1
|
Olate-Briones A, Albornoz-Muñoz S, Rodriguez-Arriaza F, Rojas-Henriquez V, Rojas SS, Escobedo N, Herrada AA. Depression-like behavior is associated with changes in the meningeal lymphatic vasculature and meningeal B cells in a murine lupus model. J Leukoc Biol 2025; 117:qiaf016. [PMID: 40276929 DOI: 10.1093/jleuko/qiaf016] [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: 11/11/2024] [Indexed: 04/26/2025] Open
Abstract
Meningeal lymphatic vasculature (mLV) comprises a network of vessels responsible for draining immune cells and fluid from the central nervous system (CNS) into the deep cervical lymph nodes. While changes in mLV function have been implicated in several neurodegenerative disorders, its role in autoimmune diseases is less clear. Systemic lupus erythematosus (SLE) is an autoimmune disease affecting multiple organs. When SLE affects the CNS, it is known as neuropsychiatric lupus (NPSLE), although the status of mLV during NPSLE has not been yet evaluated. Here, by using the lupus FcγRIIb-/- murine model, we found that this model develops NPSLE along with increased mLV coverage and function at 4 mo of age. Altered B cell developmental stages were evident in this lupus mouse model. In fact, increased B cell clusters in the meninges of FcγRIIb-/- mice were also observed. These findings suggest that mLV morphology and function are increased in FcγRIIb-/- mice together with changes in the meningeal B cell population that could have an impact on NPSLE symptoms.
Collapse
MESH Headings
- Animals
- Meninges/pathology
- Meninges/immunology
- Lymphatic Vessels/pathology
- Lymphatic Vessels/immunology
- B-Lymphocytes/immunology
- B-Lymphocytes/pathology
- Disease Models, Animal
- Mice
- Lupus Vasculitis, Central Nervous System/pathology
- Lupus Vasculitis, Central Nervous System/immunology
- Mice, Knockout
- Receptors, IgG/genetics
- Receptors, IgG/deficiency
- Female
- Mice, Inbred C57BL
- Lupus Erythematosus, Systemic/pathology
- Lupus Erythematosus, Systemic/immunology
- Behavior, Animal
Collapse
Affiliation(s)
- Alexandra Olate-Briones
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, 3467987 Talca, Chile
| | - Sofía Albornoz-Muñoz
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, 3467987 Talca, Chile
| | - Francisca Rodriguez-Arriaza
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, 3467987 Talca, Chile
| | - Victor Rojas-Henriquez
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, 3467987 Talca, Chile
| | - Stefanny S Rojas
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, 3467987 Talca, Chile
| | - Noelia Escobedo
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, 3467987 Talca, Chile
| | - Andrés A Herrada
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, 3467987 Talca, Chile
| |
Collapse
|
2
|
Wang S, Xu Z, Li J, Lai Z, Shi C, He J. A cross-sectional study on white matter hyperitensity in patients at the initial diagnosis of neuropsychiatric SLE: Correlation with Clinical and Laboratory Findings. Clin Rheumatol 2025; 44:1571-1580. [PMID: 40009312 DOI: 10.1007/s10067-025-07379-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/12/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025]
Abstract
OBJECTIVES This study aimed to investigate the differences between neuropsychiatric systemic lupus erythematosus (NPSLE) patients with and without white matter hyperintensity (WMH) on magnetic resonance imaging (MRI) and to identify factors independently associated with the development of WMH in NPSLE. METHOD A comparative analysis was conducted on 84 NPSLE patients hospitalized at Peking University People's Hospital from 2015 to 2022 at the initial diagnosis of NPSLE. Patients were categorized based on the presence of WMH on brain MRI scans. Demographic data, clinical characteristics, and laboratory parameters were reviewed and statistically analyzed. RESULTS The study included 84 NPSLE patients, 50% exhibiting WMH (NPSLE-WMH +). At the initial diagnosis of NPSLE, the NPSLE-WMH + group showed significantly higher levels of antinuclear antibody (ANA) titer, anti-double-stranded DNA antibody (anti-dsDNA), anti-nucleosome antibody (ANuA), and lower serum C3 levels. The proportion of patients with reduced WBC, elevated anti-dsDNA, elevated ANuA, elevated anticardiolipin antibody (ACA), positive urinary protein (UPR), and positive ANA was higher in the NPSLE-WMH + group. Univariate and multivariate analyses revealed that positive UPR (p = 0.040), positive ANA (p = 0.025), elevated anti-dsDNA (0.047), and elevated ACA (p = 0.025) were potentially independent factors associated with WMH development in NPSLE patients. CONCLUSIONS This study provides novel insights into the clinical and laboratory differences between NPSLE patients with and without WMH, identifying specific independently associated factors for WMH development. These findings may contribute to a better understanding of this intricate disease. Key Points • Although WMH is one of the most frequently observed lesions on MRI in patients with NPSLE, previous literature has given limited attention to it. This study focuses on the differences between NPSLE patients with and without WMH, unveiling independently associated factors for developing WMH in this disease.
Collapse
Affiliation(s)
- Shiyang Wang
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Haidian District, No. 51, North Huayuan Road, Beijing, China
| | - Zhihu Xu
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC3004, Australia
| | - Jiaxi Li
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People's Hospital, Department of Rheumatology and Immunology, No. 11, Xizhimen South Street, Xicheng District, Beijing, China
| | - Zhanhong Lai
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People's Hospital, Department of Rheumatology and Immunology, No. 11, Xizhimen South Street, Xicheng District, Beijing, China
| | - Chuan Shi
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Haidian District, No. 51, North Huayuan Road, Beijing, China.
| | - Jing He
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People's Hospital, Department of Rheumatology and Immunology, No. 11, Xizhimen South Street, Xicheng District, Beijing, China.
| |
Collapse
|
3
|
Chen S, Ren H, Fan S, Zhang S, Li M, Guan H. Autoimmune encephalitis associated with anti-LGI1 antibody: a potential cause of neuropsychiatric systemic lupus erythematosus. Lupus Sci Med 2025; 12:e001429. [PMID: 39965877 PMCID: PMC11836784 DOI: 10.1136/lupus-2024-001429] [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: 10/22/2024] [Accepted: 01/30/2025] [Indexed: 02/20/2025]
Abstract
OBJECTIVE To investigate the clinical features and treatment outcomes of anti-leucine-rich glioma-inactivated 1 (anti-LGI1) encephalitis in patients with SLE. METHODS Between October 2014 and April 2024, serum or cerebrospinal fluid samples were collected from 332 patients with SLE suspected of autoimmune encephalitis. Cell-based assays were used to detect autoimmune antibodies, including anti-LGI1 antibodies. Four patients tested positive for anti-LGI1 antibodies, and their clinical, radiological and treatment data were analysed. RESULTS All four patients exhibited signs of limbic encephalitis, including short-term memory deficits, seizures and psychiatric disturbances. Two cases also presented with faciobrachial dystonic seizures. MRI findings revealed hyperintense basal ganglia lesions in two patients. Treatment with corticosteroids, intravenous immunoglobulin and mycophenolate mofetil led to significant improvement in three patients, with no relapses during a follow-up period ranging from 33 to 60 months. One patient succumbed to pneumonia despite initial improvement of neurological function. CONCLUSION Screening for anti-LGI1 antibodies in patients with neuropsychiatric systemic lupus erythematosus (NPSLE) is crucial when limbic encephalitis presents, as it enables timely and effective treatment, potentially improving patients' outcomes. Additional basic and clinical research is required to clarify the pathogenic role of these antibodies in NPSLE.
Collapse
Affiliation(s)
- Sixian Chen
- Neurology, Peking Union Medical College Hospital, Beijing, China
| | - Haitao Ren
- Neurology, Peking Union Medical College Hospital, Beijing, China
| | - Siyuan Fan
- Neurology, Peking Union Medical College Hospital, Beijing, China
| | - Shangzhu Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, China
| | - Mengtao Li
- Department of Rheumatology, Peking Union Medical College Hospital, Beijing, China
| | - Hongzhi Guan
- Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
4
|
Wang Y, Zhao R, Liang Q, Ni S, Yang M, Qiu L, Ji J, Gu Z, Dong C. Organ-based characterization of B cells in patients with systemic lupus erythematosus. Front Immunol 2025; 16:1509033. [PMID: 39917309 PMCID: PMC11798990 DOI: 10.3389/fimmu.2025.1509033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic, inflammatory, and progressive autoimmune disease. The unclear pathogenesis, high heterogeneity, and prolonged course of the disease present significant challenges for effective clinical management of lupus patients. Dysregulation of the immune system and disruption of immune tolerance, particularly through the abnormal activation of B lymphocytes and the production of excessive autoantibodies, lead to widespread inflammation and tissue damage, resulting in multi-organ impairment. Currently, there is no systematic review that examines the specificity of B cell characteristics and pathogenic mechanisms across various organs. This paper reviews current research on B cells in lupus patients and summarizes the distinct characteristics of B cells in different organs. By integrating clinical manifestations of organ damage in patients with a focus on the organ-specific features of B cells, we provide a new perspective on enhancing the efficacy of lupus-targeted B cell therapy strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhifeng Gu
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Chen Dong
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| |
Collapse
|
5
|
Rodríguez RD, Alarcón-Riquelme ME. Exploring the contribution of genetics on the clinical manifestations of systemic lupus erythematosus. Best Pract Res Clin Rheumatol 2024; 38:101971. [PMID: 39013664 DOI: 10.1016/j.berh.2024.101971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/18/2024]
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease characterized by diverse clinical manifestations affecting multiple organs and systems. The understanding of genetic factors underlying the various manifestations of SLE has evolved considerably in recent years. This review provides an overview of the genetic implications in some of the most prevalent manifestations of SLE, including renal involvement, neuropsychiatric, cutaneous, constitutional, musculoskeletal, and cardiovascular manifestations. We discuss the current state of knowledge regarding the genetic basis of these manifestations, highlighting key genetic variants and pathways implicated in their pathogenesis. Additionally, we explore the clinical implications of genetic findings, including their potential role in risk stratification, prognosis, and personalized treatment approaches for patients with SLE. Through a comprehensive examination of the genetic landscape of SLE manifestations, this review aims to provide insights into the underlying mechanisms driving disease heterogeneity and inform future research directions in this field.
Collapse
Affiliation(s)
- Ruth D Rodríguez
- Center for Genomics and Oncological Research (GENyO). Pfizer/ University of Granada/ Andalusian Government, Spain
| | - Marta E Alarcón-Riquelme
- Center for Genomics and Oncological Research (GENyO). Pfizer/ University of Granada/ Andalusian Government, Spain; Institute for Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
6
|
Al-Ewaidat OA, Naffaa MM. Deciphering Mechanisms, Prevention Strategies, Management Plans, Medications, and Research Techniques for Strokes in Systemic Lupus Erythematosus. MEDICINES (BASEL, SWITZERLAND) 2024; 11:15. [PMID: 39189161 PMCID: PMC11348055 DOI: 10.3390/medicines11070015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/30/2024] [Accepted: 07/26/2024] [Indexed: 08/28/2024]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune rheumatic condition characterized by an unpredictable course and a wide spectrum of manifestations varying in severity. Individuals with SLE are at an increased risk of cerebrovascular events, particularly strokes. These strokes manifest with a diverse range of symptoms that cannot be solely attributed to conventional risk factors, underscoring their significance among the atypical risk factors in the context of SLE. This complexity complicates the identification of optimal management plans and the selection of medication combinations for individual patients. This susceptibility is further complicated by the nuances of neuropsychiatric SLE, which reveals a diverse array of neurological symptoms, particularly those associated with ischemic and hemorrhagic strokes. Given the broad range of clinical presentations and associated risks linking strokes to SLE, ongoing research and comprehensive care strategies are essential. These efforts are critical for improving patient outcomes by optimizing management strategies and discovering new medications. This review aims to elucidate the pathological connection between SLE and strokes by examining neurological manifestations, risk factors, mechanisms, prediction and prevention strategies, management plans, and available research tools and animal models. It seeks to explore this medical correlation and discover new medication options that can be tailored to individual SLE patients at risk of stroke.
Collapse
Affiliation(s)
- Ola A. Al-Ewaidat
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA;
| | - Moawiah M. Naffaa
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
| |
Collapse
|
7
|
Kerns S, Owen KA, Schwalbe D, Grammer AC, Lipsky PE. Examination of the shared genetic architecture between multiple sclerosis and systemic lupus erythematosus facilitates discovery of novel lupus risk loci. Hum Genet 2024; 143:703-719. [PMID: 38609570 DOI: 10.1007/s00439-024-02672-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 03/24/2024] [Indexed: 04/14/2024]
Abstract
Systemic Lupus Erythematosus (SLE) is an autoimmune disease with heterogeneous manifestations, including neurological and psychiatric symptoms. Genetic association studies in SLE have been hampered by insufficient sample size and limited power compared to many other diseases. Multiple Sclerosis (MS) is a chronic relapsing autoimmune disease of the central nervous system (CNS) that also manifests neurological and immunological features. Here, we identify a method of leveraging large-scale genome wide association studies (GWAS) in MS to identify novel genetic risk loci in SLE. Statistical genetic comparison methods including linkage disequilibrium score regression (LDSC) and cross-phenotype association analysis (CPASSOC) to identify genetic overlap in disease pathophysiology, traditional 2-sample and novel PPI-based mendelian randomization to identify causal associations and Bayesian colocalization were applied to association studies conducted in MS to facilitate discovery in the smaller, more limited datasets available for SLE. Pathway analysis using SNP-to-gene mapping identified biological networks composed of molecular pathways with causal implications for CNS disease in SLE specifically, as well as pathways likely causal of both pathologies, providing key insights for therapeutic selection.
Collapse
Affiliation(s)
- Sophia Kerns
- AMPEL BioSolutions, LLC, Charlottesville, VA, 22902, USA.
- The RILITE Research Institute, Charlottesville, VA, 22902, USA.
| | - Katherine A Owen
- AMPEL BioSolutions, LLC, Charlottesville, VA, 22902, USA
- The RILITE Research Institute, Charlottesville, VA, 22902, USA
| | - Dana Schwalbe
- AMPEL BioSolutions, LLC, Charlottesville, VA, 22902, USA
- The RILITE Research Institute, Charlottesville, VA, 22902, USA
| | - Amrie C Grammer
- AMPEL BioSolutions, LLC, Charlottesville, VA, 22902, USA
- The RILITE Research Institute, Charlottesville, VA, 22902, USA
| | - Peter E Lipsky
- AMPEL BioSolutions, LLC, Charlottesville, VA, 22902, USA
- The RILITE Research Institute, Charlottesville, VA, 22902, USA
| |
Collapse
|
8
|
Yun Y, Wang X, Xu J, Jin C, Chen J, Wang X, Wang J, Qin L, Yang P. Pristane induced lupus mice as a model for neuropsychiatric lupus (NPSLE). BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2023; 19:3. [PMID: 36765366 PMCID: PMC9921421 DOI: 10.1186/s12993-023-00205-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 01/13/2023] [Indexed: 02/12/2023]
Abstract
BACKGROUND The pristane-induced lupus (PIL) model is a useful tool for studying environmental-related systemic lupus erythematosus (SLE). However, neuropsychiatric manifestations in this model have not been investigated in detail. Because neuropsychiatric lupus (NPSLE) is an important complication of SLE, we investigated the neuropsychiatric symptoms in the PIL mouse model to evaluate its suitability for NPSLE studies. RESULTS PIL mice showed olfactory dysfunction accompanied by an anxiety- and depression-like phenotype at month 2 or 4 after pristane injection. The levels of cytokines (IL-1β, IFN-α, IFN-β, IL-10, IFN-γ, IL-6, TNF-α and IL-17A) and chemokines (CCL2 and CXCL10) in the brain and blood-brain barrier (BBB) permeability increased significantly from week 2 or month 1, and persisted throughout the observed course of the disease. Notably, IgG deposition in the choroid plexus and lateral ventricle wall were observed at month 1 and both astrocytes and microglia were activated. Persistent activation of astrocytes was detected throughout the observed course of the disease, while microglial activation diminished dramatically at month 4. Lipofuscin deposition, a sign of neuronal damage, was detected in cortical and hippocampal neurons from month 4 to 8. CONCLUSION PIL mice exhibit a series of characteristic behavioral deficits and pathological changes in the brain, and therefore might be suitable for investigating disease pathogenesis and for evaluating potential therapeutic targets for environmental-related NPSLE.
Collapse
Affiliation(s)
- Yang Yun
- grid.412467.20000 0004 1806 3501Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xuejiao Wang
- grid.412449.e0000 0000 9678 1884Department of Physiology, China Medical University, Shenyang, China
| | - Jingyi Xu
- grid.412636.40000 0004 1757 9485Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Chenye Jin
- grid.412636.40000 0004 1757 9485Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Jingyu Chen
- grid.412449.e0000 0000 9678 1884Department of Physiology, China Medical University, Shenyang, China
| | - Xueru Wang
- grid.412449.e0000 0000 9678 1884Department of Physiology, China Medical University, Shenyang, China
| | - Jianing Wang
- grid.412636.40000 0004 1757 9485Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Ling Qin
- Department of Physiology, China Medical University, Shenyang, China.
| | - Pingting Yang
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
9
|
Capasso N, Palladino R, Cerbone V, Spiezia AL, Covelli B, Fiore A, Lanzillo R, Carotenuto A, Petracca M, Stanziola L, Scalia G, Brescia Morra V, Moccia M. Ocrelizumab effect on humoral and cellular immunity in multiple sclerosis and its clinical correlates: a 3-year observational study. J Neurol 2023; 270:272-282. [PMID: 36048265 PMCID: PMC9813008 DOI: 10.1007/s00415-022-11350-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 08/18/2022] [Accepted: 08/21/2022] [Indexed: 01/13/2023]
Abstract
OBJECTIVE We aim to evaluate 3-year effects of ocrelizumab (humanized anti-CD20 monoclonal antibody for the treatment of multiple sclerosis (MS)) on lymphocytes, neutrophils and immunoglobulins: (1) when compared with pre-infusion assessment; (2) over the course of treatment; and (3) possible clinical correlates of the observed immunological modifications. METHODS This real-world observational cohort study has been conducted on prospectively collected data from 78 MS patients (mean age 47.8 ± 10.5 years; females 48.7%) commencing on ocrelizumab from 2018, with mean follow-up of 36.5 ± 6.8 months. Clinical data and blood samples were collected every three months. Total lymphocyte count and subpopulations were assessed on peripheral blood using flow cytometry. Serum immunoglobulins were evaluated with nephelometry. RESULTS When compared with pre-infusion values, we observed reduction of total, CD19 and CD20 lymphocyte counts; however, after the first infusion, their levels remained substantially stable. Over time we observed a progressive reduction of CD8 lymphocytes, while no changes were observed for CD4, CD27, CD3CD27, and CD19CD27. After the first infusion, we observed reduction in IgG, which further decreased during the follow-up. Higher probability of EDSS progression was associated with reduced modulation of CD8 lymphocytes. INTERPRETATION Ocrelizumab affects both humoral and cellular immune responses. Disability progression over the follow-up was associated with lower CD8 cytotoxic T-lymphocyte reduction. Changes in humoral response are immediate and sustained, while modulation of cellular immunity occurs progressively through regular re-treatment, and is related to clinical stability.
Collapse
Affiliation(s)
- Nicola Capasso
- Multiple Sclerosis Unit, Federico II University Hospital, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Raffaele Palladino
- Department of Public Health, Federico II University of Naples, Naples, Italy
| | | | - Antonio Luca Spiezia
- Multiple Sclerosis Unit, Federico II University Hospital, Via Sergio Pansini 5, 80131, Naples, Italy
- Department of Neuroscience, Reproductive Sciences and Odontostomatology, Federico II University of Naples, Naples, Italy
| | - Bianca Covelli
- Department of Translational Medical Sciences, Federico II University of Naples, Naples, Italy
| | - Antonia Fiore
- Centre for Advanced Biotechnology (CEINGE), Naples, Italy
| | - Roberta Lanzillo
- Multiple Sclerosis Unit, Federico II University Hospital, Via Sergio Pansini 5, 80131, Naples, Italy
- Department of Neuroscience, Reproductive Sciences and Odontostomatology, Federico II University of Naples, Naples, Italy
| | - Antonio Carotenuto
- Multiple Sclerosis Unit, Federico II University Hospital, Via Sergio Pansini 5, 80131, Naples, Italy
- Department of Neuroscience, Reproductive Sciences and Odontostomatology, Federico II University of Naples, Naples, Italy
| | - Maria Petracca
- Multiple Sclerosis Unit, Federico II University Hospital, Via Sergio Pansini 5, 80131, Naples, Italy
- Department of Human Neuroscience, Sapienza University of Rome, Rome, Italy
| | - Lucia Stanziola
- Department of Translational Medical Sciences, Federico II University of Naples, Naples, Italy
| | - Giulia Scalia
- Centre for Advanced Biotechnology (CEINGE), Naples, Italy
| | - Vincenzo Brescia Morra
- Multiple Sclerosis Unit, Federico II University Hospital, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Marcello Moccia
- Multiple Sclerosis Unit, Federico II University Hospital, Via Sergio Pansini 5, 80131, Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Via Sergio Pansini 5, 80131, Naples, Italy.
| |
Collapse
|
10
|
Raftopoulou S, Rapti A, Karathanasis D, Evangelopoulos ME, Mavragani CP. The role of type I IFN in autoimmune and autoinflammatory diseases with CNS involvement. Front Neurol 2022; 13:1026449. [PMID: 36438941 PMCID: PMC9685560 DOI: 10.3389/fneur.2022.1026449] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/17/2022] [Indexed: 07/30/2023] Open
Abstract
Type I interferons (IFNs) are major mediators of innate immunity, with well-known antiviral, antiproliferative, and immunomodulatory properties. A growing body of evidence suggests the involvement of type I IFNs in the pathogenesis of central nervous system (CNS) manifestations in the setting of chronic autoimmune and autoinflammatory disorders, while IFN-β has been for years, a well-established therapeutic modality for multiple sclerosis (MS). In the present review, we summarize the current evidence on the mechanisms of type I IFN production by CNS cellular populations as well as its local effects on the CNS. Additionally, the beneficial effects of IFN-β in the pathophysiology of MS are discussed, along with the contributory role of type I IFNs in the pathogenesis of neuropsychiatric lupus erythematosus and type I interferonopathies.
Collapse
Affiliation(s)
- Sylvia Raftopoulou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Rapti
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Karathanasis
- First Department of Neurology, National and Kapodistrian University of Athens, Aeginition Hospital, Athens, Greece
| | | | - Clio P. Mavragani
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
11
|
Monahan RC, van den Beukel MD, Borggreven NV, Fronczek R, Huizinga TWJ, Kloppenburg M, Steup-Beekman GM, Trouw LA. Autoantibodies against specific post-translationally modified proteins are present in patients with lupus and associate with major neuropsychiatric manifestations. RMD Open 2022; 8:rmdopen-2021-002079. [PMID: 35450955 PMCID: PMC9024229 DOI: 10.1136/rmdopen-2021-002079] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/27/2022] [Indexed: 11/08/2022] Open
Abstract
Background Although autoantibodies are an important hallmark of systemic lupus erythematosus (SLE), most are not specific for SLE or any of its clinical manifestations. Autoantibodies against post-translationally modified (PTM) proteins have been studied extensively in rheumatoid arthritis and associate with disease progression. While PTMs have also been detected in patients with SLE, studies on anti-PTM antibodies remain scarce. We studied the presence of anti-PTM antibodies in SLE and neuropsychiatric SLE (NPSLE), a manifestation that lacks serological markers. Methods IgG antibody responses against six PTMs (malondialdehyde–acetaldehyde adducts (MAA), advanced glycation end-products (AGE), carbamylation (CarP), citrullination, acetylation and nitration) were tested using ELISA in sera of 349 patients with SLE (mean age 44±13 years; 87% female) and compared with 108 healthy controls. Levels and positivity were correlated with clinical features and SLE manifestations. Results Anti-MAA, anti-AGE and anti-CarP antibodies were more prevalent in SLE compared with controls (MAA: 29% vs 3%, AGE: 18% vs 4%, CarP: 14% vs 5%, all p≤0.0001). Anti-MAA and anti-AGE antibodies correlated with clinical manifestations and serological inflammatory markers. Patients with major NPSLE showed higher positivity of anti-MAA (39% vs 24%, p=0.01) and anti-CarP antibodies (20% vs 11%, p=0.04) than patients without major NPSLE. In addition, anti-PTM antibody levels correlated with brain volumes, an objective measure of nervous system involvement. Conclusions In our NPSLE cohort, a subset of patients with SLE have anti-PTM antibodies against MAA, AGE and CarP modified proteins. Interestingly, anti-MAA and anti-CarP were more prevalent in NPSLE, a manifestation for which no biomarkers exist.
Collapse
Affiliation(s)
- Rory C Monahan
- Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Rolf Fronczek
- Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom W J Huizinga
- Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Margreet Kloppenburg
- Rheumatology, Leiden University Medical Center, Leiden, The Netherlands.,Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gerda M Steup-Beekman
- Rheumatology, Leiden University Medical Center, Leiden, The Netherlands.,Rheumatology, Haaglanden Medical Center, The Hague, The Netherlands
| | - Leendert A Trouw
- Immunology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
12
|
Vanarsa K, Sasidharan P, Duran V, Gokaraju S, Nidhi M, Louis Sam Titus ASC, Soomro S, Stock AD, Der E, Putterman C, Greenberg B, Mok CC, Hanly JG, Mohan C. Aptamer-based screen of Neuropsychiatric Lupus cerebrospinal fluid reveals potential biomarkers that overlap with the choroid plexus transcriptome. Arthritis Rheumatol 2022; 74:1223-1234. [PMID: 35099126 DOI: 10.1002/art.42080] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 12/28/2021] [Accepted: 01/27/2022] [Indexed: 11/07/2022]
Abstract
OBJECTIVES As no gold-standard diagnostic test exists for neuropsychiatric systemic lupus erythematosus (NPSLE), we executed a broad screen of NPSLE cerebrospinal fluid (CSF) using an aptamer-based platform. METHODS CSF were obtained from NPSLE patients and subjected to proteomic assay using the aptamer-based screen. Potential biomarkers were identified and validated in independent NPSLE cohorts in comparison with other neurological diseases. RESULTS 40 proteins out of 1129 screened were elevated in NPSLE CSF. By ELISA validation, CSF Angiostatin, α2-Macroglobulin, DAN, Fibronectin, HCC-1, IgM, Lipocalin 2, M-CSF and SERPING1 were significantly elevated in a predominantly Caucasian NPSLE cohort (n=24), compared to patients with other neurological diseases (n=54), with CSF IgM (AUC=0.95) and M-CSF (AUC=0.91) being the most discriminatory. In a second, Hong Kong NPSLE cohort, CSF IgM (AUC=0.78) and Lipocalin-2 (AUC=0.85) were the most discriminatory. Several CSF proteins exhibited high diagnostic specificity for NPSLE in both cohorts. Elevated CSF C3 was associated with acute confusional state. Eleven molecules elevated in NPSLE CSF exhibited concordant elevation in the choroid plexus, suggesting shared origins. CONCLUSIONS CSF Lipocalin-2, M-CSF, IgM and complement C3 emerge as promising CSF biomarkers of NPSLE with diagnostic potential.
Collapse
Affiliation(s)
- Kamala Vanarsa
- Department Biomedical Engineering, University of Houston, Houston, TX
| | | | - Valeria Duran
- Department Biomedical Engineering, University of Houston, Houston, TX
| | - Sirisha Gokaraju
- Department Biomedical Engineering, University of Houston, Houston, TX
| | - Malavika Nidhi
- Department Biomedical Engineering, University of Houston, Houston, TX
| | | | - Sanam Soomro
- Department Biomedical Engineering, University of Houston, Houston, TX
| | | | - Evan Der
- Albert Einstein College of Medicine, Bronx, NY
| | | | | | | | - John G Hanly
- Division of Rheumatology, Queen Elizabeth II Health Sciences Center and Dalhousie University Halifax, Nova Scotia, Canada
| | - Chandra Mohan
- Department Biomedical Engineering, University of Houston, Houston, TX
| |
Collapse
|
13
|
Kodera T, Tsutsumi T, Oka Y, Takeda T, Shirota Y, Kameoka J. Clinical efficacy of belimumab in patients with systemic lupus erythematosus and headache. A report of two cases. Mod Rheumatol Case Rep 2022; 6:25-28. [PMID: 34505625 DOI: 10.1093/mrcr/rxab003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/16/2021] [Accepted: 05/14/2021] [Indexed: 06/13/2023]
Abstract
Intractable headache, one of the manifestations of neuropsychiatric systemic lupus erythematosus (SLE), is difficult to diagnose and decide on an appropriate treatment. In addition to conventional therapy based on the type of headache, the treatment should be conducted considering the disease activity of SLE rather than the headache. We report two patients with intractable headache who were successfully treated using belimumab therapy. The headaches in both patients were relieved after 2 weeks of belimumab administration. The neutralisation of B lymphocyte stimulator and reduced production of cytokines from B lymphocytes might contribute to the early effects. The potential benefits of using belimumab as an additional immunosuppressant for treating intractable headache complicated with SLE have been discussed.
Collapse
Affiliation(s)
- Takao Kodera
- Division of Hematology and Rheumatology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Tomomi Tsutsumi
- Division of Hematology and Rheumatology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yumiko Oka
- Division of Hematology and Rheumatology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Tomoki Takeda
- Division of Hematology and Rheumatology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yuko Shirota
- Division of Hematology and Rheumatology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Junichi Kameoka
- Division of Hematology and Rheumatology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| |
Collapse
|
14
|
Aryal R, Patabendige A. Blood-brain barrier disruption in atrial fibrillation: a potential contributor to the increased risk of dementia and worsening of stroke outcomes? Open Biol 2021; 11:200396. [PMID: 33878948 PMCID: PMC8059575 DOI: 10.1098/rsob.200396] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Atrial fibrillation (AF) has become one of the most significant health problems worldwide, warranting urgent answers to currently pending questions on the effects of AF on brain function. Recent evidence has emerged to show an association between AF and an increased risk of developing dementia and worsening of stroke outcomes. A healthy brain is protected by the blood–brain barrier (BBB), which is formed by the endothelial cells that line cerebral capillaries. These endothelial cells are continuously exposed to shear stress (the frictional force generated by blood flow), which affects endothelial cell structure and function. Flow disturbances as experienced during AF can disrupt the BBB and leave the brain vulnerable to damage. Investigating the plausible mechanisms in detail, linking AF to cerebrovascular damage is difficult in humans, leading to paucity of available clinical data. Here, we discuss the available evidence for BBB disruption during AF due to altered cerebral blood flow, and how this may contribute to an increased risk of dementia and worsening of stroke outcomes.
Collapse
Affiliation(s)
- Ritambhara Aryal
- Brain Barriers Group, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia.,Brain and Mental Health Research Programme, Hunter Medical Research Institute, Newcastle, Australia
| | - Adjanie Patabendige
- Brain Barriers Group, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia.,Brain and Mental Health Research Programme, Hunter Medical Research Institute, Newcastle, Australia.,Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| |
Collapse
|
15
|
Petitdemange A, Blaess J, Sibilia J, Felten R, Arnaud L. Shared development of targeted therapies among autoimmune and inflammatory diseases: a systematic repurposing analysis. Ther Adv Musculoskelet Dis 2021; 12:1759720X20969261. [PMID: 33403021 PMCID: PMC7747103 DOI: 10.1177/1759720x20969261] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 10/05/2020] [Indexed: 12/20/2022] Open
Abstract
Background: Pathogenic inflammatory pathways are largely shared between different autoimmune and inflammatory diseases (AIDs). This offers the potential to develop a given targeted therapy in several AIDs. Methods: We analyzed two clinical trials registries (ClinicalTrials.gov and EU Clinical Trials Register) to identify the targeted therapies whose development is shared between at least two of the most common AIDs [rheumatoid arthritis (RA), spondyloarthritis (SpA), cutaneous psoriasis (cPso), inflammatory bowel diseases (IBD), systemic lupus erythematosus (SLE), primary Sjögren’s syndrome (pSS), systemic sclerosis (SSc), idiopathic inflammatory myopathies (IIM), giant cell arteritis (GCA), and multiple sclerosis (MS)] using an in-depth repurposing analysis. Results: We identified 142 shared targeted therapies. The four diseases in which shared targeted therapies were the most numerous were RA (n = 92), cPso (n = 67), IBD (n = 58), and SLE (n = 56). The two clusters of diseases between which the overlap of targeted therapies was the most important were RA and SLE as well as RA, SpA, cPso, and IBD. The targeted therapies which were shared by five diseases or more were abatacept, ustekinumab, rituximab, anakinra, etanercept, infliximab, secukinumab, tofacitinib, alemtuzumab, tocilizumab, adalimumab, apremilast, baricitinib, belimumab, brodalumab, filgotinib, and upadacitinib. The most frequently targeted molecules and pathways were (by descending frequency): JAK-STAT pathways, Th17 axis, TNF-α, IL-6, costimulation molecules, BAFF, CD20, BTK, chemokines and integrins, IL-1, and type I interferon. Conclusion: Many targeted therapies are developed in several AIDs, reflecting the overlap of pathogenic pathways and potential of drug repurposing. This suggests that a revision of the current, clinically based classification of AIDs towards a more mechanistic-based taxonomy might be relevant.
Collapse
Affiliation(s)
- Arthur Petitdemange
- Department of Rheumatology, National Reference Center for Auto-Immune Diseases (RESO), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Julien Blaess
- Department of Rheumatology, National Reference Center for Auto-Immune Diseases (RESO), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Jean Sibilia
- Department of Rheumatology, National Reference Center for Auto-Immune Diseases (RESO), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Renaud Felten
- Department of Rheumatology, National Reference Center for Auto-Immune Diseases (RESO), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Laurent Arnaud
- Service de rhumatologie, Centre National de Référence des Maladies Autoimmunes et Systémiques Rares, Hôpital de Hautepierre, 1 Avenue Molière BP 83049, Strasbourg Cedex, 67098, France
| |
Collapse
|
16
|
Zhang J, Krishnan A, Wu H, Venkataraman V. Calcium-Dependent Translocation of S100B Is Facilitated by Neurocalcin Delta. Molecules 2021; 26:molecules26010227. [PMID: 33466232 PMCID: PMC7794955 DOI: 10.3390/molecules26010227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 12/26/2020] [Accepted: 12/28/2020] [Indexed: 11/29/2022] Open
Abstract
S100B is a calcium-binding protein that governs calcium-mediated responses in a variety of cells—especially neuronal and glial cells. It is also extensively investigated as a potential biomarker for several disease conditions, especially neurodegenerative ones. In order to establish S100B as a viable pharmaceutical target, it is critical to understand its mechanistic role in signaling pathways and its interacting partners. In this report, we provide evidence to support a calcium-regulated interaction between S100B and the neuronal calcium sensor protein, neurocalcin delta both in vitro and in living cells. Membrane overlay assays were used to test the interaction between purified proteins in vitro and bimolecular fluorescence complementation assays, for interactions in living cells. Added calcium is essential for interaction in vitro; however, in living cells, calcium elevation causes translocation of the NCALD-S100B complex to the membrane-rich, perinuclear trans-Golgi network in COS7 cells, suggesting that the response is independent of specialized structures/molecules found in neuronal/glial cells. Similar results are also observed with hippocalcin, a closely related paralog; however, the interaction appears less robust in vitro. The N-terminal region of NCALD and HPCA appear to be critical for interaction with S100B based on in vitro experiments. The possible physiological significance of this interaction is discussed.
Collapse
Affiliation(s)
- Jingyi Zhang
- Department of Cell Biology and Neuroscience, Graduate School of Biomedical Sciences, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA; (J.Z.); (A.K.); (H.W.)
| | - Anuradha Krishnan
- Department of Cell Biology and Neuroscience, Graduate School of Biomedical Sciences, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA; (J.Z.); (A.K.); (H.W.)
| | - Hao Wu
- Department of Cell Biology and Neuroscience, Graduate School of Biomedical Sciences, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA; (J.Z.); (A.K.); (H.W.)
| | - Venkat Venkataraman
- Department of Cell Biology and Neuroscience, Graduate School of Biomedical Sciences, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA; (J.Z.); (A.K.); (H.W.)
- Department of Rehabilitation Medicine, NeuroMusculoskeletal Institute, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA
- Correspondence: ; Tel.: +1-856-566-6418
| |
Collapse
|
17
|
Papachristos DA, Oon S, Hanly JG, Nikpour M. Management of inflammatory neurologic and psychiatric manifestations of systemic lupus erythematosus: A systematic review. Semin Arthritis Rheum 2020; 51:49-71. [PMID: 33360230 DOI: 10.1016/j.semarthrit.2020.12.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/06/2020] [Accepted: 12/11/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND The neurological and psychiatric manifestations of systemic lupus erythematosus (NPSLE) are a heterogeneous group of conditions with variable clinical presentation and significant morbidity and mortality. OBJECTIVES Our aim was to comprehensively assess and present the evidence for treatments used in the management of inflammatory NPSLE. METHODS Medline, Embase, CINHAL and Cochrane CENTRAL were searched from 1990 to end of March 2019 using key words that related to NPSLE and treatment. Included studies comprised clinical trials, observational studies or case series with ≥5 patients and sufficient data related to treatment and outcome in NPSLE patients. RESULTS There were 7222 studies identified in the search, of which 90 were included in the review. There was a notable paucity of clinical trials, with only two randomised controlled trials and one pilot study. Treatment categories included corticosteroids (14 studies), cyclophosphamide (18 studies), synthetic DMARDs (7 studies), biologic therapies (14 studies), therapeutic plasma exchange (6 studies), intravenous immunoglobulin (2 studies), autologous stem cell transplant (3 studies), other therapies (8 studies), combination therapies (6 studies), studies with grouped outcome data (5 studies) and observational studies with therapy-specific associations (7 studies). Corticosteroids are accepted as first line treatment in NPSLE and there is low-moderate evidence supporting their benefit. Moderate evidence, based on consistent data in numerous studies and some trial data, supports the use of cyclophosphamide in the treatment of NPSLE. Limited data support some synthetic DMARDs such as mycophenolate, azathioprine and intrathecal methotrexate. In refractory disease, low-moderate evidence supports rituximab therapy and limited evidence supports benefit following autologous stem cell transplant. Regarding adjuvant treatments, limited evidence favours addition of plasma exchange, intravenous immunoglobulin and hydroxychloroquine. There exists very limited data for other therapies. CONCLUSION There are multiple therapeutic options for the management of inflammatory NPSLE including systemic, biologic and interventional therapies; however, currently there is a paucity of high-quality trial data to guide firm recommendations. In order to better understand the optimal treatment of NPSLE and its different subtypes, further well-designed clinical trials are needed.
Collapse
Affiliation(s)
- D A Papachristos
- Department of Rheumatology, St. Vincent's Hospital, Melbourne, Australia
| | - S Oon
- Department of Rheumatology, St. Vincent's Hospital, Melbourne, Australia; Department of Medicine, The University of Melbourne, Australia; Department of Rheumatology, The Royal Melbourne Hospital, Australia
| | - J G Hanly
- Division of Rheumatology, Department of Medicine and Department of Pathology, Queen Elizabeth II Health Sciences Center and Dalhousie University, Halifax, NS, Canada
| | - M Nikpour
- Department of Rheumatology, St. Vincent's Hospital, Melbourne, Australia; Department of Medicine, The University of Melbourne, Australia.
| |
Collapse
|
18
|
Karnopp TE, Chapacais GF, Freitas EC, Monticielo OA. Lupus animal models and neuropsychiatric implications. Clin Rheumatol 2020; 40:2535-2545. [PMID: 33155159 DOI: 10.1007/s10067-020-05493-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/29/2020] [Accepted: 11/01/2020] [Indexed: 02/06/2023]
Abstract
Systemic lupus erythematosus (SLE) that involves neurological complications is known as neuropsychiatric systemic lupus erythematosus (NPSLE). Research in humans is difficult due to the disease's great heterogeneity. Animal models are a resource for new discoveries. In this review, we examine experimental models of lupus that present neuropsychiatric manifestations. Spontaneous animal models such as NZB/W F1 and MRL/lpr are commonly used in NPSLE research; these models present few SLE symptoms compared to induced animal models, such as pristane-induced lupus (PIL). The PIL model is known to present eight of the main clinical and laboratory manifestations of SLE described by the American College of Rheumatology. Many cytokines associated with NPSLE are expressed in the PIL model, such as IL-6, TNF-α, and IFN. However, to date, NPSLE manifestations have been poorly studied in the PIL model. In this review article, we discuss whether the PIL model can mimic neuropsychiatric manifestations of SLE. Key Points • PIL model have a strong interferon signature. • Animals with PIL express learning and memory deficit.
Collapse
Affiliation(s)
- Thaís Evelyn Karnopp
- Laboratório de Doenças Autoimunes, Divisão de Reumatologia, Centro de Pesquisas Experimentais, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, sala 12109, Porto Alegre, 90035-003, Brazil. .,Programa de Pós-Graduação em Medicina: Ciências Médicas, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil. .,Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | - Gustavo Flores Chapacais
- Laboratório de Doenças Autoimunes, Divisão de Reumatologia, Centro de Pesquisas Experimentais, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, sala 12109, Porto Alegre, 90035-003, Brazil.,Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Curso de Graduação em Biomedicina, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Eduarda Correa Freitas
- Laboratório de Doenças Autoimunes, Divisão de Reumatologia, Centro de Pesquisas Experimentais, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, sala 12109, Porto Alegre, 90035-003, Brazil.,Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Odirlei André Monticielo
- Laboratório de Doenças Autoimunes, Divisão de Reumatologia, Centro de Pesquisas Experimentais, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, sala 12109, Porto Alegre, 90035-003, Brazil.,Programa de Pós-Graduação em Medicina: Ciências Médicas, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
19
|
Abstract
PROPOSE OF REVIEW Neuropsychiatric systemic lupus erythematosus (NPSLE) is an emerging frontier in lupus care encompassing a wide spectrum of clinical manifestations. Its pathogenesis remains poorly understood because of the complexity of pathophysiologic mechanisms involved and limited access to tissue. We highlight recent advances in the pathophysiology of neuropsychiatric lupus. RECENT FINDINGS Disruption of blood-brain barrier (BBB) facilitating entrance of neurotoxic antibodies into the central nervous system (CNS), neuroinflammation and cerebral ischemia are the key mechanisms. Disruption of the BBB may occur not only at the traditional BBB, but also at the blood-cerebrospinal fluid barrier. Certain autoantibodies, such as anti-N-methyl-D-aspartate receptors, antiribosomal P and antiphospholipid antibodies may cause injury in subsets of patients with diffuse neuropsychiatric disease. Activation of microglia via autoantibodies, interferon-a or other immune reactants, may amplify the inflammatory response and promote neuronal damage. New inflammatory pathways, such as TWEAK/Fn14, Bruton's tyrosine kinase, Nogo-a and ACE may represent additional potential targets of therapy. Novel neuroimaging techniques suggest alterations in brain perfusion and metabolism, increased concentration of neurometabolites, indicative of glial activation, vasculopathy and neuronal impairment. SUMMARY NPSLE encompasses a diverse phenotype with distinct pathogenic mechanisms, which could be targeted by novel therapies or repositioning of existing drugs.
Collapse
|
20
|
Chen C, Geng L, Xu X, Kong W, Hou Y, Yao G, Feng X, Zhang H, Liang J. Comparative proteomics analysis of plasma protein in patients with neuropsychiatric systemic lupus erythematosus. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:579. [PMID: 32566606 PMCID: PMC7290550 DOI: 10.21037/atm.2020.04.58] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background The aim of this study was to evaluate serum biomarkers of systemic lupus erythematous (SLE) patients, with and without neuropsychiatric (NP) manifestation by high-resolution proteomic analysis. Methods SLE patients with NP (NPSLE, n=9), SLE patients without NP (non-NPSLE, n=9) and healthy controls (HC, n=9) were prospectively enrolled in this study, and their plasma samples were collected and pooled into 3 NPSLE, 3 non-NPSLE and 3 HC samples for discovery profile. The TMT-LC-MS/MS-based proteomics approach was used to identify the differential proteome among the three matched groups, and the data were analyzed by bioinformatics tools, including Gene Ontology (GO) categories, Kyoto Encyclopedia of Genes and Genomes enrichment analysis, to explore canonical pathways and networks involved in the pathogenesis of NPSLE. To validation of differentially expressed proteomics results, four proteins were measured by ELISA. Results There were altogether 223 differentially expressed proteins in NPSLE groups compared with healthy controls (HC), of which 96 proteins increased while 127 proteins decreased. Compared with non-NPSLE, there were only 49 differentially expressed proteins in NPSLE groups, of which 37 proteins increased while 12 proteins decreased. The significantly changed pathway that those proteins are involved in was complement and coagulation cascades in NPSLE group compared with health controls. However, we didn't find significantly changed pathway between NPSLE group and non-NPSLE group. Five proteins were found significantly changed in all group-comparisons with consistent tendencies using Venn analysis, including Vitamin D binding protein (VDBP), C-reactive protein (CRP), KRT16, IGHV4-4 and CTRP3. Four proteins including CTRP3, VDBP, PAPPA and TRYP2 were selected to estimate the validity of the proteomics approach by ELISA. The expression levels of CTRP3 and TRYP2 were significantly changed in NPSLE patients compared with either HC or non-NPSLE patients. Conclusions Our research has successfully established serum protein profiles of NPSLE and non-NPSLE patients through TMT technology and screened out five proteins significantly changed in group-comparisons with consistent tendencies. The pathway of complement and coagulation cascades may participate in pathogenesis of NPSLE and non-NPSLE.
Collapse
Affiliation(s)
- Chen Chen
- Department of Clinical Nutrition, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Linyu Geng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Xue Xu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Wei Kong
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yayi Hou
- Institute of Brain Sciences, Medical School, Nanjing University, Nanjing 210093, China
| | - Genhong Yao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Xuebing Feng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Huayong Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jun Liang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| |
Collapse
|
21
|
Zhang Y, Liu Q, Yu Y, Wang M, Wen C, He Z. Early and Short-Term Interventions in the Gut Microbiota Affects Lupus Severity, Progression, and Treatment in MRL/lpr Mice. Front Microbiol 2020; 11:628. [PMID: 32346376 PMCID: PMC7171286 DOI: 10.3389/fmicb.2020.00628] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
There have been attempts to reveal the possible associations between systemic lupus erythematosus (SLE) and gut microbiota. Using MRL/lpr mice, this study was performed to reveal whether early and short-term interventions in gut microbiota affect lupus. MRL/lpr mice were treated with antibiotics or fecal microbiota transplantation (FMT) before onset. Then, prednisone was used to treat the lupus mice with initially different gut microbiota compositions. The compositions of gut microbiota were assessed by the V3-V4 region of 16S rRNA gene sequence. Early and short-term antibiotics exposure aggravated lupus severity by depleting beneficial gut microbiota for lupus, such as Lactobacillus and Bifidobacterium, and enriching harmful gut microbiota for lupus, such as Klebsiella and Proteus. FMT alleviated lupus severity by renovating the antibiotic-induced dysbiosis of gut microbiota in the following 1 week after antibiotics exposure. Besides, short-term antibiotics exposure before onset imposed no significant effects on lupus progression, but the following one week of FMT suppressed lupus progression. Moreover, the short-term antibiotics or FMT before onset inhibited the therapeutic efficiency of prednisone on lupus from 9 to 13 weeks old of MRL/lpr mice. These data demonstrate that the gut microbiota before onset is important for lupus severity, progression and treatment.
Collapse
Affiliation(s)
- Yun Zhang
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiuping Liu
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiran Yu
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mingzhu Wang
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chengping Wen
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhixing He
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
22
|
Ichikawa T, Shimojima Y, Kishida D, Kaneko T, Sekijima Y. Primary central nervous system lymphoma in neuropsychiatric systemic lupus erythematosus: case-based review. Rheumatol Int 2020; 41:1009-1017. [PMID: 32253501 DOI: 10.1007/s00296-020-04569-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 03/24/2020] [Indexed: 11/26/2022]
Abstract
Primary central nervous system lymphoma (PCNSL) sometimes occurs in immune-compromised hosts or patients with autoimmune diseases. Some cohort studies have previously reported an increased risk of non-Hodgkin's lymphoma in systemic lupus erythematosus (SLE), while some cases of PCNSL in patients with SLE were reported. We present the case of PCNSL which developed in a patient with the active phase of neuropsychiatric SLE (NPSLE). Furthermore, we reviewed published English articles to confirm the characteristics of PCNSL related to SLE. To our knowledge, this is the first report of PCNSL occurring in NPSLE. Histology demonstrated B-cell lymphoma with a positive Epstein-Barr virus-encoded RNA. This patient recovered following surgical resection of the lymphoma, whole brain radiation therapy, intravenous infusion of rituximab (RTX), and administration of belimumab after RTX. Given the series of reviews, our report suggests that the persistence of damage in the central nervous system (CNS) and long-term exposure to immunosuppressants may impact oncogenic immune responses within the CNS, leading to PCNSL development.
Collapse
MESH Headings
- Adult
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antineoplastic Agents, Immunological/administration & dosage
- Brain/diagnostic imaging
- Brain/pathology
- Female
- Herpesvirus 4, Human/isolation & purification
- Humans
- Immunosuppression Therapy/adverse effects
- Immunosuppressive Agents/administration & dosage
- Lupus Vasculitis, Central Nervous System/complications
- Lupus Vasculitis, Central Nervous System/drug therapy
- Lymphoma, Large B-Cell, Diffuse/complications
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/virology
- Rituximab/administration & dosage
Collapse
Affiliation(s)
- Takanori Ichikawa
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Yasuhiro Shimojima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan.
| | - Dai Kishida
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Tomoki Kaneko
- Department of Radiology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Yoshiki Sekijima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| |
Collapse
|
23
|
Leptin: an unappreciated key player in SLE. Clin Rheumatol 2019; 39:305-317. [PMID: 31707542 DOI: 10.1007/s10067-019-04831-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/18/2019] [Accepted: 10/23/2019] [Indexed: 02/08/2023]
Abstract
Leptin is the forerunner of the adipokine superfamily and plays a key role in regulating energy expenditure and neuroendocrine function. Researches into leptin put emphasize not only on the metabolic role but also its immunoregulatory effect on immune response through immunocyte activation and cytokine secretion. Leptin acts on receptors that are widespread throughout the body and that are expressed across many tissue types. As a consequence, the abnormal expression of leptin has been found to correlate with a number of diseases, including cancers, autoimmune diseases, and cardiovascular diseases. The significance of leptin in the development of autoimmune diseases is becoming increasingly prominent. Systemic lupus erythematosus (SLE) is a severe atypical autoimmune disease that causes damage to multiple organ systems. It is characterised by the following: impaired clearance of apoptotic cells, loss of tolerance to self-antigens, aberrant activation of T cells and B cells, and chronic inflammation. The heightened immunocyte response in SLE means that these physiological systems are particularly vulnerable to regulation by leptin in addition to being of great significance to the research field. Our current review provides insight into the regulatory roles that leptin plays on immune effector cells in SLE.
Collapse
|
24
|
van Mierlo HC, Broen JCA, Kahn RS, de Witte LD. B-cells and schizophrenia: A promising link or a finding lost in translation? Brain Behav Immun 2019; 81:52-62. [PMID: 31271869 DOI: 10.1016/j.bbi.2019.06.043] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/07/2019] [Accepted: 06/29/2019] [Indexed: 12/11/2022] Open
Abstract
Recent genetic studies have suggested a potential role for B-cells in the pathogenesis of schizophrenia. Greater insight in the functioning of B-cells in patients with schizophrenia is therefore of importance. In this narrative review we aim to give an overview of the current literature on B-cells and schizophrenia. We found no evidence for altered numbers of these cells in blood. We did find support for increased levels of B-cell related cytokines and certain autoantibodies. Studies on B-cell development and function, or their numbers in cerebrospinal fluid or brain tissue are very limited. Based on the available data we appraise whether various B-cell mediated pathological mechanisms are likely to play a role in schizophrenia and provide directions for future research.
Collapse
Affiliation(s)
- Hans C van Mierlo
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Jasper C A Broen
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - René S Kahn
- Department of Psychiatry, Icahn School of Medicine, New York, United States; Mental Illness Research, Education and Clinical Center (MIRECC), James J Peters VA Medical Center, Bronx, NY, United States
| | - Lot D de Witte
- Department of Psychiatry, Icahn School of Medicine, New York, United States; Mental Illness Research, Education and Clinical Center (MIRECC), James J Peters VA Medical Center, Bronx, NY, United States
| |
Collapse
|
25
|
Lushington GH, Barnes AC. Protein Glycation: An Old Villain is Shedding Secrets. Comb Chem High Throughput Screen 2019; 22:362-369. [DOI: 10.2174/1386207322666190704094356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/02/2019] [Accepted: 06/10/2019] [Indexed: 01/16/2023]
Abstract
:
The glycation of proteins is non-physiological post-translational incorporation of
carbohydrates onto the free amines or guanidines of proteins and some lipids. Although the
existence of glycated proteins has been known for forty years, a full understanding of their
pathogenic nature has been slow in accruing. In recent years, however, glycation has gained widespread
acceptance as a contributing factor in numerous metabolic, autoimmune, and neurological
disorders, tying together several confounding aspects of disease etiology. From diabetes, arthritis,
and lupus, to multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer’s, and Parkinson’s
diseases, an emerging glycation/inflammation paradigm now offers significant new insight into a
physiologically important toxicological phenomenon. It exposes novel drug targets and treatment
options, and may even lay foundations for long-awaited breakthroughs.
:
This ‘current frontier’ article briefly profiles current knowledge regarding the underlying causes
of glycation, the structural biology implications of such modifications, and their pathological
consequences. Although several emerging therapeutic strategies for addressing glycation
pathologies are introduced, the primary purpose of this mini-review is to raise awareness of the
challenges and opportunities inherent in this emerging new medicinal target area.
Collapse
|
26
|
Kang D, Mok CC. Management of Psychosis in Neuropsychiatric Lupus. JOURNAL OF CLINICAL RHEUMATOLOGY AND IMMUNOLOGY 2019. [DOI: 10.1142/s2661341719300015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Manifestations of neuropsychiatric systemic lupus erythematosus (NPSLE) are heterogeneous. Acute psychosis is an uncommon but well-recognized manifestation of NPSLE. With no specific biomarkers to date, the diagnosis of NPSLE relies on clinical acumen for circumstantial evidence and exclusion of important differential diagnoses. The attribution of psychosis to NPSLE is facilitated by the application attribution models. In particular, the American College of Rheumatology nomenclature, Systemic Lupus International Collaborating Clinics attribution models and Italian algorithm for the attribution of psychosis to NPSLE are revisited. The mainstay of treatment for psychosis attributable to NPSLE is immunosuppression and symptomatic control. In refractory cases, immunomodulatory and emerging biological agents may be considered. This article reviews the diagnostic dilemma, pathogenic mechanisms and treatment of psychosis in SLE patients.
Collapse
Affiliation(s)
- Di Kang
- Department of Medicine, Tuen Mun Hospital, Hong Kong SAR, China
| | - Chi Chiu Mok
- Department of Medicine, Tuen Mun Hospital, Hong Kong SAR, China
| |
Collapse
|
27
|
Abstract
Systemic lupus erythematosus (SLE) is a representative systemic autoimmune disease that has various types of manifestations in multiple organs. Additionally, SLE is one of the most variable diseases in its epidemiology and etiology with heterogenous types of immune dysfunction. Since the word 'lupus' has first appeared in the literature in the Middle Ages, clinical/pathological knowledges have massively accumulated that contributed to the establishments and improvements of classification criteria, therapeutic agents or assessments of disease activity. Along with them, the survival rate of patients with SLE has dramatically improved. However, the mortality rate is still higher compared with the healthy population and the progress in basic, translational and clinical research are expected to lead to new insights into pathogenesis and identifying novel targets for therapy.
Collapse
Affiliation(s)
- Kenji Oku
- a Department of Rheumatology, Endocrinology and Nephrology, Graduate School of Medicine and Faculty of Medicine , Hokkaido University , Sapporo , Japan
| | - Tatsuya Atsumi
- a Department of Rheumatology, Endocrinology and Nephrology, Graduate School of Medicine and Faculty of Medicine , Hokkaido University , Sapporo , Japan
| |
Collapse
|
28
|
Putterman C, Pisetsky DS, Petri M, Caricchio R, Wu AHB, Sanz I, Oates JC, Wallace S, Sorek R, Gerwien R, Safer P, Jakobi-Brook K, Cohen IR. The SLE-key test serological signature: new insights into the course of lupus. Rheumatology (Oxford) 2018; 57:1632-1640. [PMID: 29873771 PMCID: PMC6105919 DOI: 10.1093/rheumatology/key149] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 04/24/2018] [Indexed: 01/13/2023] Open
Abstract
Objective We previously described the multiplex autoantibody SLE-key Rule-Out test, which detects a signature of autoantibody reactivity that distinguishes healthy subjects from SLE patients with 94% sensitivity, 75% specificity and 93% negative predictive value; thus, an individual manifesting a positive Rule-Out test score is unlikely to have SLE (e.g. lupus is excluded). The objective of this current study was to evaluate the stability of the lupus-associated signature over time. Methods We used banked serum samples from healthy subjects (n = 51) and lupus patients (n = 50 individual samples and n = 181 paired samples, for a total of n = 412 serum samples). The samples were drawn at different times after diagnosis to analyse the impact on the SLE-key Rule-Out test of time elapsed since diagnosis and any changes in disease activity (as reflected by the SLEDAI score). Results The SLE signature remains stable for the first 10 years after diagnosis; in this time frame, <10% of patients manifested a positive Rule-Out score and the SLE-key Rule-Out score was independent of the underlying disease activity as reflected by the SLEDAI score. After ⩾10 years, ∼30% of lupus subjects scored as SLE Ruled-Out; the proportion of patients manifesting this status was even greater in the subset of individuals with a SLEDAI score of 0. Conclusion These findings raise the possibility that a significant number of SLE patients manifest a change in their serological signature over time, and that such a signature change may signify an evolution in the immunological features of their disease relevant to patient management.
Collapse
Affiliation(s)
- Chaim Putterman
- The Division of Rheumatology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY
| | - David S Pisetsky
- Durham VA Medical Center, Duke University Medical Center and Medical Research Service, Durham, NC
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Alan H B Wu
- Department of Laboratory Medicine, San Francisco General Hospital, San Francisco, CA
| | - Ignacio Sanz
- Division of Rheumatology, Emory University School of Medicine, Atlanta, GA
| | - Jim C Oates
- Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC
| | | | | | | | | | | | - Irun R Cohen
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
29
|
Nalakonda G, Islam M, Chukwu VE, Soliman A, Munim R, Abukraa I. Psycho-rheumatic Integration in Systemic Lupus Erythematosus: An Insight into Antibodies Causing Neuropsychiatric Changes. Cureus 2018; 10:e3091. [PMID: 30324045 PMCID: PMC6171782 DOI: 10.7759/cureus.3091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The main purpose of this paper is to bring together all the antibodies and markers related to neurological and psychiatric manifestations in systemic lupus erythematosus and also the pharmacology that could help treat these symptoms. Existing research data regarding specific antibodies involved in the disease process and drugs that were being studied was collected and analyzed. After reviewing the studies published by various authors, symptoms were shown to be mainly caused by antibodies against N-methyl-D-aspartate receptor (NMDAR) antibodies, anti-endothelial, anti-ribosomal P, antiphospholipid antibodies, cytokines like interferons and chemokines. The monoclonal antibody rituximab has shown to be beneficial in some of the cases. Based on all the articles reviewed, the antibodies and cytokines showed the most effective evidence in causing the different manifestations of neuropsychiatric systemic lupus erythematosus (NPSLE), but studies regarding the drugs being effective against all the symptoms are inconclusive as there are very few studies. Further research to support the drug’s effectiveness in managing the symptoms is needed. More studies are needed regarding early diagnosis of NPSLE using the antibodies as biomarkers as it could help in preventing these manifestations.
Collapse
Affiliation(s)
- Gouthami Nalakonda
- Medical Student, Chalmeda Anandrao Institute of Medical Sciences, Karimnagar, IND
| | - Mimsa Islam
- Internal Medicine, Sir Salimullah Medical College, Dhaka, USA
| | | | | | - Rujina Munim
- Miscellaneous, Sylhet Mag Osmani Medical College and Hospital, Sylhet, BGD
| | - Inas Abukraa
- Faculty of Medicine, Tripoli University, Tripoli, LBY
| |
Collapse
|
30
|
Tartar DM, Chung L, Fiorentino DF. Clinical significance of autoantibodies in dermatomyositis and systemic sclerosis. Clin Dermatol 2018; 36:508-524. [DOI: 10.1016/j.clindermatol.2018.04.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
31
|
Abstract
The number of peer-reviewed articles published during the 2016 solar year and retrieved using the "autoimmunity" key word remained stable while gaining a minimal edge among the immunology articles. Nonetheless, the quality of the publications has been rising significantly and, importantly, acquisitions have become available through scientific journals dedicated to immunology or autoimmunity. Major discoveries have been made in the fields of systemic lupus erythematosus, rheumatoid arthritis, autoimmunity of the central nervous system, vasculitis, and seronegative spondyloarthrithritides. Selected examples include the role of IL17-related genes and long noncoding RNAs in systemic lupus erythematosus or the effects of anti-pentraxin 3 (PTX3) in the treatment of this paradigmatic autoimmune condition. In the case of rheumatoid arthritis, there have been reports of the role of induced regulatory T cells (iTregs) or fibrocytes and T cell interactions with exciting implications. The large number of studies dealing with neuroimmunology pointed to Th17 cells, CD56(bright) NK cells, and low-level TLR2 ligands as involved in multiple sclerosis, along with a high salt intake or the micriobiome-derived Lipid 654. Lastly, we focused on the rare vasculitides to which numerous studies were devoted and suggested that unsuspected cell populations, including monocytes, mucosal-associated invariant T cells, and innate lymphoid cells, may be crucial to ANCA-associated manifestations. This brief and arbitrary discussion of the findings published in 2016 is representative of a promising background for developments that will enormously impact the work of laboratory scientists and physicians at an exponential rate.
Collapse
Affiliation(s)
- Carlo Selmi
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital, via A. Manzoni 56, 20089, Rozzano, Milan, Italy.
- Department of Medical Biotechnologies and Translational Medicine (BIOMETRA), University of Milan, Milan, Italy.
| |
Collapse
|
32
|
Chalmers SA, Wen J, Doerner J, Stock A, Cuda CM, Makinde HM, Perlman H, Bosanac T, Webb D, Nabozny G, Fine JS, Klein E, Ramanujam M, Putterman C. Highly selective inhibition of Bruton's tyrosine kinase attenuates skin and brain disease in murine lupus. Arthritis Res Ther 2018; 20:10. [PMID: 29370834 PMCID: PMC5785891 DOI: 10.1186/s13075-017-1500-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 12/20/2017] [Indexed: 05/30/2023] Open
Abstract
Background Systemic lupus erythematosus (SLE) is a systemic autoimmune disease that affects different end organs, including skin and brain. We and others have previously shown the importance of macrophages in the pathogenesis of cutaneous and neuropsychiatric lupus. Additionally, autoantibodies produced by autoreactive B cells are thought to play a role in both the skin and central nervous system pathologies associated with SLE. Methods We used a novel inhibitor of Bruton’s tyrosine kinase (BTK), BI-BTK-1, to target both macrophage and B cell function in the MRL-lpr/lpr murine model of SLE, and examined the effect of treatment on skin and brain disease. Results We found that treatment with BI-BTK-1 significantly attenuated the lupus associated cutaneous and neuropsychiatric disease phenotypes in MRL/lpr mice. Specifically, BI-BTK-1 treated mice had fewer macroscopic and microscopic skin lesions, reduced cutaneous cellular infiltration, and diminished inflammatory cytokine expression compared to control mice. BTK inhibition also significantly improved cognitive function, and decreased accumulation of T cells, B cells, and macrophages within the central nervous system, specifically the choroid plexus. Conclusions Directed therapies may improve the response rate in lupus-driven target organ involvement, and decrease the dangerous side effects associated with global immunosuppression. Overall, our results suggest that inhibition of BTK may be a promising therapeutic option for cutaneous and neuropsychiatric disease associated with SLE. Electronic supplementary material The online version of this article (doi:10.1186/s13075-017-1500-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Samantha A Chalmers
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jing Wen
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jessica Doerner
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ariel Stock
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Carla M Cuda
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hadijat M Makinde
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Harris Perlman
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Todd Bosanac
- Small Molecule Discovery Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | - Deborah Webb
- Immunology and Respiratory Disease Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | - Gerald Nabozny
- Immunology and Respiratory Disease Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | - Jay S Fine
- Immunology and Respiratory Disease Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | - Elliott Klein
- Immunology and Respiratory Disease Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | - Meera Ramanujam
- Immunology and Respiratory Disease Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | - Chaim Putterman
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA. .,Division of Rheumatology, Albert Einstein College of Medicine, F701N, 1300 Morris Park Ave, Bronx, NY, 10461, USA.
| |
Collapse
|
33
|
Muller S, Brun S, René F, de Sèze J, Loeffler JP, Jeltsch-David H. Autophagy in neuroinflammatory diseases. Autoimmun Rev 2017; 16:856-874. [DOI: 10.1016/j.autrev.2017.05.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 05/20/2017] [Indexed: 12/12/2022]
|
34
|
Platt MP, Agalliu D, Cutforth T. Hello from the Other Side: How Autoantibodies Circumvent the Blood-Brain Barrier in Autoimmune Encephalitis. Front Immunol 2017; 8:442. [PMID: 28484451 PMCID: PMC5399040 DOI: 10.3389/fimmu.2017.00442] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/30/2017] [Indexed: 12/11/2022] Open
Abstract
Antibodies against neuronal receptors and synaptic proteins are associated with autoimmune encephalitides (AE) that produce movement and psychiatric disorders. In order to exert their pathological effects on neural circuits, autoantibodies against central nervous system (CNS) targets must gain access to the brain and spinal cord by crossing the blood–brain barrier (BBB), a tightly regulated gateway formed by endothelial cells lining CNS blood vessels. To date, the pathogenic mechanisms that underlie autoantibody-triggered encephalitic syndromes are poorly understood, and how autoantibodies breach the barrier remains obscure for almost all AE syndromes. The relative importance of cellular versus humoral immune mechanisms for disease pathogenesis also remains largely unexplored. Here, we review the proposed triggers for various autoimmune encephalopathies and their animal models, as well as basic structural features of the BBB and how they differ among various CNS regions, a feature that likely underlies some regional aspects of autoimmune encephalitis pathogenesis. We then discuss the routes that antibodies and immune cells employ to enter the CNS and their implications for AE. Finally, we explore future therapeutic strategies that may either preserve or restore barrier function and thereby limit immune cell and autoantibody infiltration into the CNS. Recent mechanistic insights into CNS autoantibody entry indicate promising future directions for therapeutic intervention beyond current, short-lived therapies that eliminate circulating autoantibodies.
Collapse
Affiliation(s)
- Maryann P Platt
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Dritan Agalliu
- Department of Neurology, Columbia University Medical Center, New York, NY, USA.,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.,Department of Pharmacology, Columbia University Medical Center, New York, NY, USA.,Columbia Translational Neuroscience Initiative, Columbia University Medical Center, New York, NY, USA
| | - Tyler Cutforth
- Department of Neurology, Columbia University Medical Center, New York, NY, USA.,Columbia Translational Neuroscience Initiative, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
35
|
International and multidisciplinary expert recommendations for the use of biologics in systemic lupus erythematosus. Autoimmun Rev 2017; 16:650-657. [PMID: 28434948 DOI: 10.1016/j.autrev.2017.04.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 03/26/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND/PURPOSE Despite conventional immunosuppressants, active and steroid-dependent systemic lupus erythematosus (SLE) represents a therapeutic challenge. Only one biologic, belimumab, has been approved, but other biologics are sometimes used off-label. Given the lack of evidence-based data in some clinical situations encountered in real life, we developed expert recommendations for the use of biologics for SLE. METHODS The recommendations were developed by a formal consensus method. This method aims to formalize the degree of agreement among experts by identifying, through iterative ratings with feedback, the points on which experts agree, disagree or are undecided. Hence, the recommendations are based on the agreed-upon points. We gathered the opinion of 59 French-speaking SLE experts from 3 clinical networks dedicated to systemic autoimmune diseases (FLEUR, IMIDIATE, FAI2R) from Algeria, Belgium, France, Italy, Morocco, Switzerland and Tunisia. Represented medical specialities were internal medicine (49%), rheumatology (34%), nephrology (7%), dermatology (5%), pediatrics (3%) and cardiology (2%). Two methodologists and 3 strictly independent SLE expert groups contributed to developing these recommendations: a steering group (SG) (n=9), an evaluation group (EG) (n=28) and a reading group (RG) (n=22). Preliminary recommendations were drafted by the SG, then proposed to the EG. Each EG member rated the degree of agreement from 1 to 9 (1: lowest; 9: strongest) for each recommendation. After 2 rating rounds, the SG submitted a new version of the recommendations to the RG. With comments from the RG, the SG finalised the recommendations. RESULTS A total of 17 final recommendations were formulated by the SG, considering all agreement scores and comments by the EG and RG members and the two methodologists. These recommendations define the subset of patients who require a biologic; the type of biologics to use (belimumab, rituximab, etc.) depending on the organ involvement and associated co-treatments; what information should be given to patients; and how to evaluate treatment efficacy and when to consider discontinuation. CONCLUSION Overall, 17 recommendations for the good use of biologics in SLE were formulated by a large panel of SLE experts to provide guidance for clinicians in daily practice. These recommendations will be regularly updated according to the results of new randomized trials and increasing real life experience.
Collapse
|