1
|
Amjadi MF, Parker MH, Adyniec RR, Zheng Z, Robbins AM, Bashar SJ, Denny MF, McCoy SS, Ong IM, Shelef MA. Novel and unique rheumatoid factors cross-react with viral epitopes in COVID-19. J Autoimmun 2024; 142:103132. [PMID: 37956528 PMCID: PMC10957334 DOI: 10.1016/j.jaut.2023.103132] [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: 08/08/2023] [Revised: 10/05/2023] [Accepted: 10/12/2023] [Indexed: 11/15/2023]
Abstract
Rheumatoid factors (RFs), polyreactive antibodies canonically known to bind two conformational epitopes of IgG Fc, are a hallmark of rheumatoid arthritis but also can arise in other inflammatory conditions and infections. Also, infections may contribute to the development of rheumatoid arthritis and other autoimmune diseases. Recently, RFs only in rheumatoid arthritis were found to bind novel linear IgG epitopes as well as thousands of other rheumatoid arthritis autoantigens. Specific epitopes recognized by infection-induced polyreactive RFs remain undefined but could provide insights into loss of immune tolerance. Here, we identified novel linear IgG epitopes bound by RFs in COVID-19 but not rheumatoid arthritis or other conditions. The main COVID-19 RF was polyreactive, binding two IgG and multiple viral peptides with a tripeptide motif, as well as IgG Fc and SARS-CoV-2 spike proteins. In contrast, a rheumatoid arthritis-specific RF recognized IgG Fc, but not tripeptide motif-containing peptides or spike. Thus, RFs have disease-specific IgG reactivity and distinct polyreactivities that reflect the broader immune response. Moreover, the polyreactivity of a virus-induced RF appears to be attributable to a very short peptide motif. These findings refine our understanding of RFs and provide new insights into how viral infections may contribute to autoimmunity.
Collapse
Affiliation(s)
- Maya F Amjadi
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Maxwell H Parker
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan R Adyniec
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Zihao Zheng
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA; Department of Statistics, University of Wisconsin-Madison, Madison, WI, USA
| | - Alex M Robbins
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - S Janna Bashar
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael F Denny
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Sara S McCoy
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Irene M Ong
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, USA; Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA; Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Miriam A Shelef
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
2
|
P. Singh R, S. Bischoff D, S Singh S, H. Hahn B. Peptide-based immunotherapy in lupus: Where are we now? RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2023; 4:139-149. [PMID: 37781681 PMCID: PMC10538607 DOI: 10.2478/rir-2023-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/01/2023] [Indexed: 10/03/2023]
Abstract
In autoimmune rheumatic diseases, immune hyperactivity and chronic inflammation associate with immune dysregulation and the breakdown of immune self-tolerance. A continued, unresolved imbalance between effector and regulatory immune responses further exacerbates inflammation that ultimately causes tissue and organ damage. Many treatment modalities have been developed to restore the immune tolerance and immmunoregulatory balance in autoimmune rheumatic diseases, including the use of peptide-based therapeutics or the use of nanoparticles-based nanotechnology. This review summarizes the state-of-the-art therapeutic use of peptide-based therapies in autoimmune rheumatic diseases, with a specific focus on lupus.
Collapse
Affiliation(s)
- Ram P. Singh
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, 90073 CA, USA
| | - David S. Bischoff
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, 90073 CA, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, 90095 CA, USA
| | | | - Bevra H. Hahn
- Department of Medicine, University of California, Los Angeles, Los Angeles, 90095 CA, USA
| |
Collapse
|
3
|
Diamond B. Not Dead Yet. Annu Rev Immunol 2023; 41:1-15. [PMID: 37126416 DOI: 10.1146/annurev-immunol-101721-065214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
I have been a scientific grasshopper throughout my career, moving from question to question within the domain of lupus. This has proven to be immensely gratifying. Scientific exploration is endlessly fascinating, and succeeding in studies you care about with colleagues and trainees leads to strong and lasting bonds. Science isn't easy; being a woman in science presents challenges, but the drive to understand a disease remains strong.
Collapse
Affiliation(s)
- Betty Diamond
- Center of Autoimmune, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA;
| |
Collapse
|
4
|
Tomalla V, Schmeisser MJ, Weinmann-Menke J. Mouse models, antibodies, and neuroimaging: Current knowledge and future perspectives in neuropsychiatric systemic lupus erythematosus (NPSLE). Front Psychiatry 2023; 14:1078607. [PMID: 36970286 PMCID: PMC10031066 DOI: 10.3389/fpsyt.2023.1078607] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 02/13/2023] [Indexed: 03/11/2023] Open
Abstract
As a chronic autoimmune disease systemic lupus erythematosus (SLE) can also affect the central and the peripheral nervous system causing symptoms which are summed up as neuropsychiatric systemic lupus erythematosus (NPSLE). These symptoms are heterogenous including cognitive impairment, seizures, and fatigue, leading to morbidity or even mortality. At present, little is known about the pathophysiological processes involved in NPSLE. This review focuses on the current knowledge of the pathogenesis of NPSLE gained from the investigation of animal models, autoantibodies, and neuroimaging techniques. The antibodies investigated the most are anti-ribosomal P protein antibodies (Anti-rib P) and anti-N-Methyl-D-Aspartic Acid Receptor 2 antibodies (Anti-NR2), which represent a subpopulation of anti-dsDNA autoantibodies. Experimental data demonstrates that Anti-rib P and Anti-NR2 cause different neurological pathologies when applied intravenously (i.v.), intrathecally or intracerebrally in mice. Moreover, the investigation of lupus-prone mice, such as the MRL/MpJ-Faslpr/lpr strain (MRL/lpr) and the New Zealand black/New Zealand white mice (NZB × NZW F1) showed that circulating systemic antibodies cause different neuropsychiatric symptoms compared to intrathecally produced antibodies. Furthermore, neuroimaging techniques including magnetic resonance imaging (MRI) and positron emission tomography (PET) are commonly used tools to investigate structural and functional abnormalities in NPSLE patients. Current research suggests that the pathogenesis of NPSLE is heterogenous, complex and not yet fully understood. However, it demonstrates that further investigation is needed to develop individual therapy in NPSLE.
Collapse
Affiliation(s)
- Vanessa Tomalla
- Department of Internal Medicine, Division of Nephrology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Michael J. Schmeisser
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Focus Program Translational Neurosciences (FTN), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Julia Weinmann-Menke
- Department of Internal Medicine, Division of Nephrology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- *Correspondence: Julia Weinmann-Menke,
| |
Collapse
|
5
|
Manca E. Autoantibodies in Neuropsychiatric Systemic Lupus Erythematosus (NPSLE): Can They Be Used as Biomarkers for the Differential Diagnosis of This Disease? Clin Rev Allergy Immunol 2022; 63:194-209. [PMID: 34115263 PMCID: PMC9464150 DOI: 10.1007/s12016-021-08865-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2021] [Indexed: 01/13/2023]
Abstract
Systemic lupus erythematosus is a complex immunological disease where both environmental factors and genetic predisposition lead to the dysregulation of important immune mechanisms. Eventually, the combination of these factors leads to the production of self-reactive antibodies that can target any organ or tissue of the human body. Autoantibodies can form immune complexes responsible for both the organ damage and the most severe complications. Involvement of the central nervous system defines a subcategory of the disease, generally known with the denomination of neuropsychiatric systemic lupus erythematosus. Neuropsychiatric symptoms can range from relatively mild manifestations, such as headache, to more severe complications, such as psychosis. The evaluation of the presence of the autoantibodies in the serum of these patients is the most helpful diagnostic tool for the assessment of the disease. The scientific progresses achieved in the last decades helped researchers and physicians to discover some of autoepitopes targeted by the autoantibodies, although the majority of them have not been identified yet. Additionally, the central nervous system is full of epitopes that cannot be found elsewhere in the human body, for this reason, autoantibodies that selectively target these epitopes might be used for the differential diagnosis between patients with and without the neuropsychiatric symptoms. In this review, the most relevant data is reported with regard to mechanisms implicated in the production of autoantibodies and the most important autoantibodies found among patients with systemic lupus erythematosus with and without the neuropsychiatric manifestations.
Collapse
Affiliation(s)
- Elias Manca
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Cagliari, Italy.
| |
Collapse
|
6
|
Cardozo T, Cardozo L, Boutjdir M. Autoantibody:Autoantigen Competitor Decoys: Application to Cardiac Phenotypes. Front Immunol 2022; 13:812649. [PMID: 35154130 PMCID: PMC8832015 DOI: 10.3389/fimmu.2022.812649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/12/2022] [Indexed: 01/05/2023] Open
Abstract
Autoimmune diseases are often associated with autoantibodies that abnormally target self-antigens (autoantigens). An intuitive therapeutic strategy for diseases caused by aAbs is to design decoys, or soluble molecules that target the antigen combining site of these aAbs, thereby blocking binding of aAb to self-antigen and subsequent tissue damage. Here, we review the known decoy molecules of these types, discuss newer technological opportunities afforded by monoclonal antibody and structural biology advances, and discuss the challenges to this approach. Recent opportunities relevant to this approach for cardiac phenotypes, specifically Ro-associated long QT syndrome, are discussed.
Collapse
Affiliation(s)
- Timothy Cardozo
- Department of Biochemistry and Molecular Pharmacology, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| | - Lila Cardozo
- Department of Biochemistry and Molecular Pharmacology, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| | - Mohamed Boutjdir
- Department of Medicine, New York University (NYU) Grossman School of Medicine, New York, NY, United States.,Department of Medicine, Cell Biology and Pharmacology, State University of New York Downstate Medical Center, New York, NY, United States.,Cardiovascular Research Program, VA New York Harbor Healthcare System, New York, NY, United States
| |
Collapse
|
7
|
Wangriatisak K, Thanadetsuntorn C, Krittayapoositpot T, Leepiyasakulchai C, Suangtamai T, Ngamjanyaporn P, Khowawisetsut L, Khaenam P, Setthaudom C, Pisitkun P, Chootong P. The expansion of activated naive DNA autoreactive B cells and its association with disease activity in systemic lupus erythematosus patients. Arthritis Res Ther 2021; 23:179. [PMID: 34229724 PMCID: PMC8259008 DOI: 10.1186/s13075-021-02557-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/20/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Autoreactive B cells are well recognized as key participants in the pathogenesis of systemic lupus erythematosus (SLE). However, elucidating the particular subset of B cells in producing anti-dsDNA antibodies is limited due to their B cell heterogeneity. This study aimed to identify peripheral B cell subpopulations that display autoreactivity to DNA and contribute to lupus pathogenesis. METHODS Flow cytometry was used to detect total B cell subsets (n = 20) and DNA autoreactive B cells (n = 15) in SLE patients' peripheral blood. Clinical disease activities were assessed in SLE patients using modified SLEDAI-2 K and used for correlation analyses with expanded B cell subsets and DNA autoreactive B cells. RESULTS The increases of circulating double negative 2 (DN2) and activated naïve (aNAV) B cells were significantly observed in SLE patients. Expanded B cell subsets and DNA autoreactive B cells represented a high proportion of aNAV B cells with overexpression of CD69 and CD86. The frequencies of aNAV B cells in total B cell populations were significantly correlated with modified SLEDAI-2 K scores. Further analysis showed that expansion of aNAV DNA autoreactive B cells was more related to disease activity and serum anti-dsDNA antibody levels than to total aNAV B cells. CONCLUSION Our study demonstrated an expansion of aNAV B cells in SLE patients. The association between the frequency of aNAV B cells and disease activity patients suggested that these expanded B cells may play a role in SLE pathogenesis.
Collapse
Affiliation(s)
- Kittikorn Wangriatisak
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4 Road, Salaya, Nakhonpathom, 73170, Thailand
| | - Chokchai Thanadetsuntorn
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama 6 Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Thamonwan Krittayapoositpot
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4 Road, Salaya, Nakhonpathom, 73170, Thailand
| | - Chaniya Leepiyasakulchai
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4 Road, Salaya, Nakhonpathom, 73170, Thailand
| | - Thanitta Suangtamai
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama 6 Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Pintip Ngamjanyaporn
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama 6 Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Ladawan Khowawisetsut
- Department of Parasitology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Center of Excellence for Microparticle and Exosome in Diseases, Department of Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Prasong Khaenam
- Center of Standardization and Product Validation, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Chavachol Setthaudom
- Immunology Laboratory, Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Prapaporn Pisitkun
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama 6 Road, Ratchathewi, Bangkok, 10400, Thailand.
- Translational Medicine Program, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.
| | - Patchanee Chootong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4 Road, Salaya, Nakhonpathom, 73170, Thailand.
| |
Collapse
|
8
|
Atisha-Fregoso Y, Toz B, Diamond B. Meant to B: B cells as a therapeutic target in systemic lupus erythematosus. J Clin Invest 2021; 131:149095. [PMID: 34128474 PMCID: PMC8203443 DOI: 10.1172/jci149095] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
B cells have a prominent role in the pathogenesis of systemic lupus erythematosus (SLE). They are mediators of inflammation through the production of pathogenic antibodies that augment inflammation and cause direct tissue and cell damage. Multiple therapeutic agents targeting B cells have been successfully used in mouse models of SLE; however, these preclinical studies have led to approval of only one new agent to treat patients with SLE: belimumab, a monoclonal antibody targeting B cell-activating factor (BAFF). Integrating the experience acquired from previous clinical trials with the knowledge generated by new studies about mechanisms of B cell contributions to SLE in specific groups of patients is critical to the development of new treatment strategies that will help to improve outcomes in patients with SLE. In particular, a sharper focus on B cell differentiation to plasma cells is warranted.
Collapse
Affiliation(s)
- Yemil Atisha-Fregoso
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, New York, USA
| | - Bahtiyar Toz
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Queens Hospital Center, New York, New York, USA
| | - Betty Diamond
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, USA
| |
Collapse
|
9
|
Varley JA, Andersson M, Grant E, Berretta A, Zandi MS, Bondet V, Duffy D, Hunt D, Piehl F, Waters P, Irani SR. Absence of Neuronal Autoantibodies in Neuropsychiatric Systemic Lupus Erythematosus. Ann Neurol 2020; 88:1244-1250. [DOI: 10.1002/ana.25908] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/10/2020] [Accepted: 09/14/2020] [Indexed: 12/19/2022]
Affiliation(s)
- James A. Varley
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences University of Oxford Oxford UK
- Department of Neurology, John Radcliffe Hospital Oxford University Hospitals Oxford UK
| | - Magnus Andersson
- Department of Clinical Neuroscience, Centre for Molecular Medicine, Karolinska Institute Karolinska University Hospital Stockholm Sweden
| | - Eleanor Grant
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences University of Oxford Oxford UK
- Department of Neurology, John Radcliffe Hospital Oxford University Hospitals Oxford UK
| | - Antonio Berretta
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences University of Oxford Oxford UK
| | | | - Vincent Bondet
- Immunobiology of Dendritic Cells, Inserm U1223 Institut Pasteur Paris France
| | - Darragh Duffy
- Immunobiology of Dendritic Cells, Inserm U1223 Institut Pasteur Paris France
| | - David Hunt
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine University of Edinburgh, Western General Hospital Edinburgh UK
- Centre for Clinical Brain Sciences (CCBS) University of Edinburgh Edinburgh UK
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Centre for Molecular Medicine, Karolinska Institute Karolinska University Hospital Stockholm Sweden
| | - Patrick Waters
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences University of Oxford Oxford UK
| | - Sarosh R. Irani
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences University of Oxford Oxford UK
- Department of Neurology, John Radcliffe Hospital Oxford University Hospitals Oxford UK
| |
Collapse
|
10
|
Guthridge CJ, Gross T, Quintero M, Kheir JM, Levin J, Bourn RL, Khan S, Peercy M, Saunkeah B, Guthridge JM, James JA. Expanded Autoantibody Profiles for Subsetting of Native American, African American, and European American Patients With Systemic Lupus Erythematosus. ACR Open Rheumatol 2020; 2:415-423. [PMID: 32567819 PMCID: PMC7368137 DOI: 10.1002/acr2.11149] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVE Many Native American (NA) patients with systemic lupus erythematosus (SLE) do not exhibit the classical SLE autoantibody profiles of European American (EA) and African American (AA) patients with SLE. The poorer SLE disease outcomes noted in NA patients highlights a need for more equitable diagnostic and prognostic tools for NA patients with SLE. The objective was to identify informative autoantibody profiles for NA, AA, and EA patients with SLE using an expanded set of autoantigens. METHODS Sera from 49 NA, 49 AA, and 49 EA age-, sex-, and antinuclear autoantibody titer-matched patients with SLE who met the American College of Rheumatology classification criteria and 10 ethnicity-, sex-, and age-matched controls were tested for autoantibody reactivity by autoantigen microarrays. Autoantibodies that were significantly elevated in patients with SLE compared with ethnicity-specific controls were selected for hierarchical clustering. Differences in clinical criteria between patient clusters were determined by Fisher's exact test and corrected for multiple comparisons. RESULTS NA, AA, and EA patients with SLE each had a cluster distinguished by higher levels of anti-Ro52 and another cluster distinguished by nucleic acid-specific autoantibodies. Additional clusters were distinguished in NA patients by elevated extracellular matrix autoantibodies and were distinguished in AA patients by elevated Sm/RNP autoantibody and elevated nucleolin/histone autoantibody. Two EA patient clusters with similar nucleic acid- and Ro52-specific autoantibodies were distinguished by either high or low histone 2A reactivity. Renal manifestations trended higher in the NA Ro52 cluster and were significantly enriched in the AA nucleolin/histone cluster. The AA nucleolin/histone cluster and EA H2A cluster had higher disease activity. CONCLUSION Expanded autoantibody profiles can identify informative subsets of patients with SLE.
Collapse
Affiliation(s)
| | | | | | | | - Jeremy Levin
- Oklahoma Medical Research FoundationOklahoma City
| | | | | | | | | | - Joel M. Guthridge
- Oklahoma Medical Research Foundation and University of Oklahoma Health Sciences CenterOklahoma City
| | - Judith A. James
- Oklahoma Medical Research Foundation and University of Oklahoma Health Sciences CenterOklahoma City
| |
Collapse
|
11
|
El Khoury L, Zarfeshani A, Diamond B. Using the Mouse to Model Human Diseases: Cognitive Impairment in Systemic Lupus Erythematosus. J Rheumatol 2020; 47:1145-1149. [PMID: 32295852 PMCID: PMC11207199 DOI: 10.3899/jrheum.200410] [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] [Indexed: 11/22/2022]
Abstract
In this 2020 Dunlop-Dottridge Lecture, the authors discuss cognitive impairment (CI), one of the most prevalent neuropsychiatric syndromes in systemic lupus erythematosus (SLE). Patients often report CI as the most bothersome disease-related manifestation, with a great effect on their quality of life. Nevertheless, studies focusing on CI remain scarce and no effective targeted therapy has been identified. We herein present murine models of CI in SLE with insights into the pathogenesis of this condition as well as the role of the renin angiotensin system in microglial activation. We will discuss the role of neuroimaging as a useful objective assessment tool, describing our experience in previous and ongoing clinical trials of CI in patients with SLE.
Collapse
Affiliation(s)
- Lara El Khoury
- From the Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
- L. El Khoury, MD, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health; A. Zarfeshani, PhD, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health; B. Diamond, MD, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health
| | - Aida Zarfeshani
- From the Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
- L. El Khoury, MD, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health; A. Zarfeshani, PhD, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health; B. Diamond, MD, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health
| | - Betty Diamond
- From the Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA.
- L. El Khoury, MD, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health; A. Zarfeshani, PhD, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health; B. Diamond, MD, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health.
| |
Collapse
|
12
|
Wreczycka K, Franke V, Uyar B, Wurmus R, Bulut S, Tursun B, Akalin A. HOT or not: examining the basis of high-occupancy target regions. Nucleic Acids Res 2019; 47:5735-5745. [PMID: 31114922 PMCID: PMC6582337 DOI: 10.1093/nar/gkz460] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 05/02/2019] [Accepted: 05/13/2019] [Indexed: 01/16/2023] Open
Abstract
High-occupancy target (HOT) regions are segments of the genome with unusually high number of transcription factor binding sites. These regions are observed in multiple species and thought to have biological importance due to high transcription factor occupancy. Furthermore, they coincide with house-keeping gene promoters and consequently associated genes are stably expressed across multiple cell types. Despite these features, HOT regions are solely defined using ChIP-seq experiments and shown to lack canonical motifs for transcription factors that are thought to be bound there. Although, ChIP-seq experiments are the golden standard for finding genome-wide binding sites of a protein, they are not noise free. Here, we show that HOT regions are likely to be ChIP-seq artifacts and they are similar to previously proposed ‘hyper-ChIPable’ regions. Using ChIP-seq data sets for knocked-out transcription factors, we demonstrate presence of false positive signals on HOT regions. We observe sequence characteristics and genomic features that are discriminatory of HOT regions, such as GC/CpG-rich k-mers, enrichment of RNA–DNA hybrids (R-loops) and DNA tertiary structures (G-quadruplex DNA). The artificial ChIP-seq enrichment on HOT regions could be associated to these discriminatory features. Furthermore, we propose strategies to deal with such artifacts for the future ChIP-seq studies.
Collapse
Affiliation(s)
- Katarzyna Wreczycka
- Bioinformatics and Omics Data Science Platform, Berlin Institute for Medical Systems Biology, Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Vedran Franke
- Bioinformatics and Omics Data Science Platform, Berlin Institute for Medical Systems Biology, Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Bora Uyar
- Bioinformatics and Omics Data Science Platform, Berlin Institute for Medical Systems Biology, Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Ricardo Wurmus
- Bioinformatics and Omics Data Science Platform, Berlin Institute for Medical Systems Biology, Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Selman Bulut
- Gene Regulation and Cell Fate Decision in C. elegans, Berlin Institute for Medical Systems Biology, Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Baris Tursun
- Gene Regulation and Cell Fate Decision in C. elegans, Berlin Institute for Medical Systems Biology, Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Altuna Akalin
- Bioinformatics and Omics Data Science Platform, Berlin Institute for Medical Systems Biology, Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
13
|
Ploran E, Tang C, Mackay M, Small M, Anderson E, Storbeck J, Bascetta B, Kang S, Aranow C, Sartori C, Watson P, Volpe B, Diamond B, Eidelberg D. Assessing cognitive impairment in SLE: examining relationships between resting glucose metabolism and anti-NMDAR antibodies with navigational performance. Lupus Sci Med 2019; 6:e000327. [PMID: 31413849 PMCID: PMC6667777 DOI: 10.1136/lupus-2019-000327] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/17/2019] [Accepted: 06/10/2019] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Resting Fluorine-18 fluorodeoxyglucose positron emission tomography (FDG-PET) brain imaging and neuropsychological testing were used to investigate the usefulness of a spatial navigation task (SNT) as a performance benchmark for cognitive impairment related to anti-N-methyl D-aspartate (anti-NMDA) receptor antibodies (DNRAb) in SLE. METHODS Neuropsychological assessments, including a desktop 3-D virtual SNT, were performed on 19 SLE participants and 9 healthy control (HC) subjects. SLE participants had stable disease activity and medication doses and no history of neuropsychiatric illness or current use of mind-altering medications. Resting FDG-PET scans were obtained on all SLE participants and compared with a historical set from 25 age-matched and sex-matched HCs. Serum DNRAb titres were measured by ELISA. RESULTS 11/19 (58%) of SLE participants failed to complete the SNT (SNT-) compared with 2/9 (22%) of HCs. Compared with 7/9 (78%) in HCs, only 2/9 (22%; p=0.037) of SLE participants with high serum DNRAb titres completed the SNT, in contrast to 6/10 (60%; p=0.810) in SLE participants with low DNRAb titres. Voxel-wise comparison of FDG-PET scans between the 8 SLE participants successfully completing the SNT task (SNT+) and the 11 SNT- SLE participants revealed increased metabolism in the SNT+ participants (p<0.001) in the left anterior putamen/caudate, right anterior putamen, left prefrontal cortex (BA 9), right prefrontal cortex (BA 9/10) and left lateral and medial frontal cortex (BA 8). Compared with HCs, the SNT+ group demonstrated increased metabolism in all regions (p<0.02) except for the right prefrontal cortex (BA 9), whereas the SNT- group demonstrated either significantly decreased or similar metabolism in these seven regions. CONCLUSIONS SNT performance is associated with serum DNRAb titres and resting glucose metabolism in the anterior putamen/caudate and frontal cortex, suggesting compensatory neural recruitment in SNT-associated regions is necessary for successful completion of the task. The SNT therefore has potential for use as a marker for SLE-mediated cognitive impairment.
Collapse
Affiliation(s)
- Elisabeth Ploran
- Department of Psychology, Hofstra University, Hempstead, New York, USA
| | - Chris Tang
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Meggan Mackay
- The Center for Autoimmune, Musculoskeletal, and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Michael Small
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Erik Anderson
- The Center for Autoimmune, Musculoskeletal, and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Justin Storbeck
- Department of Psychology, Queens College, Flushing, New York, USA
| | | | - Simran Kang
- Department of Psychology, Queens College, Flushing, New York, USA
| | - Cynthia Aranow
- The Center for Autoimmune, Musculoskeletal, and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Carl Sartori
- The Center for Autoimmune, Musculoskeletal, and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Philip Watson
- Department of Psychiatry, Zucker Hillside Hospital, Glen Oaks, New York, USA
| | - Bruce Volpe
- Center for Biomedical Science, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Betty Diamond
- The Center for Autoimmune, Musculoskeletal, and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - David Eidelberg
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| |
Collapse
|
14
|
Yasuda S. Emerging targets for the treatment of lupus erythematosus: There is no royal road to treating lupus. Mod Rheumatol 2019; 29:60-69. [PMID: 29947283 DOI: 10.1080/14397595.2018.1493909] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Systemic lupus erythematosus (SLE) is a highly heterogeneous autoimmune disease that preferentially affects women of child-bearing age. Most current treatments for SLE with the exception of belimumab are not target-specific. Nontargeted therapy such as corticosteroids, cyclophosphamide, and other immunosuppressive drugs results in unwanted adverse effects. Although progress in treatment, including supportive therapy, has dramatically improved the prognosis of patients with SLE, better treatment drugs and protocols with fewer adverse effects and higher efficacy for the most severe form of SLE are needed. Advancements in genomics, immunology, and pathophysiology in the field of systemic autoimmunity have provided physicians with increasing knowledge, but the most appropriate treatment for each patient with SLE remains to be established. Therefore, the search for novel treatment targets in patients with SLE is ongoing. This review focuses on recent findings in the genetics of lupus and the abnormalities in cellular interactions, cytokine profiles, and intracellular signaling in patients with SLE. Novel molecular targets for lupus, mostly introduced through clinical trials, are then discussed based on these findings.
Collapse
Affiliation(s)
- Shinsuke Yasuda
- a Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine , Hokkaido University , Sapporo , Japan
| |
Collapse
|
15
|
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
|
16
|
Porat A, Giat E, Kowal C, He M, Son M, Latz E, Ben-Zvi I, Al-Abed Y, Diamond B. DNA-Mediated Interferon Signature Induction by SLE Serum Occurs in Monocytes Through Two Pathways: A Mechanism to Inhibit Both Pathways. Front Immunol 2018; 9:2824. [PMID: 30619247 PMCID: PMC6297782 DOI: 10.3389/fimmu.2018.02824] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 11/15/2018] [Indexed: 02/05/2023] Open
Abstract
A primary mechanism for activation of innate immunity is recognition of damage or pathogen associated molecular patterns by pattern recognition receptors (PRRs). Nucleic acid is a damage associated molecular pattern molecule that when internalized into a monocyte and recognized by intracellular nucleic acid sensing toll like receptors will cause production of type 1 interferon. The process by which DNA or RNA is delivered into the cytosol of monocytes in systemic lupus erythematosus remains incompletely understood, and therapeutic approaches to prevent DNA-mediated monocyte activation are needed. We identified two mechanisms for internalization of DNA by monocytes. IgG-bound DNA was internalized by interacting with Fc gamma receptor IIa, while high-mobility group box-1 protein-bound DNA was internalized by interacting with the receptor for advanced glycation end products. Both pathways contribute to an inflammatory phenotype in monocytes exposed to serum from patients with SLE. Moreover, both of these pathways can be inhibited by a pentapeptide, DWEYS, which is a DNA mimetope. In one instance DWEYS directly competes with DNA for antibody binding and in the other DWEYS binds high-mobility group box-1 and blocks its interaction with RAGE. Our data highlight distinct pathways involved in nucleic acid enters monocytes in SLE, and identify a potential therapeutic to prevent nucleic acid internalization in SLE.
Collapse
Affiliation(s)
- Amit Porat
- Elmezzi Graduate School for Molecular Medicine, Manhasset, NY, United States.,Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Eitan Giat
- Elmezzi Graduate School for Molecular Medicine, Manhasset, NY, United States.,Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Czeslawa Kowal
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Mingzhu He
- Center for Molecular Innovation, Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Myoungsun Son
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Eicke Latz
- Biomedical Centre (BMZ), Institute of Innate Immunity, 1G007 University Hospital, University of Bonn, Bonn, Germany
| | - Ilan Ben-Zvi
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Yousef Al-Abed
- Center for Molecular Innovation, Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Betty Diamond
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, NY, United States
| |
Collapse
|
17
|
Fujieda Y, Mader S, Jeganathan V, Arinuma Y, Shimizu Y, Kato M, Oku K, Minami A, Shimizu C, Yasuda S, Atsumi T. Clinical significance of anti-DNA/N-methyl-D-aspartate receptor 2 antibodies in de novo and post-steroid cases with neuropsychiatric systemic lupus erythematosus. Int J Rheum Dis 2018; 22:443-448. [PMID: 30358102 DOI: 10.1111/1756-185x.13392] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 07/11/2018] [Accepted: 08/05/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND Anti-DNA/N-methyl-D-aspartate receptor 2 (NR2) antibodies (anti-DNA/NR2 antibodies) are a subset of anti-DNA autoantibodies that cross-react with the extracellular domain of the GluN2A/GluN2B subunits of NR2. These antibodies induce apoptosis of hippocampus neurons and psychiatric disorder in mice and humans. Neuropsychiatric system lupus erythematosus (NPSLE) can develop after initiation of corticosteroids (post-steroid neuropsychiatric manifestation: PSNP) or before treatment (de novo NPSLE); however, pathophysiological differences between these subtypes remain unclear. The objective of this study was to clarify the prevalence of anti-DNA/NR2 antibodies in patients with NPSLE. METHODS This study involved a cohort of patients with NPSLE admitted to our hospital. NPSLE patients were classified into two groups, de novo NPSLE and PSNP-SLE. Serum anti-DNA antibodies and anti-DNA/NR2 antibodies were measured by enzyme-linked immunosorbent assays. RESULTS Serum samples were obtained from 24 patients with de novo NPSLE, 25 with PSNP-SLE and 76 healthy controls (HC). The level of anti-DNA/NR2 antibodies in patients with de novo NPSLE and PSNP-SLE were also higher than those in HC. Positive correlation between anti-DNA antibodies and anti-DNA/NR2 antibodies were found in PSNP-SLE, but was not significant in de novo NPSLE. CONCLUSION The levels of anti-DNA/NR2 antibodies in PSNP-SLE were similar to those in de novo NPSLE. Anti-DNA/NR2 antibodies in PSNP-SLE were suggested as a dominant subset of anti-DNA antibodies, indicating that anti-DNA/NR2 antibodies may be a predictive factor in PSNP-SLE.
Collapse
Affiliation(s)
- Yuichiro Fujieda
- Department of Rheumatology, Endocrinology and Nephrology, Graduate School of Medicine and Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Simone Mader
- Center of Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, Manhasset, New York
| | - Venkatesh Jeganathan
- Center of Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, Manhasset, New York
| | - Yoshiyuki Arinuma
- Department of Rheumatology and Infectious Disease, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Yuka Shimizu
- Department of Rheumatology, Endocrinology and Nephrology, Graduate School of Medicine and Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Masaru Kato
- Department of Rheumatology, Endocrinology and Nephrology, Graduate School of Medicine and Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Kenji Oku
- Department of Rheumatology, Endocrinology and Nephrology, Graduate School of Medicine and Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Akiko Minami
- Division of Laboratory and Transfusion Medicine, Hokkaido University, Sapporo, Japan
| | - Chikara Shimizu
- Division of Laboratory and Transfusion Medicine, Hokkaido University, Sapporo, Japan
| | - Shinsuke Yasuda
- Department of Rheumatology, Endocrinology and Nephrology, Graduate School of Medicine and Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Tatsuya Atsumi
- Department of Rheumatology, Endocrinology and Nephrology, Graduate School of Medicine and Faculty of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
18
|
Heyduk E, Hickey R, Pozzi N, Heyduk T. Peptide ligand-based ELISA reagents for antibody detection. Anal Biochem 2018; 559:55-61. [PMID: 30130491 DOI: 10.1016/j.ab.2018.08.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 07/26/2018] [Accepted: 08/15/2018] [Indexed: 12/17/2022]
Abstract
Detection of specific antibodies has numerous research, therapeutic and diagnostic applications. Short peptide ligands that bind specifically to antibodies with continuous epitopes can be derived from epitope mapping experiments. Short peptide ligands (mimotopes) specific to antibodies with discontinuous epitopes can be identified by screening complex peptide libraries. In an effort to enhance practical utility of such peptide ligands, we describe here a simple approach to turn such target antibody-specific peptide ligands into specific ELISA detection reagents. We show that a simple addition of biotinylated peptide ligands to commonly available horseradish peroxidase (HRP)-labeled streptavidin (or HRP-anti-biotin antibody), or digoxigenin-labeled peptides to HRP-anti-digoxigenin antibody detection reagents transformed these generic detection reagents into sensitive target antibody-specific reagents. ELISA assays performed using these reagents exhibited excellent analytical properties indicating their practical utility for antibody detection. One generic detection reagent can be readily transformed into many different specific ELISA reagents by a simple mix and match design using an appropriate target-specific peptide ligand. Simplicity of preparation of these ELISA reagents for detecting antibodies should facilitate their practical applications.
Collapse
Affiliation(s)
- Ewa Heyduk
- Edward A. Doisy Department of Biochemistry and Molecular Biology, St. Louis University Medical School, 1100 S. Grand Blvd, St. Louis, MO, 63104, USA
| | - Rachel Hickey
- Edward A. Doisy Department of Biochemistry and Molecular Biology, St. Louis University Medical School, 1100 S. Grand Blvd, St. Louis, MO, 63104, USA
| | - Nicola Pozzi
- Edward A. Doisy Department of Biochemistry and Molecular Biology, St. Louis University Medical School, 1100 S. Grand Blvd, St. Louis, MO, 63104, USA
| | - Tomasz Heyduk
- Edward A. Doisy Department of Biochemistry and Molecular Biology, St. Louis University Medical School, 1100 S. Grand Blvd, St. Louis, MO, 63104, USA.
| |
Collapse
|
19
|
Bashford-Rogers RJM, Smith KGC, Thomas DC. Antibody repertoire analysis in polygenic autoimmune diseases. Immunology 2018; 155:3-17. [PMID: 29574826 PMCID: PMC6099162 DOI: 10.1111/imm.12927] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/01/2018] [Accepted: 03/06/2018] [Indexed: 12/18/2022] Open
Abstract
High-throughput sequencing of the DNA/RNA encoding antibody heavy- and light-chains is rapidly transforming the field of adaptive immunity. It can address key questions, including: (i) how the B-cell repertoire differs in health and disease; and (ii) if it does differ, the point(s) in B-cell development at which this occurs. The advent of technologies, such as whole-genome sequencing, offers the chance to link abnormalities in the B-cell antibody repertoire to specific genomic variants and polymorphisms. Here, we discuss the current research using B-cell antibody repertoire sequencing in three polygenic autoimmune diseases where there is good evidence for a pathological role for B-cells, namely systemic lupus erythematosus, multiple sclerosis and rheumatoid arthritis. These autoimmune diseases exhibit significantly skewed B-cell receptor repertoires compared with healthy controls. Interestingly, some common repertoire defects are shared between diseases, such as elevated IGHV4-34 gene usage. B-cell clones have effectively been characterized and tracked between different tissues and blood in autoimmune disease. It has been hypothesized that these differences may signify differences in B-cell tolerance; however, the mechanisms and implications of these defects are not clear.
Collapse
Affiliation(s)
| | | | - David C Thomas
- Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
20
|
A structural investigation of FISLE-412, a peptidomimetic compound derived from saquinavir that targets lupus autoantibodies. Bioorg Med Chem Lett 2017; 27:4725-4729. [PMID: 28927788 DOI: 10.1016/j.bmcl.2017.08.070] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 08/30/2017] [Accepted: 08/31/2017] [Indexed: 01/17/2023]
Abstract
FISLE-412 is the first reported small molecule peptidomimetic that neutralizes anti-dsDNA autoantibodies associated with systemic lupus erythematosus (SLE) pathogenesis. FISLE-412 is a complex small molecule that involves a challenging synthesis scheme, but has attractive pharmacological activities as a potential small molecule therapeutic in lupus. Therefore, we initiated a Structure-Activity Relationship study to simplify the complexity of FISLE-412. We synthesized a small library of mimetopes around the FISLE-412 structure and identified several analogues which could neutralize anti-DNA lupus antibodies in vitro and ex vivo. Our strategies reduced the structural complexity of FISLE-412 and provide important information that may guide development of potential autoantibody-targeted lupus therapeutics.
Collapse
|
21
|
Mader S, Brimberg L, Diamond B. The Role of Brain-Reactive Autoantibodies in Brain Pathology and Cognitive Impairment. Front Immunol 2017; 8:1101. [PMID: 28955334 PMCID: PMC5601985 DOI: 10.3389/fimmu.2017.01101] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/22/2017] [Indexed: 12/15/2022] Open
Abstract
Antibodies to different brain proteins have been recently found to be associated with an increasing number of different autoimmune diseases. They need to penetrate the blood–brain barrier (BBB) in order to bind antigens within the central nervous system (CNS). They can target either neuronal or non-neuronal antigen and result in damage either by themselves or in synergy with other inflammatory mediators. Antibodies can lead to acute brain pathology, which may be reversible; alternatively, they may trigger irreversible damage that persists even though the antibodies are no longer present. In this review, we will describe two different autoimmune conditions and the role of their antibodies in causing brain pathology. In systemic lupus erythematosus (SLE), patients can have double stranded DNA antibodies that cross react with the neuronal N-methyl-d-aspartate receptor (NMDAR), which have been recently linked to neurocognitive dysfunction. In neuromyelitis optica (NMO), antibodies to astrocytic aquaporin-4 (AQP4) are diagnostic of disease. There is emerging evidence that pathogenic T cells also play an important role for the disease pathogenesis in NMO since they infiltrate in the CNS. In order to enable appropriate and less invasive treatment for antibody-mediated diseases, we need to understand the mechanisms of antibody-mediated pathology, the acute and chronic effects of antibody exposure, if the antibodies are produced intrathecally or systemically, their target antigen, and what triggers their production. Emerging data also show that in utero exposure to some brain-reactive antibodies, such as those found in SLE, can cause neurodevelopmental impairment since they can penetrate the embryonic BBB. If the antibody exposure occurs at a critical time of development, this can result in irreversible damage of the offspring that persists throughout adulthood.
Collapse
Affiliation(s)
- Simone Mader
- The Feinstein Institute for Medical Research, The Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, Northwell Health System, Manhasset, NY, United States
| | - Lior Brimberg
- The Feinstein Institute for Medical Research, The Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, Northwell Health System, Manhasset, NY, United States
| | - Betty Diamond
- The Feinstein Institute for Medical Research, The Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, Northwell Health System, Manhasset, NY, United States
| |
Collapse
|
22
|
Pozsgay J, Szekanecz Z, Sármay G. Antigen-specific immunotherapies in rheumatic diseases. Nat Rev Rheumatol 2017; 13:525-537. [DOI: 10.1038/nrrheum.2017.107] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
23
|
Malkiel S, Jeganathan V, Wolfson S, Manjarrez Orduño N, Marasco E, Aranow C, Mackay M, Gregersen PK, Diamond B. Checkpoints for Autoreactive B Cells in the Peripheral Blood of Lupus Patients Assessed by Flow Cytometry. Arthritis Rheumatol 2017; 68:2210-20. [PMID: 27059652 DOI: 10.1002/art.39710] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 03/31/2016] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Antinuclear antibodies (ANAs) are diagnostic in several autoimmune disorders, yet the failure to achieve B cell tolerance in these diseases is still poorly understood. Although secreted ANAs detected by an indirect immunofluorescence assay are the gold standard for autoreactivity, there has been no convenient assay with which to measure the frequency of circulating B cells that recognize nuclear antigens (ANA+ B cells) in patients. The aim of this study was to generate an assay to easily identify these B cells and to examine its utility in a study of autoreactive B cells in systemic lupus erythematosus (SLE). METHODS We developed and validated a novel flow cytometry-based assay that identifies ANA+ B cells using biotinylated nuclear extracts, and utilized it to examine B cell tolerance checkpoints in peripheral blood mononuclear cells obtained from SLE patients and healthy controls. RESULTS We observed progressive selection against ANA+ B cells as they matured from transitional to naive to CD27+IgD- and CD27+IgD+ memory cells in both healthy subjects and SLE patients; however, ANA+ naive B cells in SLE patients were not anergized to the same extent as in healthy individuals. We also showed that anergy induction is restored in SLE patients treated with belimumab, an inhibitor of BAFF. CONCLUSION This assay will enable studies of large populations to identify potential genetic or environmental factors affecting B cell tolerance checkpoints in healthy subjects and patients with autoimmune disease and permit monitoring of the B cell response to therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | | | - Cynthia Aranow
- Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York
| | - Meggan Mackay
- Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York
| | - Peter K Gregersen
- Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York
| | - Betty Diamond
- Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York
| |
Collapse
|
24
|
Tay SH, Fairhurst AM, Mak A. Clinical utility of circulating anti-N-methyl- d-aspartate receptor subunits NR2A/B antibody for the diagnosis of neuropsychiatric syndromes in systemic lupus erythematosus and Sjögren's syndrome: An updated meta-analysis. Autoimmun Rev 2016; 16:114-122. [PMID: 27988431 DOI: 10.1016/j.autrev.2016.12.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 09/08/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND/PURPOSE Neuropsychiatric (NP) events are found in patients with rheumatic diseases, commonly in systemic lupus erythematosus (SLE) and Sjögren's syndrome (SS). The standard nomenclature and case definitions for 19 NPSLE syndromes by the American College of Rheumatology (ACR) Committee on Research cover a wide range of NP events seen in both SLE and SS. Despite advances in the understanding of SLE and SS, NP syndromes continue to pose diagnostic challenges. Correct attribution of NP events is critical in determining the correct treatment and prognosis. Anti-N-methyl-d-aspartate receptor subunits NR2A/B (anti-NR2A/B) antibodies have been demonstrated in the sera of SLE and SS patients and have been associated with collective or specific NP syndromes, though not consistently. Interpretation of anti-NR2A/B antibody data in the medical literature is rendered difficult by small sample size of patient groups. By combining different studies to generate a pooled effect size, a meta-analysis can increase the power to detect differences in the presence or absence of NP syndromes. Hence, we set out to perform a meta-analysis to assess the association between anti-NR2A/B antibodies and NP syndromes in SLE and SS. METHODS A literature search was conducted using PubMed and other databases from inception to June 2016. We abstracted data relating to anti-NR2A/B antibodies from the identified studies. The random effects model was used to calculate overall combined odds ratio (OD) with its corresponding 95% confidence interval (CI) to evaluate the relationship between anti-NR2A/B antibodies and NP syndromes in SLE and SS patients with and without NP events. We also included our own cohort of 57 SLE patients fulfilling the ACR 1997 revised classification criteria and 58 healthy controls (HCs). RESULTS In total, 17 studies with data on anti-NR2A/B antibodies in 2212 SLE patients, 66 SS patients, 99 disease controls (DCs) (e.g. antiphospholipid syndrome, myasthenia gravis and autoimmune polyendocrine syndrome I) and 538 HCs were used in this analysis. Overall pooled prevalence of serum/plasma anti-NR2A/B antibodies was higher in SLE patients [24.6% (95% CI 18.5-32.0%)] and SS patients [19.7% (95% CI 11.8-31.0%)] compared to DCs [14.8% (95% CI 2.2-56.9)] and HCs [7.6% (95% CI 4.6-12.4%)] (p=0.001). There was a significantly greater proportion of SLE and SS patients with NP syndromes who demonstrated positivity for serum/plasma anti-NR2A/B antibody [pooled OR=1.607 (95% CI 1.041-2.479), p=0.032] as compared to SLE and SS patients without NP syndromes in 13 studies. Usable data for cerebrospinal fluid anti-NR2A/B antibodies were available in only 4 studies [pooled OR=0.831 (95% CI 0.365-1.888), p=0.658]. Among the 19 NP syndromes, serum/plasma anti-NR2A/B antibodies were not specifically associated with any NP syndrome, including cognitive dysfunction (p=0.259) and mood disorder (p=0.503). Meta-regression identified proportion of anti-double-stranded deoxyribonucleic acid antibody positivity (p=0.009) and SLE Disease Activity Index (p=0.028) as moderators for the heterogeneity of serum/plasma anti-NR2A/B antibodies. CONCLUSION Circulating anti-NR2A/B antibody testing has a diagnostic value for NP syndromes in SLE and SS collectively. However, the evidence to date suggests that anti-NR2A/B antibody positivity cannot distinguish specific NP syndromes.
Collapse
Affiliation(s)
- Sen Hee Tay
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Division of Rheumatology, Department of Medicine, National University Hospital, National University Health System, Singapore.
| | - Anna-Marie Fairhurst
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore; Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Immunology, UT Southwestern Medical Center at Dallas, TX, USA
| | - Anselm Mak
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Division of Rheumatology, Department of Medicine, National University Hospital, National University Health System, Singapore
| |
Collapse
|
25
|
VanPatten S, Sun S, He M, Cheng KF, Altiti A, Papatheodorou A, Kowal C, Jeganathan V, Crawford JM, Bloom O, Volpe BT, Grant C, Meurice N, Coleman TR, Diamond B, Al-Abed Y. Amending HIV Drugs: A Novel Small-Molecule Approach To Target Lupus Anti-DNA Antibodies. J Med Chem 2016; 59:8859-8867. [PMID: 27603688 DOI: 10.1021/acs.jmedchem.6b00694] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Systemic lupus erythematosus is an autoimmune disease that can affect numerous tissues and is characterized by the production of nuclear antigen-directed autoantibodies (e.g., anti-dsDNA). Using a combination of virtual and ELISA-based screens, we made the intriguing discovery that several HIV-protease inhibitors can function as decoy antigens to specifically inhibit the binding of anti-dsDNA antibodies to target antigens such as dsDNA and pentapeptide DWEYS. Computational modeling revealed that HIV-protease inhibitors comprised structural features present in DWEYS and predicted that analogues containing more flexible backbones would possess preferred binding characteristics. To address this, we reduced the internal amide backbone to improve flexibility, producing new small-molecule decoy antigens, which neutralize anti-dsDNA antibodies in vitro, in situ, and in vivo. Pharmacokinetic and SLE model studies demonstrated that peptidomimetic FISLE-412,1 a reduced HIV protease inhibitor analogue, was well-tolerated, altered serum reactivity to DWEYS, reduced glomeruli IgG deposition, preserved kidney histology, and delayed SLE onset in NZB/W F1 mice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Christian Grant
- BioMedical Research Models, Inc. , 67 Millbrook Street, Worcester, Massachusetts 01606, United States
| | - Nathalie Meurice
- Department of Research, Mayo Clinic , 13400 East Shea Boulevard, Scottsdale, Arizona 85259, United States
| | | | | | | |
Collapse
|
26
|
Lupus brain fog: a biologic perspective on cognitive impairment, depression, and fatigue in systemic lupus erythematosus. Immunol Res 2016; 63:26-37. [PMID: 26481913 DOI: 10.1007/s12026-015-8716-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Cognitive disturbances, mood disorders and fatigue are common in SLE patients with substantial adverse effects on function and quality of life. Attribution of these clinical findings to immune-mediated disturbances associated with SLE remains difficult and has compromised research efforts in these areas. Improved understanding of the role of the immune system in neurologic processes essential for cognition including synaptic plasticity, long term potentiation and adult neurogenesis suggests multiple potential mechanisms for altered central nervous system function associated with a chronic inflammatory illness such as SLE. This review will focus on the biology of cognition and neuroinflammation in normal circumstances and potential biologic mechanisms for cognitive impairment, depression and fatigue attributable to SLE.
Collapse
|
27
|
Suurmond J, Calise J, Malkiel S, Diamond B. DNA-reactive B cells in lupus. Curr Opin Immunol 2016; 43:1-7. [PMID: 27504587 DOI: 10.1016/j.coi.2016.07.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 07/12/2016] [Accepted: 07/19/2016] [Indexed: 12/20/2022]
Abstract
IgG anti-DNA antibodies are both diagnostic and pathogenic for systemic lupus erythematosus (SLE). They contribute to tissue inflammation through direct tissue binding and to systemic inflammation through activation of Toll-like receptors by nucleic acid-containing immune complexes. IgG DNA-reactive antibodies originate when B cell tolerance mechanisms are impaired. The heterogeneous immune perturbations in SLE lead to the survival and activation of DNA-reactive B cells in various B cell subsets at distinct stages of B cell maturation and differentiation. We propose that the spectrum of B cell alterations and failed tolerance mechanisms for DNA-reactive B cells in lupus patients is best understood by studying genetic risk alleles. This implies that the B cells producing IgG anti-DNA antibodies and the failed tolerance mechanisms(s) will differ across patients. A better understanding of these differences should lead to better patient stratification, improved outcomes of clinical trials, and the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Jolien Suurmond
- Center for Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, North Shore-LIJ, 350 Community Drive, Manhasset, NY 11030, USA
| | - Justine Calise
- Center for Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, North Shore-LIJ, 350 Community Drive, Manhasset, NY 11030, USA; PhD Program in Molecular Medicine, Hofstra-Northwell School of Medicine, 500 Hofstra Blvd, Hempstead, NY 11549, USA
| | - Susan Malkiel
- Center for Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, North Shore-LIJ, 350 Community Drive, Manhasset, NY 11030, USA
| | - Betty Diamond
- Center for Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, North Shore-LIJ, 350 Community Drive, Manhasset, NY 11030, USA.
| |
Collapse
|
28
|
Yadav P, Carr MT, Yu R, Mumbey-Wafula A, Spatz LA. Mapping an epitope in EBNA-1 that is recognized by monoclonal antibodies to EBNA-1 that cross-react with dsDNA. IMMUNITY INFLAMMATION AND DISEASE 2016; 4:362-75. [PMID: 27621818 PMCID: PMC5004290 DOI: 10.1002/iid3.119] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 06/29/2016] [Accepted: 07/01/2016] [Indexed: 01/19/2023]
Abstract
Introduction The Epstein Barr Virus (EBV) has been associated with the autoimmune disease, Systemic Lupus Erythematosus (SLE). EBV nuclear antigen‐I (EBNA‐1) is the major nuclear protein of EBV. We previously generated an IgG monoclonal antibody (MAb) to EBNA‐1, 3D4, and demonstrated that it cross‐reacts with double stranded DNA (dsDNA) and binds the 148 amino acid viral binding site (VBS) in the carboxyl region of EBNA‐1. The aim of the present study was to characterize another antibody to EBNA‐1 that cross‐reacts with dsDNA, compare its immunoglobulin genes to 3D4, and finely map the epitope in EBNA‐1 that is recognized by these cross‐reactive antibodies. Methods We generated an IgM MAb to EBNA‐1, 16D2, from EBNA‐1 injected mice and demonstrated by ELISA that it cross‐reacts with dsDNA and binds the 148 amino acid VBS. We sequenced the variable heavy and light chain genes of 3D4 and 16D2 and compared V gene usage. To more finely map the epitope in EBNA‐1 recognized by these MAbs, we examined their binding by ELISA to 15 overlapping peptides spanning the 148 amino acid domain. Results Sequence analysis revealed that 3D4 and 16D2 utilize different VH and VL genes but identical JH and Jk regions with minimal junctional diversity. This accounts for similarities in their CDR3 regions and may explain their similar dual binding specificity. Epitope mapping revealed 3D4 and 16D2 bind the same peptide in the VBS. Based on the crystal structure of EBNA‐1, we observed that this peptide resides at the base of an exposed proline rich loop in EBNA‐1. Conclusion We have demonstrated that two MAbs that bind EBNA‐1 and cross‐react with dsDNA, recognize the same peptide in the VBS. This peptide may serve as a mimetope for dsDNA and may be of diagnostic and therapeutic value in SLE.
Collapse
Affiliation(s)
- Pragya Yadav
- Department of ChemistryCity College of New York160 Convent AvenueNew YorkNew York10031; Graduate Program in BiochemistryGraduate Center of the City University of New York160 Convent AvenueNew YorkNew York10031
| | - Matthew T Carr
- Department of ChemistryCity College of New York160 Convent AvenueNew YorkNew York10031; Graduate Program in BiochemistryGraduate Center of the City University of New York160 Convent AvenueNew YorkNew York10031
| | - Ruby Yu
- Department of Biology City College of New York 160 Convent Avenue New York New York 10031
| | - Alice Mumbey-Wafula
- Department of Pathobiology, Sophie Davis School of Biomedical Education City College of New York 160 Convent Avenue New York New York 10031
| | - Linda A Spatz
- Department of Pathobiology, Sophie Davis School of Biomedical Education City College of New York 160 Convent Avenue New York New York 10031
| |
Collapse
|
29
|
Xia Y, Eryilmaz E, Der E, Pawar RD, Guo X, Cowburn D, Putterman C. A peptide mimic blocks the cross-reaction of anti-DNA antibodies with glomerular antigens. Clin Exp Immunol 2015; 183:369-79. [PMID: 26482679 DOI: 10.1111/cei.12734] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2015] [Indexed: 12/29/2022] Open
Abstract
Anti-DNA antibodies play a pivotal role in the pathogenesis of lupus nephritis by cross-reacting with renal antigens. Previously, we demonstrated that the binding affinity of anti-DNA antibodies to self-antigens is isotype-dependent. Furthermore, significant variability in renal pathogenicity was seen among a panel of anti-DNA isotypes [derived from a single murine immunoglobulin (Ig)G3 monoclonal antibody, PL9-11] that share identical variable regions. In this study, we sought to select peptide mimics that effectively inhibit the binding of all murine and human anti-DNA IgG isotypes to glomerular antigens. The PL9-11 panel of IgG anti-DNA antibodies (IgG1, IgG2a, IgG2b and IgG3) was used for screening a 12-mer phage display library. Binding affinity was determined by surface plasmon resonance. Enzyme-linked immunosorbent assay (ELISA), flow cytometry and glomerular binding assays were used for the assessment of peptide inhibition of antibody binding to nuclear and kidney antigens. We identified a 12 amino acid peptide (ALWPPNLHAWVP, or 'ALW') which binds to all PL9-11 IgG isotypes. Preincubation with the ALW peptide reduced the binding of the PL9-11 anti-DNA antibodies to DNA, laminin, mesangial cells and isolated glomeruli significantly. Furthermore, we confirmed the specificity of the amino acid sequence in the binding of ALW to anti-DNA antibodies by alanine scanning. Finally, ALW inhibited the binding of murine and human lupus sera to dsDNA and glomeruli significantly. In conclusion, by inhibiting the binding of polyclonal anti-DNA antibodies to autoantigens in vivo, the ALW peptide (or its derivatives) may potentially be a useful approach to block anti-DNA antibody binding to renal tissue.
Collapse
Affiliation(s)
- Y Xia
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.,Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - E Eryilmaz
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - E Der
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.,Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - R D Pawar
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.,Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - X Guo
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - D Cowburn
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - C Putterman
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.,Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
30
|
Antibodies as Mediators of Brain Pathology. Trends Immunol 2015; 36:709-724. [PMID: 26494046 DOI: 10.1016/j.it.2015.09.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 09/17/2015] [Accepted: 09/17/2015] [Indexed: 01/04/2023]
Abstract
The brain is normally sequestered from antibody exposure by the blood brain barrier. However, antibodies can access the brain during fetal development before the barrier achieves full integrity, and in disease states when barrier integrity is compromised. Recent studies suggest that antibodies contribute to brain pathology associated with autoimmune diseases such as systemic lupus erythematosus and neuromyelitis optica, and can lead to transient or permanent behavioral or cognitive abnormalities. We review these findings here and examine the circumstances associated with antibody entry into the brain, the routes of access and the mechanisms that then effect pathology. Understanding these processes and the nature and specificity of neuronal autoantibodies may reveal therapeutic strategies toward alleviating or preventing the neurological pathologies and behavioral abnormalities associated with autoimmune disease.
Collapse
|
31
|
Fernandes-Cerqueira C, Ossipova E, Gunasekera S, Hansson M, Mathsson L, Catrina AI, Sommarin Y, Klareskog L, Lundberg K, Rönnelid J, Göransson U, Jakobsson PJ. Targeting of anti-citrullinated protein/peptide antibodies in rheumatoid arthritis using peptides mimicking endogenously citrullinated fibrinogen antigens. Arthritis Res Ther 2015; 17:155. [PMID: 26059223 PMCID: PMC4484629 DOI: 10.1186/s13075-015-0666-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 05/29/2015] [Indexed: 01/27/2023] Open
Abstract
INTRODUCTION We have previously identified endogenously citrullinated peptides derived from fibrinogen in rheumatoid arthritis (RA) synovial tissues. In this study, we have investigated the auto-antigenicity of four of those citrullinated peptides, and explored their feasibility to target anti-citrullinated protein/peptide antibodies (ACPA). METHODS The autoantigenic potential of the fibrinogen peptides was investigated by screening 927 serum samples from the Epidemiological Investigation of RA (EIRA) cohort on a peptide microarray based on the ImmunoCAP ISAC® system. In order to assay for ACPA blocking, two independent pools of purified ACPA were incubated with the respective targeting peptide prior to binding to cyclic citrullinated peptide (CCP)2 using the CCPlus® ELISA kit. RESULTS Two peptides derived from the fibrinogen α chain, Arg573Cit (563-583) and Arg591Cit (580-600), referred to as Cit573 and Cit591, and two peptides from the fibrinogen β chain, Arg72Cit (62-81) and Arg74Cit (62-81) (Cit72 and Cit74), displayed 65%, 15%, 35%, and 53% of immune reactivity among CCP2-positive RA sera, respectively. In CCP2-negative RA sera, a positive reactivity was detected in 5% (Cit573), 6% (Cit591), 8% (Cit72), and 4% (Cit74). In the competition assay, Cit573 and Cit591 peptides reduced ACPA binding to CCP2 by a maximum of 84% and 63% respectively. An additive effect was observed when these peptides were combined. In contrast, Cit74 and Cit72 were less effective. Cyclization of the peptide structure containing Cit573 significantly increased the blocking efficiency. CONCLUSIONS Here we demonstrate extensive autoantibody reactivity against in vivo citrullinated fibrinogen epitopes, and further show the potential use of these peptides for antagonizing ACPA.
Collapse
Affiliation(s)
- Cátia Fernandes-Cerqueira
- Rheumatology Unit, Department Medicine, Karolinska University Hospital, Solna, Rheumatology Clinic D2:01, 171 76, Stockholm, Sweden.
| | - Elena Ossipova
- Rheumatology Unit, Department Medicine, Karolinska University Hospital, Solna, Rheumatology Clinic D2:01, 171 76, Stockholm, Sweden.
| | - Sunithi Gunasekera
- Division of Pharmacognosy, Department of Medicinal Chemistry, Biomedical Centre, Uppsala University, Husargatan 3, 751 23, Uppsala, Sweden.
| | - Monika Hansson
- Rheumatology Unit, Department Medicine, Karolinska University Hospital, Solna, Rheumatology Clinic D2:01, 171 76, Stockholm, Sweden.
| | - Linda Mathsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Dag Hammarskjölds v 20, 751 85, Uppsala, Sweden.
| | - Anca I Catrina
- Rheumatology Unit, Department Medicine, Karolinska University Hospital, Solna, Rheumatology Clinic D2:01, 171 76, Stockholm, Sweden.
| | - Yngve Sommarin
- Euro-Diagnostica AB, Lundavägen 151, 202 11, Malmö, Sweden.
| | - Lars Klareskog
- Rheumatology Unit, Department Medicine, Karolinska University Hospital, Solna, Rheumatology Clinic D2:01, 171 76, Stockholm, Sweden.
| | - Karin Lundberg
- Rheumatology Unit, Department Medicine, Karolinska University Hospital, Solna, Rheumatology Clinic D2:01, 171 76, Stockholm, Sweden.
| | - Johan Rönnelid
- Department of Immunology, Genetics and Pathology, Uppsala University, Dag Hammarskjölds v 20, 751 85, Uppsala, Sweden.
| | - Ulf Göransson
- Division of Pharmacognosy, Department of Medicinal Chemistry, Biomedical Centre, Uppsala University, Husargatan 3, 751 23, Uppsala, Sweden.
| | - Per-Johan Jakobsson
- Rheumatology Unit, Department Medicine, Karolinska University Hospital, Solna, Rheumatology Clinic D2:01, 171 76, Stockholm, Sweden.
| |
Collapse
|
32
|
Tay SH, Mak A. Anti-NR2A/B Antibodies and Other Major Molecular Mechanisms in the Pathogenesis of Cognitive Dysfunction in Systemic Lupus Erythematosus. Int J Mol Sci 2015; 16:10281-300. [PMID: 25955648 PMCID: PMC4463646 DOI: 10.3390/ijms160510281] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 04/20/2015] [Accepted: 04/29/2015] [Indexed: 12/26/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that affects approximately 1–45.3 per 100,000 people worldwide. Although deaths as a result of active and renal diseases have been substantially declining amongst SLE patients, disease involving the central nervous system (CNS), collectively termed neuropsychiatric systemic lupus erythematosus (NPSLE), remains one of the important causes of death in these patients. Cognitive dysfunction is one of the most common manifestations of NPSLE, which comprises deficits in information-processing speed, attention and executive function, in conjunction with preservation of speech. Albeit a prevalent manifestation of NPSLE, the pathogenetic mechanisms of cognitive dysfunction remain unclear. Recent advances in genetic studies, molecular techniques, neuropathology, neuroimaging and cognitive science have gleaned valuable insights into the pathophysiology of lupus-related cognitive dysfunction. In recent years, a role for autoantibodies, molecular and cellular mechanisms in cognitive dysfunction, has been emerging, challenging our previous concept of the brain as an immune privileged site. This review will focus on the potential pathogenic factors involved in NPSLE, including anti-N-methyl-d-aspartate receptor subunit NR2A/B (anti-NR2A/B) antibodies, matrix metalloproteinase-9, neutrophil extracellular traps and pro-inflammatory mediators. Better understanding of these mechanistic processes will enhance identification of new therapeutic modalities to halt the progression of cognitive decline in SLE patients.
Collapse
Affiliation(s)
- Sen Hee Tay
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 21 Lower Kent Ridge Road, Singapore 119077, Singapore.
- Divison of Rheumatology, Department of Medicine, National University Hospital, National University Health System, 1E Kent Ridge Road, Singapore 119228, Singapore.
| | - Anselm Mak
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 21 Lower Kent Ridge Road, Singapore 119077, Singapore.
- Divison of Rheumatology, Department of Medicine, National University Hospital, National University Health System, 1E Kent Ridge Road, Singapore 119228, Singapore.
| |
Collapse
|
33
|
Suurmond J, Diamond B. Autoantibodies in systemic autoimmune diseases: specificity and pathogenicity. J Clin Invest 2015; 125:2194-202. [PMID: 25938780 DOI: 10.1172/jci78084] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In this Review we focus on the initiation of autoantibody production and autoantibody pathogenicity, with a special emphasis on the targeted antigens. Release of intracellular antigens due to excessive cell death or to ineffective clearance of apoptotic debris, modification of self-antigens during inflammatory responses, and molecular mimicry contribute to the initiation of autoantibody production. We hypothesize that those autoreactive B cells that survive and produce pathogenic autoantibodies have specificity for self-antigens that are TLR ligands. Such B cells experience both B cell receptor (BCR) activation and TLR engagement, leading to an escape from tolerance. Moreover, the autoantibodies they produce form immune complexes that can activate myeloid cells and thereby establish the proinflammatory milieu that further negates tolerance mechanisms of both B and T cells.
Collapse
|
34
|
Mackay M, Tang CC, Volpe BT, Aranow C, Mattis PJ, Korff RA, Diamond B, Eidelberg D. Brain metabolism and autoantibody titres predict functional impairment in systemic lupus erythematosus. Lupus Sci Med 2015; 2:e000074. [PMID: 25861456 PMCID: PMC4379887 DOI: 10.1136/lupus-2014-000074] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 02/11/2015] [Accepted: 03/02/2015] [Indexed: 01/18/2023]
Abstract
Objective We investigated whether systemic lupus erythematosus (SLE) disease duration or serology associate with abnormal regional glucose metabolism as measured with [18F]2-fluoro-2-deoxy-D-glucose positron emission tomography (FDG-PET) and deficits on neuropsychological testing. Methods Subjects with SLE with stable disease activity, without brain damage or clinical symptoms of neuropsychiatric (NP) SLE, stratified by disease duration (short-term (ST)-SLE=disease ≤2 years, long-term (LT)-SLE=disease ≥10 years), underwent clinical assessments, neuropsychological testing, resting FDG-PET scan imaging and measurement of serum titres of antibody to N-methyl-d-aspartate receptor (DNRAb). FDG-PET scans were compared with age-matched and gender-matched healthy controls. Results Subjects with LT-SLE demonstrated hypometabolism in the prefrontal and premotor cortices that correlated with accrued SLE-related damage, but not with DNRAb titre or performance on NP testing. Independent of disease duration, subjects with SLE demonstrated hypermetabolism in the hippocampus and orbitofrontal cortex that correlated with impaired memory performance and mood alterations (depression, anxiety, fatigue). Serum DNRAb also correlated independently with impaired memory performance and increased anxiety. Together, serum DNRAb titre and regional hypermetabolism were more powerful predictors of performance than either alone. Interpretation The presence of serum DNRAbs can account for some aspects of brain dysfunction in patients with SLE, and the addition of regional measurements of resting brain metabolism improves the assessment and precise attribution of central nervous system manifestations related to SLE.
Collapse
Affiliation(s)
- Meggan Mackay
- Center for Autoimmune and Musculoskeletal Disease, The Feinstein Institute for Medical Research , Manhasset, New York , USA
| | - Chris C Tang
- Center for Neurosciences, The Feinstein Institute for Medical Research , Manhasset, New York , USA
| | - Bruce T Volpe
- Center for Autoimmune and Musculoskeletal Disease, The Feinstein Institute for Medical Research , Manhasset, New York , USA
| | - Cynthia Aranow
- Center for Autoimmune and Musculoskeletal Disease, The Feinstein Institute for Medical Research , Manhasset, New York , USA
| | - Paul J Mattis
- Center for Neurosciences, The Feinstein Institute for Medical Research , Manhasset, New York , USA
| | - Ricki A Korff
- Center for Autoimmune and Musculoskeletal Disease, The Feinstein Institute for Medical Research , Manhasset, New York , USA
| | - Betty Diamond
- Center for Autoimmune and Musculoskeletal Disease, The Feinstein Institute for Medical Research , Manhasset, New York , USA
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institute for Medical Research , Manhasset, New York , USA
| |
Collapse
|
35
|
Suurmond J, Zou YR, Kim SJ, Diamond B. Therapeutics to block autoantibody initiation and propagation in systemic lupus erythematosus and rheumatoid arthritis. Sci Transl Med 2015; 7:280ps5. [DOI: 10.1126/scitranslmed.aaa3809] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
36
|
Sankowski R, Mader S, Valdés-Ferrer SI. Systemic inflammation and the brain: novel roles of genetic, molecular, and environmental cues as drivers of neurodegeneration. Front Cell Neurosci 2015; 9:28. [PMID: 25698933 PMCID: PMC4313590 DOI: 10.3389/fncel.2015.00028] [Citation(s) in RCA: 235] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 01/15/2015] [Indexed: 12/20/2022] Open
Abstract
The nervous and immune systems have evolved in parallel from the early bilaterians, in which innate immunity and a central nervous system (CNS) coexisted for the first time, to jawed vertebrates and the appearance of adaptive immunity. The CNS feeds from, and integrates efferent signals in response to, somatic and autonomic sensory information. The CNS receives input also from the periphery about inflammation and infection. Cytokines, chemokines, and damage-associated soluble mediators of systemic inflammation can also gain access to the CNS via blood flow. In response to systemic inflammation, those soluble mediators can access directly through the circumventricular organs, as well as open the blood–brain barrier. The resulting translocation of inflammatory mediators can interfere with neuronal and glial well-being, leading to a break of balance in brain homeostasis. This in turn results in cognitive and behavioral manifestations commonly present during acute infections – including anorexia, malaise, depression, and decreased physical activity – collectively known as the sickness behavior (SB). While SB manifestations are transient and self-limited, under states of persistent systemic inflammatory response the cognitive and behavioral changes can become permanent. For example, cognitive decline is almost universal in sepsis survivors, and a common finding in patients with systemic lupus erythematosus. Here, we review recent genetic evidence suggesting an association between neurodegenerative disorders and persistent immune activation; clinical and experimental evidence indicating previously unidentified immune-mediated pathways of neurodegeneration; and novel immunomodulatory targets and their potential relevance for neurodegenerative disorders.
Collapse
Affiliation(s)
- Roman Sankowski
- Elmezzi Graduate School of Molecular Medicine , Manhasset, NY , USA ; Feinstein Institute for Medical Research , Manhasset, NY , USA
| | - Simone Mader
- Feinstein Institute for Medical Research , Manhasset, NY , USA
| | - Sergio Iván Valdés-Ferrer
- Elmezzi Graduate School of Molecular Medicine , Manhasset, NY , USA ; Feinstein Institute for Medical Research , Manhasset, NY , USA ; Department of Neurology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , México City , Mexico
| |
Collapse
|
37
|
Chalmers SA, Chitu V, Herlitz LC, Sahu R, Stanley ER, Putterman C. Macrophage depletion ameliorates nephritis induced by pathogenic antibodies. J Autoimmun 2014; 57:42-52. [PMID: 25554644 DOI: 10.1016/j.jaut.2014.11.007] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Revised: 11/27/2014] [Accepted: 11/27/2014] [Indexed: 02/07/2023]
Abstract
Kidney involvement affects 40-60% of patients with lupus, and is responsible for significant morbidity and mortality. Using depletion approaches, several studies have suggested that macrophages may play a key role in the pathogenesis of lupus nephritis. However, "off target" effects of macrophage depletion, such as altered hematopoiesis or enhanced autoantibody production, impeded the determination of a conclusive relationship. In this study, we investigated the role of macrophages in mice receiving rabbit anti-glomerular antibodies, or nephrotoxic serum (NTS), an experimental model which closely mimics the immune complex mediated disease seen in murine and human lupus nephritis. GW2580, a selective inhibitor of the colony stimulating factor-1 (CSF-1) receptor kinase, was used for macrophage depletion. We found that GW2580-treated, NTS challenged mice did not develop the increased levels of proteinuria, serum creatinine, and BUN seen in control-treated, NTS challenged mice. NTS challenged mice exhibited significantly increased kidney expression of inflammatory cytokines including RANTES, IP-10, VCAM-1 and iNOS, whereas GW2580-treated mice were protected from the robust expression of these inflammatory cytokines that are associated with lupus nephritis. Quantification of macrophage related gene expression, flow cytometry analysis of kidney single cell suspensions, and immunofluorescence staining confirmed the depletion of macrophages in GW2580-treated mice, specifically within renal glomeruli. Our results strongly implicate a specific and necessary role for macrophages in the development of immune glomerulonephritis mediated by pathogenic antibodies, and support the development of macrophage targeting approaches for the treatment of lupus nephritis.
Collapse
Affiliation(s)
- Samantha A Chalmers
- The Department of Microbiology and Immunology and the Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Violeta Chitu
- The Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Leal C Herlitz
- Department of Pathology, Columbia-Presbyterian Medical Center, New York, NY 10032, USA
| | - Ranjit Sahu
- The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | - E Richard Stanley
- The Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Chaim Putterman
- The Department of Microbiology and Immunology and the Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
38
|
Mostafa GA, El-Sherif DF, Al-Ayadhi LY. Systemic auto-antibodies in children with autism. J Neuroimmunol 2014; 272:94-8. [PMID: 24837704 DOI: 10.1016/j.jneuroim.2014.04.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 04/12/2014] [Accepted: 04/17/2014] [Indexed: 01/18/2023]
Abstract
Autoimmunity to central nervous system may have a role in the pathogenesis of autism. A subset of anti-ds-DNA antibodies has been recently proved to be pathogenic to the brain as well as to the kidney. Due to the paucity of studies investigating the frequency of systemic auto-antibodies in autism, we are the first to investigate the frequency of anti-ds-DNA antibodies in a group of autistic children. The seropositivity of anti-nuclear antibodies (ANA) was also investigated. Serum anti-ds-DNA antibodies and ANA were measured in 100 autistic children, aged between 4 and 11 years, in comparison to 100 healthy-matched children. The seropositivity of anti-ds-DNA antibodies and ANA in autistic children was 34% and 25%, respectively. In addition, 42% of autistic children were seropositive for anti-ds-DNA antibodies and/or ANA. The frequencies of anti-ds-DNA antibodies and ANA in autistic children were significantly higher than that in healthy children (4% and 2%, respectively), (P<0.001 and P<0.001, respectively). Autistic children with a family history of autoimmunity (45%) had significantly higher frequency of serum anti-ds-DNA antibodies (48.9%) than patients without such a history (21.8%), P=0.008. There was a significant positive association between the seropositivity of anti-ds-DNA antibodies and ANA (P<0.001). In conclusion, anti-ds-DNA antibodies and ANA were found in the sera of a subgroup of autistic children. However, replication studies of larger samples are warranted to validate whether these antibodies are a mere association or have a pathogenic role in some autistic children.
Collapse
Affiliation(s)
- Gehan A Mostafa
- Department of Pediatrics, Faculty of Medicine, Ain Shams University, Cairo, Egypt; Autism Research and Treatment Center, Al-Amodi Autism Research Chair, Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia.
| | - Dalia F El-Sherif
- Department of Pediatrics, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Laila Y Al-Ayadhi
- Autism Research and Treatment Center, Al-Amodi Autism Research Chair, Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
39
|
Rahman A, Giles IP. Structure and function of autoantibodies and their role in autoimmune rheumatic diseases. Expert Rev Clin Immunol 2014; 2:225-36. [DOI: 10.1586/1744666x.2.2.225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
40
|
Cerqueira CF, Klareskog L, Jakobsson PJ. Neutralization of AntiCitrullinated Protein Antibodies in Rheumatoid Arthritis - A Way to Go? Basic Clin Pharmacol Toxicol 2013; 114:13-7. [DOI: 10.1111/bcpt.12157] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 10/06/2013] [Indexed: 11/30/2022]
Affiliation(s)
- Cátia F. Cerqueira
- Rheumatology Unit; Department of Medicine; Karolinska Institutet; Karolinska University Hospital; Stockholm Sweden
| | - Lars Klareskog
- Rheumatology Unit; Department of Medicine; Karolinska Institutet; Karolinska University Hospital; Stockholm Sweden
| | - Per-Johan Jakobsson
- Rheumatology Unit; Department of Medicine; Karolinska Institutet; Karolinska University Hospital; Stockholm Sweden
| |
Collapse
|
41
|
Franchin G, Son M, Kim SJ, Ben-Zvi I, Zhang J, Diamond B. Anti-DNA antibodies cross-react with C1q. J Autoimmun 2013; 44:34-9. [PMID: 23834843 DOI: 10.1016/j.jaut.2013.06.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 06/03/2013] [Indexed: 12/19/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder that involves multiple organ systems and typically presents as a chronic inflammatory disease. Antibodies to double-stranded (ds) DNA are present in approximately 70% of patients and form nucleic acid containing immune complexes which activate dendritic cells through engagement of toll-like receptors, leading to a pro-inflammatory, pro-immunogenic milieu. In addition, anti-dsDNA antibodies deposit in kidneys to initiate glomerulonephritis. Antibodies to C1q have also been implicated in lupus nephritis and are found in 30-50% of patients. C1q is a known suppressor of immune activation and C1q deficiency is the strongest risk factor for SLE. We previously identified a subset of anti-DNA antibodies that binds the N-methyl-D-aspartate receptor. We now show that both mouse and human anti-DNA antibodies with this specificity bind C1q. These antibodies bind to Clq in glomeruli and exhibit decreased glomerular deposition in the absence of C1q. We propose that this subset of anti-DNA antibodies participates in lupus pathogenesis through direct targeting of C1q on glomeruli and also through removal of soluble C1q thereby limiting the ability of C1q to mediate immune homeostasis.
Collapse
Affiliation(s)
- Giovanni Franchin
- The Center for Autoimmune and Musculoskeletal Disease, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
Autoimmune diseases currently affect 5-7% of the world's population; in most diseases there are circulating autoantibodies. Brain-reactive antibodies are present in approximately 2-3% of the general population but do not usually contribute to brain pathology. These antibodies penetrate brain tissue only early in development or under pathologic conditions. This restriction on their pathogenicity and the lack of correlation between serum titers and brain pathology have, no doubt, contributed to a delayed appreciation of the contribution of autoantibodies in diseases of the central nervous system. Nonetheless, it is increasingly clear that antibodies can cause damage in the brain and likely initiate or aggravate multiple neurologic conditions; brain-reactive antibodies contribute to symptomatology in autoimmune disease, infectious disease, and malignancy.
Collapse
Affiliation(s)
- B Diamond
- Feinstein Institute for Medical Research, Manhasset, New York 11030, USA.
| | | | | | | | | |
Collapse
|
43
|
The constant region affects antigen binding of antibodies to DNA by altering secondary structure. Mol Immunol 2013; 56:28-37. [PMID: 23665381 DOI: 10.1016/j.molimm.2013.04.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 04/08/2013] [Accepted: 04/09/2013] [Indexed: 11/23/2022]
Abstract
We previously demonstrated an important role of the constant region in the pathogenicity of anti-DNA antibodies. To determine the mechanisms by which the constant region affects autoantibody binding, a panel of isotype-switch variants (IgG1, IgG2a, IgG2b) was generated from the murine PL9-11 IgG3 autoantibody. The affinity of the PL9-11 antibody panel for histone was measured by surface plasmon resonance (SPR). Tryptophan fluorescence was used to determine wavelength shifts of the antibody panel upon binding to DNA and histone. Finally, circular dichroism spectroscopy was used to measure changes in secondary structure. SPR analysis revealed significant differences in histone binding affinity between members of the PL9-11 panel. The wavelength shifts of tryptophan fluorescence emission were found to be dependent on the antibody isotype, while circular dichroism analysis determined that changes in antibody secondary structure content differed between isotypes upon antigen binding. Thus, the antigen binding affinity is dependent on the particular constant region expressed. Moreover, the effects of antibody binding to antigen were also constant region dependent. Alteration of secondary structures influenced by constant regions may explain differences in fine specificity of anti-DNA antibodies between antibodies with similar variable regions, as well as cross-reactivity of anti-DNA antibodies with non-DNA antigens.
Collapse
|
44
|
Yoshio T, Okamoto H, Hirohata S, Minota S. IgG anti-NR2 glutamate receptor autoantibodies from patients with systemic lupus erythematosus activate endothelial cells. ACTA ACUST UNITED AC 2013; 65:457-63. [DOI: 10.1002/art.37745] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 10/04/2012] [Indexed: 11/09/2022]
|
45
|
Lakhan SE, Caro M, Hadzimichalis N. NMDA Receptor Activity in Neuropsychiatric Disorders. Front Psychiatry 2013; 4:52. [PMID: 23772215 PMCID: PMC3677126 DOI: 10.3389/fpsyt.2013.00052] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Accepted: 05/25/2013] [Indexed: 01/03/2023] Open
Abstract
N-Methyl-d-aspartate (NMDA) receptors play a variety of physiologic roles and their proper signaling is essential for cellular homeostasis. Any disruption in this pathway, leading to either enhanced or decreased activity, may result in the manifestation of neuropsychiatric pathologies such as schizophrenia, mood disorders, substance induced psychosis, Huntington's disease, Alzheimer's disease, and neuropsychiatric systemic lupus erythematosus. Here, we explore the notion that the overlap in activity of at least one biochemical pathway, the NMDA receptor pathway, may be the link to understanding the overlap in psychotic symptoms between diseases. This review intends to present a broad overview of those neuropsychiatric disorders for which alternations in NMDA receptor activity is prominent thus suggesting that continued direction of pharmaceutical intervention to this pathway may present a viable option for managing symptoms.
Collapse
Affiliation(s)
- Shaheen E Lakhan
- Biosciences Department, Global Neuroscience Initiative Foundation , Beverly Hills, CA , USA ; Neurological Institute, Cleveland Clinic , Cleveland, OH , USA
| | | | | |
Collapse
|
46
|
Gesheva V, Szekeres Z, Mihaylova N, Dimitrova I, Nikolova M, Erdei A, Prechl J, Tchorbanov A. Generation of gene-engineered chimeric DNA molecules for specific therapy of autoimmune diseases. Hum Gene Ther Methods 2012; 23:357-65. [PMID: 23075110 PMCID: PMC4015069 DOI: 10.1089/hgtb.2012.051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 10/15/2012] [Indexed: 12/20/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the development of self-reactive B and T cells and autoantibody production. In particular, double-stranded DNA-specific B cells play an important role in lupus progression, and their selective elimination is a reasonable approach for effective therapy of SLE. DNA-based vaccines aim at the induction of immune response against the vector-encoded antigen. Here, we are exploring, as a new DNA-based therapy of SLE, a chimeric DNA molecule encoding a DNA-mimotope peptide, and the Fv but not the immunogenic Fc fragment of an FcγRIIb-specific monoclonal antibody. This DNA construct was inserted in the expression vector pNut and used as a naked DNA vaccine in a mouse model of lupus. The chimeric DNA molecule can be expressed in eukaryotic cells and cross-links cell surface receptors on DNA-specific B cells, delivering an inhibitory intracellular signal. Intramuscular administration of the recombinant DNA molecule to lupus-prone MRL/lpr mice prevented increase in IgG anti-DNA antibodies and was associated with a low degree of proteinuria, modulation of cytokine profile, and suppression of lupus nephritis.
Collapse
Affiliation(s)
- Vera Gesheva
- Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Wang F, Tian F, Whitman SA, Zhang DD, Nishinaka T, Zhang N, Jiang T. Regulation of transforming growth factor β1-dependent aldose reductase expression by the Nrf2 signal pathway in human mesangial cells. Eur J Cell Biol 2012; 91:774-81. [PMID: 22951256 DOI: 10.1016/j.ejcb.2012.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 07/13/2012] [Accepted: 07/16/2012] [Indexed: 01/26/2023] Open
Abstract
Aldose reductase (AR) is a key enzyme in the alternative glucose metabolism pathway, the polyol pathway. To date, AR is known to be involved in several secondary complications of diabetes and various kidney diseases. The goal of this study was to elucidate how the Nrf2-anti-oxidant response element (ARE) signal pathway plays a role in TGFβ1's regulation of AR expression in human renal mesangial cells (HRMCs). As an in vitro model system, HRMCs were used to investigate AR mRNA by qPCR, protein by Western blot and enzymatic activity by spectrophotometric assay. The ability of TGFβ1 to induce reactive oxygen species (ROS) in cells was measured by electron-spin resonance (ESR) trapping method. Reporter assays were used to test the activity of the AR promoter region, and ChIP was employed to test the direct binding of Nrf2 with the endogenous AR promoter. Treatment of HRMCs with TGFβ1 up-regulated the expression of AR mRNA, protein, and activity level. Additionally, TGFβ1 rapidly increased cellular ROS levels, which in turn activated the Nrf2-ARE pathway. Either inhibition of ROS production or knockdown of Nrf2 in HRMCs decreased the TGFβ1-induction of AR expression. Nrf2 regulated AR luciferase activity specifically via two AREs within the AR promoter, and bound directly to the endogenous AR promoter. Furthermore, the TGFβ1-mediated expression of AR required Nrf2 and was significantly abrogated in Nrf2-/- cells. These data show the regulation of AR by TGFβ1 is induced by TGFβ1 stimulation of ROS, which activates the Nrf2-ARE pathway allowing Nrf2 to directly increase AR expression in HRMCs.
Collapse
Affiliation(s)
- Fei Wang
- Department of Pathology, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
48
|
Xia Y, Campbell SR, Broder A, Herlitz L, Abadi M, Wu P, Michaelson JS, Burkly LC, Putterman C. Inhibition of the TWEAK/Fn14 pathway attenuates renal disease in nephrotoxic serum nephritis. Clin Immunol 2012; 145:108-21. [PMID: 22982296 DOI: 10.1016/j.clim.2012.08.008] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 08/07/2012] [Accepted: 08/08/2012] [Indexed: 02/08/2023]
Abstract
Previously it was shown that the TNF superfamily member TWEAK (TNFSF12) acts through its receptor, Fn14, to promote proinflammatory responses in kidney cells, including the production of MCP-1, RANTES, IP-10 and KC. In addition, the TWEAK/Fn14 pathway promotes mesangial cell proliferation, vascular cell activation, and renal cell death. To study the relevance of the TWEAK/Fn14 pathway in the pathogenesis of antibody-induced nephritis using the mouse model of nephrotoxic serum nephritis (NTN), we induced NTN by passive transfer of rabbit anti-glomerular antibodies into Fn14 knockout (KO) and wild type (WT) mice. Severe proteinuria as well as renal histopathology were induced in WT but not in Fn14 KO mice. Similarly, a pharmacologic approach of anti-TWEAK mAb administration into WT mice in the NTN model significantly ameliorated proteinuria and improved kidney histology. Anti-TWEAK treatment did not affect the generation of mouse anti-rabbit antibodies; however, within the kidney there was a significant decrease in glomerular immunoglobulin deposition, as well as macrophage infiltrates and tubulointerstitial fibrosis. The mechanism of action is most likely due to reductions in downstream targets of TWEAK/Fn14 signaling, including reduced renal expression of MCP-1, VCAM-1, IP-10, RANTES as well as Fn14 itself, and other molecular pathways associated with fibrosis in anti-TWEAK treated mice. Thus, TWEAK/Fn14 interactions are instrumental in the pathogenesis of nephritis in the NTN model, apparently mediating a cascade of pathologic events locally in the kidney rather than by impacting the systemic immune response. Disrupting TWEAK/Fn14 interactions may be an innovative kidney-protective approach for the treatment of lupus nephritis and other antibody-induced renal diseases.
Collapse
Affiliation(s)
- Yumin Xia
- Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Systemic lupus erythematosus is an autoimmune disease characterized by antibodies that bind target autoantigens in multiple organs in the body. In peripheral organs, immune complexes engage the complement cascade, recruiting blood-borne inflammatory cells and initiating tissue inflammation. Immune complex-mediated activation of Fc receptors on infiltrating blood-borne cells and tissue resident cells amplifies an inflammatory cascade with resulting damage to tissue function, ultimately leading to tissue destruction. This pathophysiology appears to explain tissue injury throughout the body, except in the central nervous system. This review addresses a paradigm we have developed for autoantibody-mediated brain damage. This paradigm suggests that antibody-mediated brain disease does not depend on immune complex formation but rather on antibody-mediated alterations in neuronal activation and survival. Moreover, antibodies only access brain tissue when blood-brain barrier integrity is impaired, leading to a lack of concurrence of brain disease and tissue injury in other organs. We discuss the implications of this model for lupus and for identifying other antibodies that may contribute to brain disease.
Collapse
Affiliation(s)
- Betty Diamond
- Center for Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, North Shore-Long Island Jewish Health System, Manhasset, NY 11030, USA.
| | | |
Collapse
|
50
|
Bosch X, Ramos-Casals M, Khamashta MA. The DWEYS peptide in systemic lupus erythematosus. Trends Mol Med 2012; 18:215-23. [DOI: 10.1016/j.molmed.2012.01.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 01/24/2012] [Accepted: 01/27/2012] [Indexed: 11/26/2022]
|