging technologies in autoantibody testing for rheumatic diseases. Arthritis Res Ther 2017; 19:172. [PMID: 28738887 PMCID: PMC5525353 DOI: 10.1186/s13075-017-1380-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Testing for the presence of antinuclear antibodies (ANAs) is a key step in the diagnosis of systemic lupus erythematosus (SLE) and other systemic autoimmune rheumatic diseases (SARD). The standard slide-based indirect immunofluorescence (IIF) test is widely used, but is limited by a relative lack of specificity for SLE and not all SARD-ANAs are detected. Alternative immunoassays that might offer enhanced diagnostic and prognostic information have evolved, and some of these have entered clinical practice. This review summarizes the current state of ANA testing and multiplex techniques for detecting other autoantibodies, the possibility of point-of-care testing, and approaches for applications in early disease stages.
Collapse
Affiliation(s)
- Nancy J Olsen
- Penn State M.S. Hershey Medical Center, 500 University Drive, Hershey, PA, 17033, USA.
| | - May Y Choi
- Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada
| | - Marvin J Fritzler
- Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada
| |
Collapse
|
19
|
Abstract
BACKGROUND Inflammatory bowel disease (IBD), composed of Crohn's disease (CD) and ulcerative colitis (UC), is an inflammatory autoimmune disease. CD99 has been reported to participate in migration of leukocytes and T cell activation. However, the roles of CD99 in IBD are obscure. MATERIALS AND METHODS CD99 expression was examined in peripheral blood mononuclear cells (PBMCs) and inflamed mucosa from IBD patients by qRT-PCR. Serum TNF-α and IL-17A levels were detected by ELISA. Correlations of CD99 expression with TNF-α, IL-17A, Crohn's disease activity index (CDAI), simple endoscopic score for CD (SES-CD), Mayo index, and Truelove grading were performed by Pearson's correlation. RESULTS CD99 expression was increased in PBMCs and inflamed mucosa from active CD and UC patients, and CD99 expression was also increased in the inflamed mucosa compared with unaffected control from the same patients. Serum TNF-α and IL-17A levels were increased in active CD or UC patients, and positively correlated with CD99 expression in PBMCs (CD: r = .402, p = .009; r = .350, p = .025. UC: r = .289, p = .028; r = .322, p = .014). Moreover, CD99 expression in inflamed mucosa was correlated with CDAI, SES-CD, Mayo index, and Truelove grading (r = .410, p = .012; r = .341, p = .005; r = .366, p = .002; r = .312, p = .011). CONCLUSION CD99 expression is increased in patients with active IBD, and positively correlated with disease activity. Therefore, CD99 expression can be used as an index to evaluate the activity of IBD.
Collapse
Affiliation(s)
- Guangxi Zhou
- a Department of Gastroenterology , The Shanghai Tenth People's Hospital, Tongji University , Shanghai , China
| | - Wenjing Yang
- a Department of Gastroenterology , The Shanghai Tenth People's Hospital, Tongji University , Shanghai , China
| | - Lin Yu
- a Department of Gastroenterology , The Shanghai Tenth People's Hospital, Tongji University , Shanghai , China
| | - Tianming Yu
- a Department of Gastroenterology , The Shanghai Tenth People's Hospital, Tongji University , Shanghai , China
| | - Zhanju Liu
- a Department of Gastroenterology , The Shanghai Tenth People's Hospital, Tongji University , Shanghai , China
| |
Collapse
|
20
|
Devarapu SK, Lorenz G, Kulkarni OP, Anders HJ, Mulay SR. Cellular and Molecular Mechanisms of Autoimmunity and Lupus Nephritis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 332:43-154. [PMID: 28526137 DOI: 10.1016/bs.ircmb.2016.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Autoimmunity involves immune responses directed against self, which are a result of defective self/foreign distinction of the immune system, leading to proliferation of self-reactive lymphocytes, and is characterized by systemic, as well as tissue-specific, inflammation. Numerous mechanisms operate to ensure the immune tolerance to self-antigens. However, monogenetic defects or genetic variants that weaken immune tolerance render susceptibility to the loss of immune tolerance, which is further triggered by environmental factors. In this review, we discuss the phenomenon of immune tolerance, genetic and environmental factors that influence the immune tolerance, factors that induce autoimmunity such as epigenetic and transcription factors, neutrophil extracellular trap formation, extracellular vesicles, ion channels, and lipid mediators, as well as costimulatory or coinhibitory molecules that contribute to an autoimmune response. Further, we discuss the cellular and molecular mechanisms of autoimmune tissue injury and inflammation during systemic lupus erythematosus and lupus nephritis.
Collapse
Affiliation(s)
- S K Devarapu
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - G Lorenz
- Klinikum rechts der Isar, Abteilung für Nephrologie, Technische Universität München, Munich, Germany
| | | | - H-J Anders
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - S R Mulay
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany.
| |
Collapse
|
21
|
Budde P, Zucht HD, Vordenbäumen S, Goehler H, Fischer-Betz R, Gamer M, Marquart K, Rengers P, Richter J, Lueking A, Schulz-Knappe P, Schneider M. Multiparametric detection of autoantibodies in systemic lupus erythematosus. Lupus 2016; 25:812-22. [DOI: 10.1177/0961203316641770] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous disease with respect to disease manifestations, disease progression and treatment response. Therefore, strategies to identify biomarkers that help distinguishing SLE subgroups are a major focus of biomarker research. We reasoned that a multiparametric autoantibody profiling approach combined with data mining tools could be applied to identify SLE patient clusters. We used a bead-based array containing 86 antigens including diverse nuclear and immune defense pathway proteins. Sixty-four autoantibodies were significantly ( p < 0.05) increased in SLE ( n = 69) compared to healthy controls (HC, n = 59). Using binary cut-off thresholds (95% quantile of HC), hierarchical clustering of SLE patients yields five clusters, which differ qualitatively and in their total number of autoantibodies. In two patient clusters the overall accumulated autoantibody reactivity of all antigens tested was 31% and 48%, respectively. We observed a positive association between the autoantibody signature present in these two patient clusters and the clinical manifestation of glomerulonephritis (GLMN). In addition, groups of autoantibodies directed against distinct intracellular compartments and/or biological motifs characterize the different SLE subgroups. Our findings highlight the relevant potential of multiparametric autoantibody detection and may contribute to a deeper understanding of the clinical and serological diversity of SLE.
Collapse
Affiliation(s)
| | | | - S Vordenbäumen
- Heinrich-Heine-University, Medical Faculty, Policlinic of Rheumatology, Düsseldorf, Germany
| | | | - R Fischer-Betz
- Heinrich-Heine-University, Medical Faculty, Policlinic of Rheumatology, Düsseldorf, Germany
| | | | | | | | - J Richter
- Heinrich-Heine-University, Medical Faculty, Policlinic of Rheumatology, Düsseldorf, Germany
| | | | | | - M Schneider
- Heinrich-Heine-University, Medical Faculty, Policlinic of Rheumatology, Düsseldorf, Germany
| |
Collapse
|
22
|
Abstract
Autoantibodies are a key component for the diagnosis, prognosis and monitoring of various diseases. In order to discover novel autoantibody targets, highly multiplexed assays based on antigen arrays hold a great potential and provide possibilities to analyze hundreds of body fluid samples for their reactivity pattern against thousands of antigens in parallel. Here, we provide an overview of the available technologies for producing antigen arrays, highlight some of the technical and methodological considerations and discuss their applications as discovery tools. Together with recent studies utilizing antigen arrays, we give an overview on how the different types of antigen arrays have and will continue to deliver novel insights into autoimmune diseases among several others.
Collapse
|
23
|
Putterman C, Wu A, Reiner-Benaim A, Batty DS, Sanz I, Oates J, Jakobi K, Petri M, Safer P, Gerwien R, Sorek R, Blumenstein Y, Cohen IR. SLE-key(®) rule-out serologic test for excluding the diagnosis of systemic lupus erythematosus: Developing the ImmunArray iCHIP(®). J Immunol Methods 2016; 429:1-6. [PMID: 26678162 PMCID: PMC4824168 DOI: 10.1016/j.jim.2015.12.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 12/03/2015] [Accepted: 12/07/2015] [Indexed: 01/10/2023]
Abstract
We describe here the development, verification and validation of the SLE-key(®) rule-out test for a definitive rule-out of a diagnosis of systemic lupus erythematosus (SLE). The test uses the proprietary iCHIP(®) micro-array technology platform (Fattal et al., 2010) to identify discriminating patterns of circulating autoantibodies among SLE patients compared with self-declared healthy individuals. Given the challenges associated with the diagnosis of SLE and the healthcare costs of delayed diagnosis and misdiagnosis, a definitive rule-out test can provide significant clinical benefits to patients and potentially major cost savings to healthcare systems.
Collapse
Affiliation(s)
- Chaim Putterman
- Division of Rheumatology, Albert Einstein College of Medicine, NY, United States.
| | - Alan Wu
- Department of Laboratory Medicine, San Francisco General Hospital, CA, United States
| | - Anat Reiner-Benaim
- ImmunArray Ltd., Rehovot, Israel; Department of Statistics, University of Haifa, Haifa, Israel
| | | | - Ignacio Sanz
- Division of Rheumatology, Emory University School of Medicine, GA, United States
| | - Jim Oates
- Division of Rheumatology & Immunology, Medical University of South Carolina, SC, United States
| | | | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, MD, United States
| | | | | | | | | | | |
Collapse
|
24
|
Hinchliffe TE, Lin ZT, Wu T. Protein arrays for biomarker discovery in lupus. Proteomics Clin Appl 2016; 10:625-34. [PMID: 26684273 DOI: 10.1002/prca.201500060] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 11/05/2015] [Accepted: 12/09/2015] [Indexed: 11/08/2022]
Abstract
Lupus is one of the most common autoimmune diseases, yet many mechanisms of its pathogenesis are not fully known. Over the last few years, advances in protein array technology have accelerated rapidly, resulting in many promising insights toward the discovery of novel lupus biomarkers that may become useful in disease diagnosis and management. Still, only two types of analytical protein arrays thus far, being antibody and antigen arrays, have found notable usage toward lupus biomarker discovery. In this review, we summarize current protein array technologies being used for biomarker discoveries in lupus and associated biomarker findings, as well as protein arrays that are likely to be used for lupus biomarker discovery in the near future.
Collapse
Affiliation(s)
- Taylor E Hinchliffe
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Zuan-Tao Lin
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Tianfu Wu
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| |
Collapse
|
25
|
Autoantigen Microarray for High-throughput Autoantibody Profiling in Systemic Lupus Erythematosus. GENOMICS PROTEOMICS & BIOINFORMATICS 2015; 13:210-8. [PMID: 26415621 PMCID: PMC4610965 DOI: 10.1016/j.gpb.2015.09.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 09/21/2015] [Accepted: 09/23/2015] [Indexed: 12/19/2022]
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease characterized by the production of autoantibodies to a broad range of self-antigens. Profiling the autoantibody repertoire using array-based technology has emerged as a powerful tool for the identification of biomarkers in SLE and other autoimmune diseases. Proteomic microarray has the capacity to hold large number of self-antigens on a solid surface and serve as a high-throughput screening method for the determination of autoantibody specificities. The autoantigen arrays carrying a wide variety of self-antigens, such as cell nuclear components (nucleic acids and associated proteins), cytoplasmic proteins, phospholipid proteins, cell matrix proteins, mucosal/secreted proteins, glomeruli, and other tissue-specific proteins, have been used for screening of autoantibody specificities associated with different manifestations of SLE. Arrays containing synthetic peptides and molecular modified proteins are also being utilized for identification of autoantibodies targeting to special antigenic epitopes. Different isotypes of autoantibodies, including IgG, IgM, IgA, and IgE, as well as other Ig subtypes, can be detected simultaneously with multi-color labeled secondary antibodies. Serum and plasma are the most common biologic materials for autoantibody detection, but other body fluids such as cerebrospinal fluid, synovial fluid, and saliva can also be a source of autoantibody detection. Proteomic microarray as a multiplexed high-throughput screening platform is playing an increasingly-important role in autoantibody diagnostics. In this article, we highlight the use of autoantigen microarrays for autoantibody exploration in SLE.
Collapse
|
26
|
Fattal I, Shental N, Ben-Dor S, Molad Y, Gabrielli A, Pokroy-Shapira E, Oren S, Livneh A, Langevitz P, Zandman-Goddard G, Sarig O, Margalit R, Gafter U, Domany E, Cohen IR. Guanine polynucleotides are self-antigens for human natural autoantibodies and are significantly reduced in the human genome. Immunology 2015; 146:401-10. [PMID: 26227667 DOI: 10.1111/imm.12514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 07/16/2015] [Accepted: 07/21/2015] [Indexed: 11/28/2022] Open
Abstract
In the course of investigating anti-DNA autoantibodies, we examined IgM and IgG antibodies to poly-G and other oligonucleotides in the sera of healthy persons and those diagnosed with systemic lupus erythematosus (SLE), scleroderma (SSc), or pemphigus vulgaris (PV); we used an antigen microarray and informatic analysis. We now report that all of the 135 humans studied, irrespective of health or autoimmune disease, manifested relatively high amounts of IgG antibodies binding to the 20-mer G oligonucleotide (G20); no participants entirely lacked this reactivity. IgG antibodies to homo-nucleotides A20, C20 or T20 were present only in the sera of SLE patients who were positive for antibodies to dsDNA. The prevalence of anti-G20 antibodies led us to survey human, mouse and Drosophila melanogaster (fruit fly) genomes for runs of T20 and G20 or more: runs of T20 appear > 170,000 times compared with only 93 runs of G20 or more in the human genome; of these runs, 40 were close to brain-associated genes. Mouse and fruit fly genomes showed significantly lower T20/G20 ratios than did human genomes. Moreover, sera from both healthy and SLE mice contained relatively little or no anti-G20 antibodies; so natural anti-G20 antibodies appear to be characteristic of humans. These unexpected observations invite investigation of the immune functions of anti-G20 antibodies in human health and disease and of runs of G20 in the human genome.
Collapse
Affiliation(s)
- Ittai Fattal
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel.,Department of Physics of Complex Systems, The Weizmann Institute of Science, Rehovot, Israel.,Department of Nephrology, Rabin Medical Centre, Petach Tikva, Israel
| | - Noam Shental
- Department of Computer Science, The Open University of Israel, Raanana, Israel
| | - Shifra Ben-Dor
- Bioinformatics and Biological Computing Unit, The Weizmann Institute of Science, Rehovot, Israel
| | - Yair Molad
- The Rheumatology Unit, Rabin Medical Centre, Beilinson Hospital, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Armando Gabrielli
- Istituto di, Dipartimento di Scienze Cliniche e Molecolari- Clinica Medica, Università Politecnica delle Marche, Ancona, Italy
| | - Elisheva Pokroy-Shapira
- The Rheumatology Unit, Rabin Medical Centre, Beilinson Hospital, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shirly Oren
- The Rheumatology Unit, Rabin Medical Centre, Beilinson Hospital, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Avi Livneh
- Department of Medicine F, Sheba Medical Centre, Tel Hashomer, Israel
| | - Pnina Langevitz
- The Rheumatology Unit, Sheba Medical Centre, Tel Hashomer, Israel
| | | | - Ofer Sarig
- Department of Dermatology, Tel Aviv Sourasky Medical Centre, Tel Aviv, Israel
| | - Raanan Margalit
- Science in Action Ltd, Weizmann Science Park, Ness-Ziona, Israel
| | - Uzi Gafter
- Department of Nephrology, Rabin Medical Centre, Petach Tikva, Israel
| | - Eytan Domany
- Department of Physics of Complex Systems, The Weizmann Institute of Science, Rehovot, Israel
| | - Irun R Cohen
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
27
|
Neutrophil extracellular traps (Nets) impact upon autoimmune disorders. Cent Eur J Immunol 2015; 40:217-24. [PMID: 26557037 PMCID: PMC4637397 DOI: 10.5114/ceji.2015.52836] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 06/10/2015] [Indexed: 12/16/2022] Open
Abstract
Friend or foe? This is often asked question when it comes to neutrophil extracellular traps studies. There is no simple answer to that. At the time of their discovery they were considered to be protectors of our well-being. Excellent pathogen fighting skills were described as purely beneficial. But it was not long before those guardians of immunity reveal their dark side. What seemed to be profitable could also take its toll. They are perfectly constructed, made from nucleic deoxyribonucleic acid ornamented with cytoplasmic and granular proteins, to fight invaders. But this unique build is prone to become considered by our body as a threat. Since there is a thin line which when crossed turns a savior into enemy, it was postulated that Nets can play a significant role in autoimmune disorders pathogenesis and disease exacerbation. Recent years have brought a new insight into autoimmune disorders trying to connect the old knowledge and suspicions with modern discoveries.
Collapse
|
28
|
Anti-alpha-actinin antibodies are part of the anti-cell membrane antibody spectrum that characterize patients with lupus nephritis. J Autoimmun 2015; 61:54-61. [DOI: 10.1016/j.jaut.2015.05.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 05/19/2015] [Accepted: 05/21/2015] [Indexed: 01/01/2023]
|
29
|
Hamilton JA, Li J, Wu Q, Yang P, Luo B, Li H, Bradley JE, Taylor JJ, Randall TD, Mountz JD, Hsu HC. General Approach for Tetramer-Based Identification of Autoantigen-Reactive B Cells: Characterization of La- and snRNP-Reactive B Cells in Autoimmune BXD2 Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 194:5022-34. [PMID: 25888644 PMCID: PMC4417409 DOI: 10.4049/jimmunol.1402335] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 03/15/2015] [Indexed: 11/19/2022]
Abstract
Autoreactive B cells are associated with the development of several autoimmune diseases, including systemic lupus erythematosus and rheumatoid arthritis. The low frequency of these cells represents a major barrier to their analysis. Ag tetramers prepared from linear epitopes represent a promising strategy for the identification of small subsets of Ag-reactive immune cells. This is challenging given the requirement for identification and validation of linear epitopes and the complexity of autoantibody responses, including the broad spectrum of autoantibody specificities and the contribution of isotype to pathogenicity. Therefore, we tested a two-tiered peptide microarray approach, coupled with epitope mapping of known autoantigens, to identify and characterize autoepitopes using the BXD2 autoimmune mouse model. Microarray results were verified through comparison with established age-associated profiles of autoantigen specificities and autoantibody class switching in BXD2 and control (C57BL/6) mice and high-throughput ELISA and ELISPOT analyses of synthetic peptides. Tetramers were prepared from two linear peptides derived from two RNA-binding proteins (RBPs): lupus La and 70-kDa U1 small nuclear ribonucleoprotein. Flow cytometric analysis of tetramer-reactive B cell subsets revealed a significantly higher frequency and greater numbers of RBP-reactive marginal zone precursor, transitional T3, and PDL-2(+)CD80(+) memory B cells, with significantly elevated CD69 and CD86 observed in RBP(+) marginal zone precursor B cells in the spleens of BXD2 mice compared with C57BL/6 mice, suggesting a regulatory defect. This study establishes a feasible strategy for the characterization of autoantigen-specific B cell subsets in different models of autoimmunity and, potentially, in humans.
Collapse
Affiliation(s)
- Jennie A Hamilton
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jun Li
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Qi Wu
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - PingAr Yang
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Bao Luo
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Hao Li
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - John E Bradley
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Justin J Taylor
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109; and
| | - Troy D Randall
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - John D Mountz
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294; Department of Medicine, Birmingham, Alabama VA Medical Center, Birmingham, AL 35233
| | - Hui-Chen Hsu
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294;
| |
Collapse
|
30
|
Soriano A, Shoenfeld Y. Haemophagocytic lymphohistiocytosis: from diagnostic challenges to predictive possibilities. Lupus 2015; 24:655-8. [PMID: 25701564 DOI: 10.1177/0961203315572719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 01/19/2015] [Indexed: 11/16/2022]
Affiliation(s)
- A Soriano
- Department of Clinical Medicine and Rheumatology, Campus Bio-Medico University Hospital and School of Medicine, Rome, Italy Zabludowicz Center for Autoimmune Diseases, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Y Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Chaim Sheba Medical Center, Tel Hashomer, Israel Incumbent of the Laura Schwarz-Kipp Chair for Research of Autoimmune Diseases, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|
31
|
Prechl J, Czirják L. The endothelial deprotection hypothesis for lupus pathogenesis: the dual role of C1q as a mediator of clearance and regulator of endothelial permeability. F1000Res 2015; 4:24. [DOI: 10.12688/f1000research.6075.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/26/2015] [Indexed: 11/20/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous multifactorial systemic autoimmune disease affecting several organs. SLE can start relatively early in life and results in impaired quality of life and shortened life expectancy because of a gradual disease progression leading to cardiovascular, renal and neoplastic disease. The basic mechanisms of the pathogenesis of the disease still remain to be clarified. It is clear that complement proteins play a key and complex role in the development of SLE. Complement component C1q has been known to be a fundamental component of lupus development, but most explanations focus on its role in apoptotic debris removal. Importantly, C1q was recently found to play a key role in the maintenance of vascular endothelial integrity.We suggest that apoptotic products, endothelial cells and extracellular matrix components, which display negatively charged moieties, compete for binding to molecules of the innate humoral immune response, like C1q. Genetic or acquired factors leading to an increased load of apoptotic cell debris and decrease or absence of C1q therefore interfere with the regulation of endothelial permeability and integrity. Furthermore, we suggest that lupus is the net result of an imbalance between the two functions of immune clearance and vascular endothelial integrity maintenance, an imbalance triggered and sustained by autoimmunity, which skews C1q consumption by IgG-mediated complement classical pathway activation on autoantigens. In this triangle of innate clearance, autoimmunity and endothelial integrity, C1q plays a central role.Hence, we interpret the pathogenesis of lupus by identifying three key components, namely innate immune clearance, autoimmunity and endothelial integrity and we establish a link between these components based on the protective role that innate clearance molecules play in endothelial renewal. By including the vasoprotective role of C1q in the interpretation of SLE development we attempt to provide novel explanations for the symptoms, organ damage, diagnostic and therapeutic difficulties of the disease.
Collapse
|
32
|
Prechl J, Czirják L. The endothelial deprotection hypothesis for lupus pathogenesis: the dual role of C1q as a mediator of clearance and regulator of endothelial permeability. F1000Res 2015; 4:24. [PMID: 25901277 PMCID: PMC4392829 DOI: 10.12688/f1000research.6075.2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/08/2015] [Indexed: 12/31/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous multifactorial systemic autoimmune disease affecting several organs. SLE can start relatively early in life and results in impaired quality of life and shortened life expectancy because of a gradual disease progression leading to cardiovascular, renal and neoplastic disease. The basic mechanisms of the pathogenesis of the disease still remain to be clarified. It is clear that complement proteins play a key and complex role in the development of SLE. Complement component C1q has been known to be a fundamental component of lupus development, but most explanations focus on its role in apoptotic debris removal. Importantly, C1q was recently found to play a key role in the maintenance of vascular endothelial integrity. We suggest that apoptotic products, endothelial cells and extracellular matrix components, which display negatively charged moieties, compete for binding to molecules of the innate humoral immune response, like C1q. Genetic or acquired factors leading to an increased load of apoptotic cell debris and decrease or absence of C1q therefore interfere with the regulation of endothelial permeability and integrity. Furthermore, we suggest that lupus is the net result of an imbalance between the two functions of immune clearance and vascular endothelial integrity maintenance, an imbalance triggered and sustained by autoimmunity, which skews C1q consumption by IgG-mediated complement classical pathway activation on autoantigens. In this triangle of innate clearance, autoimmunity and endothelial integrity, C1q plays a central role. Hence, we interpret the pathogenesis of lupus by identifying three key components, namely innate immune clearance, autoimmunity and endothelial integrity and we establish a link between these components based on the protective role that innate clearance molecules play in endothelial renewal. By including the vasoprotective role of C1q in the interpretation of SLE development we attempt to provide novel explanations for the symptoms, organ damage, diagnostic and therapeutic difficulties of the disease.
Collapse
Affiliation(s)
- József Prechl
- Diagnosticum Zrt, Budapest, 1047, Hungary ; MTA-ELTE Immunology Research Group, Budapest, 1117, Hungary
| | - László Czirják
- Department of Rheumatology and Immunology, Clinic Center, University of Pécs, Pécs, 7632, Hungary
| |
Collapse
|
33
|
Devaraju P, Gulati R, Antony PT, Mithun CB, Negi VS. Susceptibility to SLE in South Indian Tamils may be influenced by genetic selection pressure on TLR2 and TLR9 genes. Mol Immunol 2014; 64:123-6. [PMID: 25466615 DOI: 10.1016/j.molimm.2014.11.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Revised: 09/28/2014] [Accepted: 11/06/2014] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is a multisystem autoimmune disorder with complex etiology. Genetics plays an important role in lupus pathogenesis through its influence on clinical and autoantibody phenotype of the disease. Toll like receptors (TLR) recognize molecular patterns of pathogens and activate the innate immune system. Their ability to identify nucleic acids makes them suitable candidates for investigation of their role in lupus pathogenesis. Hence, this study was carried out to analyze the G to A and C to T transitions in TLR2 and TLR9 genes respectively and to test their association with lupus susceptibility, clinical and autoantibody phenotypes in South Indian Tamils. METHOD Three hundred SLE patients fulfilling ACR 2012 criteria for SLE and 460 age, sex similar, ethnicity matched controls were recruited as cases and controls. TLR2 (R753Q) and TLR9 (-1237C/T) polymorphisms were analyzed by real time PCR. RESULTS The TLR2 gene remained monomorphic in patients and controls, the frequency of the homozygous wild type allele being 100% and 99.6% respectively. Hence, it did not confer susceptibility to SLE. The more frequent T allele of TLR9 gene conferred a significant risk to develop SLE (p=0.011, OR 1.69, 95% CI 1.1-2.6). Both the polymorphisms did not influence clinical or autoantibody phenotype of the disease. CONCLUSION Prevailing endemic infections in the Indian subcontinent may have exerted a selection pressure resulting in TLR2 gene remaining monomorphic and the TLR9 adapting to a mutation for its increased expression. These may have an additive effect in the presence of other genetic and environmental risk factors to confer susceptibility to SLE in South Indian Tamils.
Collapse
Affiliation(s)
- Panneer Devaraju
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry 605 006, India
| | - Reena Gulati
- Genetic Services Unit, Department of Pediatrics, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry 605 006, India
| | - Paul T Antony
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry 605 006, India
| | - C B Mithun
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry 605 006, India
| | - Vir S Negi
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry 605 006, India.
| |
Collapse
|
34
|
Selective adsorption of antiphospholipid and anti-dsDNA autoantibodies on histidine based pseudobioaffinity adsorbent from sera of patients with systemic lupus erythematosus (SLE). J Chromatogr B Analyt Technol Biomed Life Sci 2014; 975:77-83. [PMID: 25438246 DOI: 10.1016/j.jchromb.2014.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/29/2014] [Accepted: 11/08/2014] [Indexed: 11/22/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune diseases characterized by the presence of antiphospholipid and anti-dsDNA autoantibodies in the sera of patients. These autoantibodies and their subclasses have received increasing attention by medical community due to their association with recurrent venous thrombosis, fetal loss and thrombocytopenia. In particular, attention has been paid to IgG subclasses in SLE. The biological and functional properties together with the subclass distribution might therefore influence the course of SLE. The separation and elimination of these autoantibodies from sera of patients can be effective in clinical therapy. In the present study, histidine based pseudobioaffinity adsorbents have been used for the selective adsorption and separation of anti-double stranded DNA (anti-dsDNA), anticardiolipin (aCL) and anti-β2-glycoprotein-I (anti-β2-GPI) antibodies from sera of patients with SLE. For this purpose histidine acting as a pseudobiospecific ligand has been coupled to bisoxirane activated sepharose CL-6B for the adsorption and separation of these autoantibodies. The removal of autoantibodies was carried out under gentle adsorption and elution chromatographic conditions at pH values 7.0 and 8.0. Autoantibodies isotypes and subclasses distribution in the separated fractions were studied by enzyme-linked immune-sorbent assay. The obtained results showed that the separated anticardiolipin and anti-β2-glycoprotein-I autoantibodies belong to IgG1, IgG2 and IgG3subclasses, while those of anti-dsDNA belong to IgM isotype and were shown to have a DNA hydrolyzing activity that hydrolyzes plasmid DNA. The results also indicate a total IgM and IgG recovery superior to 90% of the fraction loaded at pH 7.4 and pH 8.0 respectively.
Collapse
|
35
|
Agmon-Levin N, Shoenfeld Y. The spectrum between antiphospholipid syndrome and systemic lupus erythematosus. Clin Rheumatol 2014; 33:293-5. [PMID: 24435353 DOI: 10.1007/s10067-014-2486-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Nancy Agmon-Levin
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, 52621, Israel
| | | |
Collapse
|
36
|
Detection and quantification of lysine acetyl-alteration using antibody microarray. Bioanalysis 2014; 5:2469-80. [PMID: 24138621 DOI: 10.4155/bio.13.191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Lysine acetylation is a reversible and dynamic post-translational modification on proteins, and plays an important role in diverse biological processes. Technological limitations have so far prevented comparative quantification of lysine acetylation in different samples. RESULTS We developed a method to efficiently study lysine acetylation on individual proteins from complex mixtures, using antibody microarrays to capture individual proteins followed by detection with lysine acetyl antibody. By profiling both protein and acetylation variations in multiple samples using this microarray, we found cancer-associated lysine acetylation alteration on VEGF in the serum of hepatocellular carcinoma patients. CONCLUSION Microarrays of lysine acetylation are highly effective for detecting acetylation, and should be useful in identifying and validating disease-associated acetylation alterations as biomarkers under both normal and pathological circumstances.
Collapse
|
37
|
Fattal I, Rimer J, Shental N, Molad Y, Gabrielli A, Livneh A, Sarig O, Goldberg I, Gafter U, Domany E, Cohen IR. Pemphigus vulgaris is characterized by low IgG reactivities to specific self-antigens along with high IgG reactivity to desmoglein 3. Immunology 2014; 143:374-80. [PMID: 24820664 DOI: 10.1111/imm.12316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 04/14/2014] [Accepted: 05/01/2014] [Indexed: 11/28/2022] Open
Abstract
Pemphigus vulgaris (PV) is an autoimmune skin disease, which has been characterized by IgG autoantibodies to desmoglein 3. Here we studied the antibody signatures of PV patients compared with healthy subjects and with patients with two other autoimmune diseases with skin manifestations (systemic lupus erythematosus and scleroderma), using an antigen microarray and informatics analysis. We now report a previously unobserved phenomenon--patients with PV, compared with the healthy subjects and the two other diseases, show a significant decrease in IgG autoantibodies to a specific set of self-antigens. This novel finding demonstrates that an autoimmune disease may be associated with a loss of specific, healthy IgG autoantibodies and not only with a gain of specific, pathogenic IgG autoantibodies.
Collapse
Affiliation(s)
- Ittai Fattal
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel; Department of Physics of Complex Systems, The Weizmann Institute of Science, Rehovot, Israel; Department of Nephrology, Rabin Medical Centre, Petach Tikva, Israel
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Fattal I, Shental N, Molad Y, Gabrielli A, Pokroy-Shapira E, Oren S, Livneh A, Langevitz P, Pauzner R, Sarig O, Gafter U, Domany E, Cohen IR. Epstein-Barr virus antibodies mark systemic lupus erythematosus and scleroderma patients negative for anti-DNA. Immunology 2014; 141:276-85. [PMID: 24164500 DOI: 10.1111/imm.12200] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 10/20/2013] [Accepted: 10/21/2013] [Indexed: 01/01/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that can attack many different body organs; the triggering event is unknown. SLE has been associated with more than 100 different autoantibody reactivities - anti-dsDNA is prominent. Nevertheless, autoantibodies to dsDNA occur in only two-thirds of SLE patients. We previously reported the use of an antigen microarray to characterize SLE serology. We now report the results of an expanded study of serology in SLE patients and scleroderma (SSc) patients compared with healthy controls. The analysis validated and extended previous findings: two-thirds of SLE patients reacted to a large spectrum of self-molecules that overlapped with their reactivity to dsDNA; moreover, some SLE patients manifested a deficiency of natural IgM autoantibodies. Most significant was the finding that many SLE patients who were negative for autoantibodies to dsDNA manifested abnormal antibody responses to Epstein-Barr virus (EBV): these subjects made IgG antibodies to EBV antigens to which healthy subjects did not respond or they failed to make antibodies to EBV antigens to which healthy subjects did respond. This observation suggests that SLE may be associated with a defective immune response to EBV. The SSc patients shared many of these serological abnormalities with SLE patients, but differed from them in increased IgG autoantibodies to topoisomerase and centromere B; 84% of SLE patients and 58% of SSc patients could be detected by their abnormal antibodies to EBV. Hence an aberrant immune response to a ubiquitous viral infection such as EBV might set the stage for an autoimmune disease.
Collapse
Affiliation(s)
- Ittai Fattal
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel; Department of Physics of Complex Systems, The Weizmann Institute of Science, Rehovot, Israel; Department of Nephrology, Rabin Medical Centre, Petach Tikva, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sivakumar PM, Moritsugu N, Obuse S, Isoshima T, Tashiro H, Ito Y. Novel microarrays for simultaneous serodiagnosis of multiple antiviral antibodies. PLoS One 2013; 8:e81726. [PMID: 24367491 PMCID: PMC3867344 DOI: 10.1371/journal.pone.0081726] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 10/07/2013] [Indexed: 11/19/2022] Open
Abstract
We developed an automated diagnostic system for the detection of virus-specific immunoglobulin Gs (IgGs) that was based on a microarray platform. We compared efficacies of our automated system with conventional enzyme immunoassays (EIAs). Viruses were immobilized to microarrays using a radical cross-linking reaction that was induced by photo-irradiation. A new photoreactive polymer containing perfluorophenyl azide (PFPA) and poly(ethylene glycol) methacrylate was prepared and coated on plates. Inactivated measles, rubella, mumps, Varicella-Zoster and recombinant Epstein-Barr viruse antigen were added to coated plates, and irradiated with ultraviolet light to facilitate immobilization. Virus-specific IgGs in healthy human sera were assayed using these prepared microarrays and the results obtained compared with those from conventional EIAs. We observed high correlation (0.79-0.96) in the results between the automated microarray technique and EIAs. The microarray-based assay was more rapid, involved less reagents and sample, and was easier to conduct compared with conventional EIA techniques. The automated microarray system was further improved by introducing reagent storage reservoirs inside the chamber, thereby conserving the use of expensive reagents and antibodies. We considered the microarray format to be suitable for rapid and multiple serological diagnoses of viral diseases that could be developed further for clinical applications.
Collapse
Affiliation(s)
| | - Nozomi Moritsugu
- Nano Medical Engineering Laboratory, RIKEN, Wako, Saitama, Japan
| | - Sei Obuse
- Nano Medical Engineering Laboratory, RIKEN, Wako, Saitama, Japan
| | - Takashi Isoshima
- Nano Medical Engineering Laboratory, RIKEN, Wako, Saitama, Japan
| | - Hideo Tashiro
- Nano Medical Engineering Laboratory, RIKEN, Wako, Saitama, Japan
- Consonal Biotechnologies Co., Ltd., Funabashi, Chiba, Japan
| | - Yoshihiro Ito
- Nano Medical Engineering Laboratory, RIKEN, Wako, Saitama, Japan
- Consonal Biotechnologies Co., Ltd., Funabashi, Chiba, Japan
- Emergent Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science, Wako, Saitama, Japan
| |
Collapse
|
40
|
Arriens C, Mohan C. Systemic lupus erythematosus diagnostics in the 'omics' era. ACTA ACUST UNITED AC 2013; 8:671-687. [PMID: 24860621 DOI: 10.2217/ijr.13.59] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Systemic lupus erythematosus is a complex autoimmune disease affecting multiple organ systems. Currently, diagnosis relies upon meeting at least four out of eleven criteria outlined by the ACR. The scientific community actively pursues discovery of novel diagnostics in the hope of better identifying susceptible individuals in early stages of disease. Comprehensive studies have been conducted at multiple biological levels including: DNA (or genomics), mRNA (or transcriptomics), protein (or proteomics) and metabolites (or metabolomics). The 'omics' platforms allow us to re-examine systemic lupus erythematosus at a greater degree of molecular resolution. More importantly, one is hopeful that these 'omics' platforms may yield newer biomarkers for systemic lupus erythematosus that can help clinicians track the disease course with greater sensitivity and specificity.
Collapse
Affiliation(s)
- Cristina Arriens
- Rheumatic Diseases Division, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8884, USA
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, 3605 Cullen Blvd, Room 2018, Houston, TX 77204, USA
| |
Collapse
|
41
|
The deleterious role of basophils in systemic lupus erythematosus. Curr Opin Immunol 2013; 25:704-11. [PMID: 24209595 DOI: 10.1016/j.coi.2013.10.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 10/02/2013] [Accepted: 10/03/2013] [Indexed: 11/23/2022]
Abstract
Systemic lupus erythematosus is a complex autoimmune disease of multifactorial origins. All compartments of the immune system appear to be affected, at least in some way, and to contribute to disease pathogenesis. Because of an escape from negative selection autoreactive T and B cells accumulate in SLE patients leading to the production of autoantibodies mainly raised against nuclear components and their subsequent deposition into target organs. We recently showed that basophils, in an IgE and IL-4 dependent manner, contribute to SLE pathogenesis by amplifying autoantibody production. Here, we summarize what we have learned about the deleterious role of basophils in lupus both in a mouse model and in SLE patients. We discuss which possible pathways could be involved in basophil activation and recruitment to secondary lymphoid organs during SLE, and how basophils may amplify autoantibody production.
Collapse
|
42
|
An integrated peptide-antigen microarray on plasmonic gold films for sensitive human antibody profiling. PLoS One 2013; 8:e71043. [PMID: 23923050 PMCID: PMC3726620 DOI: 10.1371/journal.pone.0071043] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 06/24/2013] [Indexed: 12/19/2022] Open
Abstract
High-throughput screening for interactions of peptides with a variety of antibody targets could greatly facilitate proteomic analysis for epitope mapping, enzyme profiling, drug discovery and biomarker identification. Peptide microarrays are suited for such undertaking because of their high-throughput capability. However, existing peptide microarrays lack the sensitivity needed for detecting low abundance proteins or low affinity peptide-protein interactions. This work presents a new peptide microarray platform constructed on nanostructured plasmonic gold substrates capable of metal enhanced NIR fluorescence enhancement (NIR-FE) by hundreds of folds for screening peptide-antibody interactions with ultrahigh sensitivity. Further, an integrated histone peptide and whole antigen array is developed on the same plasmonic gold chip for profiling human antibodies in the sera of systemic lupus erythematosus (SLE) patients, revealing that collectively a panel of biomarkers against unmodified and post-translationally modified histone peptides and several whole antigens allow more accurate differentiation of SLE patients from healthy individuals than profiling biomarkers against peptides or whole antigens alone.
Collapse
|
43
|
Cohen IR. Autoantibody repertoires, natural biomarkers, and system controllers. Trends Immunol 2013; 34:620-5. [PMID: 23768955 DOI: 10.1016/j.it.2013.05.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 05/04/2013] [Accepted: 05/14/2013] [Indexed: 12/20/2022]
Abstract
The immune system is composed of networks of interacting cells and molecules; therefore, to understand and control immune behavior we need to adopt the thinking and tools of systems immunology. This review describes the use of an antigen microarray device and informatics to profile the repertoires of autoantibodies in health and disease. Autoantibody profiling provides an insight into the biomarkers used by the immune system in its dialog with the body. Heat shock protein 60 (HSP60) and HSP70 are cited as examples of key hubs in physiological regulatory networks; HSP molecules and peptides can be viewed as natural regulators because the immune system itself deploys them to modulate inflammatory reactions. The discovery of such natural biomarkers paves the way towards natural control.
Collapse
Affiliation(s)
- Irun R Cohen
- Department of Immunology, The Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
44
|
Lin MW, Ho JWK, Harrison LC, dos Remedios CG, Adelstein S. An antibody-based leukocyte-capture microarray for the diagnosis of systemic lupus erythematosus. PLoS One 2013; 8:e58199. [PMID: 23516448 PMCID: PMC3596412 DOI: 10.1371/journal.pone.0058199] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Accepted: 02/01/2013] [Indexed: 12/17/2022] Open
Abstract
The diagnosis of Systemic Lupus Erythematosus (SLE) is challenging due to its heterogeneous clinical presentation and the lack of robust biomarkers to distinguish it from other autoimmune diseases. Further, currently used laboratory tests do not readily distinguish active and inactive disease. Several groups have attempted to apply emerging high throughput profiling technologies to diagnose and monitor SLE. Despite showing promise, many are expensive and technically challenging for routine clinical use. The goal of this work is to develop a better diagnostic and monitoring tool for SLE. We report a highly customisable antibody microarray that consists of a duplicate arrangement of 82 antibodies directed against surface antigens on peripheral blood mononuclear cells (PMBCs). This high-throughput array was used to profile SLE patients (n = 60) with varying disease activity, compared to healthy controls (n = 24), patients with rheumatoid arthritis (n = 25), and other autoimmune diseases (n = 28). We used a computational algorithm to calculate a score from the entire microarray profile and correlated it with SLE disease activity. Our results demonstrate that leukocyte-capture microarray profiles can readily distinguish active SLE patients from healthy controls (AUROC = 0.84). When combined with the standard laboratory tests (serum anti-dsDNA, complements C3 and C4), the microarrays provide significantly increased discrimination. The antibody microarrays can be enhanced by the addition of other markers for potential application to the diagnosis and stratification of SLE, paving the way for the customised and accurate diagnosis and monitoring of SLE.
Collapse
Affiliation(s)
- Ming-Wei Lin
- Department of Clinical Immunology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Discipline of Medicine, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Joshua W. K. Ho
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Leonard C. Harrison
- The Walter and Eliza Hall Institute of Medical Research, Department of Clinical Immunology and Burnet Clinical Research Unit, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Cristobal G. dos Remedios
- Discipline of Anatomy, Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Stephen Adelstein
- Department of Clinical Immunology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Discipline of Medicine, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
45
|
Peretti CS, Peretti CR, Kozora E, Papathanassiou D, Chouinard VA, Chouinard G. Cognitive impairment in systemic lupus erythematosus women with elevated autoantibodies and normal single photon emission computerized tomography. PSYCHOTHERAPY AND PSYCHOSOMATICS 2013; 81:276-85. [PMID: 22832425 DOI: 10.1159/000336555] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 01/13/2012] [Indexed: 11/19/2022]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is known to induce psychiatric disorders, from psychoses to maladaptive coping. Brain autoantibodies were proposed to explain SLE neuropsychiatric disorders and found to be elevated before the onset of clinical symptoms. We assessed cognition in Caucasian SLE women with elevated autoantibodies without overt neuropsychiatric syndromes, in conjunction with single photon emission computerized tomography (SPECT). METHODS 31 women meeting SLE criteria of the American College of Rheumatology (ACR) were included. Patients who met the ACR neuropsychiatric definition were excluded. Matched controls were 23 healthy women from the Champagne-Ardenne region, France. Participants completed neuropsychological and autoantibodies measurements, and 19 completed SPECT. RESULTS 61% (19/31) of women with SLE and 53% (9/17) of those with normal SPECT had significant global cognitive impairment defined as 4 T-scores <40 in cognitive tests, compared to 0% (0/23) of controls. SLE women also had significantly greater cognitive dysfunction (mean T-score) on the Wechsler Adult Intelligence Scale (WAIS) visual backspan, Trail Making Test A and B, WAIS Digit Symbol Substitution Test and Stroop Interference, compared to controls. Elevated antinuclear antibody correlated with impairment in the WAIS visual span, WAIS visual backspan, and cancellation task; elevated anti-double-stranded DNA antibody and anticardiolipin correlated respectively with impairment in the Trail Making Test A and WAIS auditive backspan. Two SLE women had abnormal SPECT. CONCLUSIONS A high prevalence of cognitive deficits was found in Caucasian SLE women compared to normal women, which included impairment in cognitive domains important for daily activities. Elevated autoantibodies tended to correlate with cognitive dysfunction.
Collapse
Affiliation(s)
- Charles-Siegfried Peretti
- Department of Psychiatry, Université Pierre et Marie Curie and Saint-Antoine Hospital, Paris, France. charles.peretti @ sat.aphp.fr
| | | | | | | | | | | |
Collapse
|
46
|
Papp K, Végh P, Hóbor R, Szittner Z, Vokó Z, Podani J, Czirják L, Prechl J. Immune complex signatures of patients with active and inactive SLE revealed by multiplex protein binding analysis on antigen microarrays. PLoS One 2012; 7:e44824. [PMID: 22984570 PMCID: PMC3439431 DOI: 10.1371/journal.pone.0044824] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 08/07/2012] [Indexed: 01/15/2023] Open
Abstract
Systemic lupus erythematosus is characterized by dysfunctional clearance of apoptotic debris and the development of pathogenic autoantibodies. While the complement system is also involved in the disease no attempt has been made to generate a comprehensive view of immune complex formation from various autoantigens. We increased the complexity of autoantibody profiles by measuring the binding of two complement proteins, C3 and C4, in addition to two antibody classes, IgG and IgM, to a collection of autoantigens. These complement components covalently bind to those microarray features where antibodies and other serum components induce complement activation. Using this technology, we compared functional serum antibody profiles of control subjects (n = 31) and patients with lupus erythematosus (n = 61) in the active (n = 22) and inactive (n = 39) phase of the disease. Multivariate analysis was applied to identify contributions of binding data on 25 antigens to the discrimination of the study groups. Receiver operating characteristic analysis was used to portray the discriminative property of each measured parameter for each antigen in pairwise group comparisons. Complement C3 and C4 deposition increased on autoantibody targets in spite of the decreased serum complement concentrations, and decreased on other autoantigens, demonstrating the imbalance of complement function in patients with lupus erythematosus. Our observations confirmed previously known markers of disease and showed that C3 and C4 deposition data were at least as powerful as Ig binding data in separating the study groups.
Collapse
Affiliation(s)
- Krisztián Papp
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
- Diagnosticum Ltd., Budapest, Hungary
| | | | - Renáta Hóbor
- Department of Rheumatology and Immunology, Clinic Center, University of Pécs, Pécs, Hungary
| | - Zoltán Szittner
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
- Diagnosticum Ltd., Budapest, Hungary
| | - Zoltán Vokó
- Department of Health Policy and Health Economics, Eötvös Loránd University, Budapest, Hungary
- Syreon Research Institute, Budapest, Hungary
| | - János Podani
- Department of Plant Systematics, Ecology and Theoretical Biology, Eötvös Loránd University, Budapest, Hungary
| | - László Czirják
- Department of Rheumatology and Immunology, Clinic Center, University of Pécs, Pécs, Hungary
| | - József Prechl
- Diagnosticum Ltd., Budapest, Hungary
- Immunology Research Group of the Hungarian Academy of Sciences, Eötvös Loránd University, Budapest, Hungary
- * E-mail:
| |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW Systemic lupus erythematosus (SLE) is a heterogeneous human disease influenced by a complex interplay of necessary, but not individually sufficient, factors. Although many genetic and environmental factors are associated with SLE, this review will focus on the evolving evidence for key Epstein-Barr virus (EBV)-specific roles in SLE, focusing on new experimental studies published during 2009, 2010, and 2011. RECENT FINDINGS SLE patients have a dysregulated immune response against EBV. EBV antigens exhibit structural molecular mimicry with common SLE antigens and functional molecular mimicry with critical immune-regulatory components. SLE patients, from a number of unique geographic regions, are shown to have higher rates of EBV seroconversion, especially against early EBV antigens, suggesting frequent viral reactivation. SLE patients also have increased EBV viral loads and impaired EBV-specific CD8 cytotoxic T cells, with impaired cytokine responses to EBV in lupus patients. Irregular cytokine production in plasmacytoid dendritic cells and CD69 CD4 T cells after stimulation with EBV has also been demonstrated. SUMMARY Recent advances demonstrate SLE-specific serologic responses, gene expression, viral load, T-cell responses, humoral fine specificity, and molecular mimicry with EBV, further supporting potential roles for EBV in lupus etiology and pathogenesis.
Collapse
|
48
|
Li L, Nukala S, Du Y, Han J, Liu K, Hutcheson J, Pathak S, Li Q, Mohan C. Murine lupus strains differentially model unique facets of human lupus serology. Clin Exp Immunol 2012; 168:178-185. [PMID: 22471278 PMCID: PMC3390518 DOI: 10.1111/j.1365-2249.2012.04568.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2012] [Indexed: 12/01/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a polygenic autoimmune disease characterized by the production of anti-nuclear autoantibodies that lead to subsequent end organ damage. Previous array-based studies in patients with SLE have shown that high immunoglobulin (Ig)G anti-nuclear autoantibody reactivity was associated with severe renal lupus, whereas IgM polyreactivity was associated with less severe disease. To ascertain how different murine lupus strains recapitulate these different autoantibody profiles seen in patients, serum from New Zealand black (NZB)/NZ white (W) F(1), Murphy Roths large (MRL)/lpr, NZ mixed (M)2410 and BXSB strains were compared using a comprehensive array-based screen. The array results were verified using enzyme-linked immunosorbent assays (ELISAs). Serum from MRL/lpr mice exhibited high levels of IgG anti-nuclear antibodies as well as anti-glomerular antibodies and variable levels of antibodies to myosin, Matrigel and thyroglobulin. Elevated anti-nuclear IgG antibodies were associated with severe nephritis in this strain. In contrast, NZM2410 mice exhibited lower IgG autoantibody levels with less severe nephritis but a significantly higher polyreactive IgM autoantibody profile. ELISA analysis confirmed these results. The NZB/NZW F(1) and BXSB strains exhibited an intermediate serological profile. Hence, just as in patients with SLE, whereas strong IgG reactivity to nuclear antigens is associated with severe renal disease, a polyreactive IgM seroprofile is also less ominous in murine lupus.
Collapse
Affiliation(s)
- L Li
- Department of Internal Medicine and Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390-8884, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Gibson DS, Qiu J, Mendoza EA, Barker K, Rooney ME, LaBaer J. Circulating and synovial antibody profiling of juvenile arthritis patients by nucleic acid programmable protein arrays. Arthritis Res Ther 2012; 14:R77. [PMID: 22510425 PMCID: PMC3446451 DOI: 10.1186/ar3800] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Revised: 02/22/2012] [Accepted: 04/17/2012] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Juvenile idiopathic arthritis (JIA) is a heterogeneous disease characterized by chronic joint inflammation of unknown cause in children. JIA is an autoimmune disease and small numbers of autoantibodies have been reported in JIA patients. The identification of antibody markers could improve the existing clinical management of patients. METHODS A pilot study was performed on the application of a high-throughput platform, the nucleic acid programmable protein array (NAPPA), to assess the levels of antibodies present in the systemic circulation and synovial joint of a small cohort of juvenile arthritis patients. Plasma and synovial fluid from 10 JIA patients was screened for antibodies against 768 proteins on NAPPAs. RESULTS Quantitative reproducibility of NAPPAs was demonstrated with > 0.95 intra-array and inter-array correlations. A strong correlation was also observed for the levels of antibodies between plasma and synovial fluid across the study cohort (r = 0.96). Differences in the levels of 18 antibodies were revealed between sample types across all patients. Patients were segregated into two clinical subtypes with distinct antibody signatures by unsupervised hierarchical cluster analysis. CONCLUSION The NAPPAs provide a high-throughput quantitatively reproducible platform to screen for disease-specific autoantibodies at the proteome level on a microscope slide. The strong correlation between the circulating antibody levels and those of the inflamed joint represents a novel finding and provides confidence to use plasma for discovery of autoantibodies in JIA, thus circumventing the challenges associated with joint aspiration. We expect that autoantibody profiling of JIA patients on NAPPAs could yield antibody markers that can act as criteria to stratify patients, predict outcomes and understand disease etiology at the molecular level.
Collapse
Affiliation(s)
- David S Gibson
- Arthritis Research Group, Centre for Infection and Immunity, Health Science Building, Queen's University of Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK.
| | | | | | | | | | | |
Collapse
|
50
|
Tomizaki KY, Obi M, Mihara H. Noncompetitive On-Chip Immunoassays for Detection of Nonlabeled Antibodies Based on the Excluded Volume Effect of the Target Itself. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2012. [DOI: 10.1246/bcsj.20110239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Kin-ya Tomizaki
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology
- Innovative Materials and Processing Research Center, Ryukoku University
- Department of Materials Chemistry, Ryukoku University
| | - Masaki Obi
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology
| | - Hisakazu Mihara
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology
| |
Collapse
|