1
|
Moritz CP, Tholance Y, Boutahar N, Borowczyk C, Berger AE, Paul S, Antoine JC, Camdessanché JP. The antibody repertoire of autoimmune sensory neuronopathies targets pathways of the innate and adaptive immune system. An autoantigenomic approach. J Transl Autoimmun 2025; 10:100277. [PMID: 40026393 PMCID: PMC11870273 DOI: 10.1016/j.jtauto.2025.100277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/09/2025] [Accepted: 01/29/2025] [Indexed: 03/05/2025] Open
Abstract
Sensory neuronopathies (SNN) encompass diverse etiologies, with autoimmunity playing a major role through both cellular and humoral responses. To investigate the humoral autoantibody repertoire in autoimmune SNN, we conducted a retrospective cohort study using large Human Proteome-wide protein microarrays (HuProt 3.1, HuProt 4.0, ProtoArrays). We specifically analyzed immune system pathways targeted within the autoantigen repertoire (the autoantigenome). We included 131 participants: 44 patients with non-paraneoplastic autoimmune SNN (12 with anti-FGFR3 and/or anti-AGO antibodies), 8 with paraneoplastic SNN, and 79 controls. Findings were validated in an independent cohort of 16 SNN patients. Overrepresentation of immune-system-related proteins was assessed using the Reactome database, and serum levels of IFN-γ and IL-6 were measured with the Bio-Plex Pro™ Reagent Kit. Autoimmune SNN sera interact with significantly more immune system proteins than healthy controls (ProtoArrays: 271/863 vs. 14/863, HuProt: 112/1694 vs. 39/1694, both p < 0.0001). Overrepresentation was observed across all major immune sub-pathways, including innate and adaptive immune responses as well as cytokine signaling. Anti-FGFR3-positive SNN patients showed more frequent reactivity to immune system proteins than anti-FGFR3-negative ones. The independent SNN cohort validated the overrepresentation of targeted immune system pathways. Validation with dot blot and ELISA confirmed reactivity to TRIM21 and IL-6 and identified anti-IFN-γ-positive SNN patients. IFN-γ levels correlated weakly with levels of anti-IFN-γ antibodies (Pearson's r = 0.22, p = 0.03). We conclude that the antibody repertoire of autoimmune SNN targets pathways of the innate and adaptive immune system, potentially reflecting key disease-related immune pathways and highlighting the systemic role of immune dysregulation in SNN.
Collapse
Affiliation(s)
- Christian P. Moritz
- Synaptopathies and Autoantibodies (SynatAc) Team, Institut NeuroMyoGène, MELIS, INSERM U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1, 43 Boulevard Du 11 Novembre 1918, Villeurbanne, 69622, France
- University Jean Monnet, 10, Rue de Marandière, 42270, Saint-Étienne, France
- Department of Neurology, University Hospital of Saint-Etienne, Avenue Albert Raimond, 42270, Saint-Etienne, France
| | - Yannick Tholance
- Synaptopathies and Autoantibodies (SynatAc) Team, Institut NeuroMyoGène, MELIS, INSERM U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1, 43 Boulevard Du 11 Novembre 1918, Villeurbanne, 69622, France
- University Jean Monnet, 10, Rue de Marandière, 42270, Saint-Étienne, France
- Department of Biochemistry, University Hospital of Saint-Etienne, Avenue Albert Raimond, 42270, Saint-Etienne, France
| | - Nadia Boutahar
- Synaptopathies and Autoantibodies (SynatAc) Team, Institut NeuroMyoGène, MELIS, INSERM U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1, 43 Boulevard Du 11 Novembre 1918, Villeurbanne, 69622, France
- University Jean Monnet, 10, Rue de Marandière, 42270, Saint-Étienne, France
- Department of Biochemistry, University Hospital of Saint-Etienne, Avenue Albert Raimond, 42270, Saint-Etienne, France
| | - Coralie Borowczyk
- Synaptopathies and Autoantibodies (SynatAc) Team, Institut NeuroMyoGène, MELIS, INSERM U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1, 43 Boulevard Du 11 Novembre 1918, Villeurbanne, 69622, France
- Department of Biochemistry, University Hospital of Saint-Etienne, Avenue Albert Raimond, 42270, Saint-Etienne, France
| | - Anne-Emmanuelle Berger
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, Saint-Etienne, F42023, France
- Department of Immunology and Biotherapies, University Hospital of Saint-Etienne, Avenue Albert Raimond, 42270, Saint-Étienne, France
| | - Stéphane Paul
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, Saint-Etienne, F42023, France
- Department of Immunology and Biotherapies, University Hospital of Saint-Etienne, Avenue Albert Raimond, 42270, Saint-Étienne, France
| | - Jean-Christophe Antoine
- Synaptopathies and Autoantibodies (SynatAc) Team, Institut NeuroMyoGène, MELIS, INSERM U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1, 43 Boulevard Du 11 Novembre 1918, Villeurbanne, 69622, France
- University Jean Monnet, 10, Rue de Marandière, 42270, Saint-Étienne, France
- Department of Neurology, University Hospital of Saint-Etienne, Avenue Albert Raimond, 42270, Saint-Etienne, France
| | - Jean-Philippe Camdessanché
- Synaptopathies and Autoantibodies (SynatAc) Team, Institut NeuroMyoGène, MELIS, INSERM U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1, 43 Boulevard Du 11 Novembre 1918, Villeurbanne, 69622, France
- University Jean Monnet, 10, Rue de Marandière, 42270, Saint-Étienne, France
- Department of Neurology, University Hospital of Saint-Etienne, Avenue Albert Raimond, 42270, Saint-Etienne, France
| |
Collapse
|
2
|
Chamessian A, Tavares-Ferriera D, Payne M, Govindarajan R, Pestronk A, Bertels Z, Widman AJ, Slivicki RA, Del Rosario JS, Yi J, Copits B, Ornitz DM, Price TJ, Gereau RW. Expression of Fibroblast Growth Factor Receptor 3 (FGFR3) in the Human Peripheral Nervous System: Implications for the Putative Pathogenic Role of FGFR3 Autoantibodies in Neuropathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.03.639509. [PMID: 40093173 PMCID: PMC11908139 DOI: 10.1101/2025.03.03.639509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Introduction Autoantibodies to the Fibroblast Growth Factor Receptor 3 (FGFR3-AAbs) have been associated with idiopathic sensory-predominant neuropathy. The pathogenicity of FGFR3-AAbs in this disorder is unknown. Pathogenic mechanisms of autoantibodies in other dysimmune neuropathies commonly involve their direct binding to antigens on either neurons or glia. The expression of FGFR3 in the human peripheral nervous system is unknown. Therefore, as an initial step toward clarifying the pathogenicity of FGFR3-AAbs, we characterized the expression of FGFR3 in nerve (hNerve), dorsal root ganglia (hDRG) and spinal cord (hSC) in human. Methods FGFR3 mRNA was assayed via in situ hybridization (ISH) on post-mortem sections of hNerve, hDRG and hSC, and by re-analysis of RNA-sequencing data from hDRG. FGFR3 protein was assayed in these tissues using capillary electrophoretic immunoassays (CEIA) with several validated anti-FGFR3 antibodies. Results FGFR3 mRNA was not detected in hNerve or hDRG but was abundant in hSC by ISH. FGFR3 protein was absent from hNerve and hDRG by CEIA but was moderately expressed in hSC. Discussion A direct pathogenic mechanism of FGFR3-AAbs in sensory neuropathy would require the expression of FGFR3 in either neurons or non-neuronal cells in nerve or DRG. Using multiple methods, we did not detect FGFR3 expression at the mRNA or protein levels in these tissues. Given the absence of FGFR3 from hNerve and hDRG, it is improbable that FGFR3-AAbs cause direct damage to the neural components involved in neuropathy and thus are unlikely to be pathogenic, although indirect mechanisms via non-neural cells cannot be excluded.
Collapse
Affiliation(s)
- Alexander Chamessian
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Diana Tavares-Ferriera
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, US
| | - Maria Payne
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Raghav Govindarajan
- Hospital Sisters Health System, Division of Neurology, Edwardsville, IL 62025
| | - Alan Pestronk
- Washington University Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Zach Bertels
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Allie J Widman
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Richard A Slivicki
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - John S Del Rosario
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Jiwon Yi
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Bryan Copits
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, US
| | - Robert W Gereau
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| |
Collapse
|
3
|
Yeganeh O, Abdolahinia ED, Soofiyani SR, Faghfuri E, Shafie A, Pahlavan Y. Biosensors for autoimmune diseases. Clin Chim Acta 2025; 565:119998. [PMID: 39454986 DOI: 10.1016/j.cca.2024.119998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024]
Abstract
Diagnosis of autoimmune diseases (ADs) is usually based on symptoms and laboratory tests that measure the occurrence of serological and genetic biomarkers such as peptides, autoantibodies, and complement proteins. Early detection of AD is essential to reduce the severity of symptoms and organ damage as a result of progressive disease. Biosensors are tools that convert biochemical signals produced by molecular elements into optical, electrical, and other physical signals for analysis. In recent years, peptides, antigens, aptamers, autoantibodies, and other biomolecules have provided suitable diagnostic features for development of biosensors in detecting and follow up the diagnoses and treatment of diseases. This study reviews the introducing of different biomarkers in ADs with the novel vision to use of biosensor technology for research and development in this regard. Therefore, this study has the required innovation for using biosensor technology with more attention to electrochemical based biosensors to developing, targeting and designing the easy applicable and available diagnostic and response to treatment products using key biomolecules for ADs. It will help readers to understand the research trends of biosensors in ADs and further advance the development of this paramount field.
Collapse
Affiliation(s)
- Omid Yeganeh
- Department of Microbiology, Faculty of Bioscience, North Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Elaheh Dalir Abdolahinia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Saeideh Razi Soofiyani
- Clinical Research Development Unit, Sina Educational, Research and Treatment Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Elnaz Faghfuri
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Abbas Shafie
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran; Biosensor Sciences and Technologies Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Yasamin Pahlavan
- Biosensor Sciences and Technologies Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
4
|
Antoine JC. Antibodies in immune-mediated peripheral neuropathies. Where are we in 2024? Rev Neurol (Paris) 2024; 180:876-887. [PMID: 39322491 DOI: 10.1016/j.neurol.2024.09.002] [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: 06/14/2024] [Revised: 08/28/2024] [Accepted: 09/09/2024] [Indexed: 09/27/2024]
Abstract
Over the past 30 years, about 20 antibodies have been identified in immune-mediated neuropathies, recognizing membrane or intracellular proteins or glycolipids of neuron and Schwann cells. This article reviews the different methods used for their detection, what we know about their pathogenic role, how they have helped identify several disorders, and how they are essential for diagnosis. Despite sustained efforts, some immune-mediated disorders still lack identified autoantibodies, notably the classical form of Guillain-Barré syndrome and chronic inflammatory demyelinating polyneuropathy. The reasons for this are discussed. The article also tries to determine potential future developments in antibody research, particularly the use of omic approaches and the search for other types of biomarkers beyond diagnostic ones, such as those that can identify patients who will respond to a given treatment.
Collapse
Affiliation(s)
- J-C Antoine
- Service de neurologie, CHU de Saint-Étienne, 42055 Saint-Étienne cedex, France; Inserm CNRS, laboratoire SynAtac, MeliS, université Jean-Monnet, Saint-Étienne, France.
| |
Collapse
|
5
|
Chang CW, Wang HY, Lin WY, Wang YC, Lo WL, Lin TW, Yu JR, Tseng YJ. Identifying heterogeneous subgroups of systemic autoimmune diseases by applying a joint dimension reduction and clustering approach to immunomarkers. BioData Min 2024; 17:36. [PMID: 39285420 PMCID: PMC11403832 DOI: 10.1186/s13040-024-00389-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/02/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND The high complexity of systemic autoimmune diseases (SADs) has hindered precise management. This study aims to investigate heterogeneity in SADs. METHODS We applied a joint cluster analysis, which jointed multiple correspondence analysis and k-means, to immunomarkers and measured the heterogeneity of clusters by examining differences in immunomarkers and clinical manifestations. The electronic health records of patients who received an antinuclear antibody test and were diagnosed with SADs, namely systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and Sjögren's syndrome (SS), were retrieved between 2001 and 2016 from hospitals in Taiwan. RESULTS With distinctive patterns of immunomarkers, a total of 11,923 patients with the three SADs were grouped into six clusters. None of the clusters was composed only of a single SAD, and these clusters demonstrated considerable differences in clinical manifestation. Both patients with SLE and SS had a more dispersed distribution in the six clusters. Among patients with SLE, the occurrence of renal compromise was higher in Clusters 3 and 6 (52% and 51%) than in the other clusters (p < 0.001). Cluster 3 also had a high proportion of patients with discoid lupus (60%) than did Cluster 6 (39%; p < 0.001). Patients with SS in Cluster 3 were the most distinctive because of the high occurrence of immunity disorders (63%) and other and unspecified benign neoplasm (58%) with statistical significance compared with the other clusters (all p < 0.05). CONCLUSIONS The immunomarker-driven clustering method could recognise more clinically relevant subgroups of the SADs and would provide a more precise diagnosis basis.
Collapse
Affiliation(s)
- Chia-Wei Chang
- Department of Computer Science, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Hsin-Yao Wang
- Department of Laboratory Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Wan-Ying Lin
- Department of Laboratory Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Syu Kang Sport Clinic, Taipei, Taiwan
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Yu-Chiang Wang
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
- Queen's Heart Institute, Queens Medical Center, Honolulu, HI, USA
| | - Wei-Lin Lo
- Department of Rheumatology, Chang Gung Memorial Hospital at Keelung, Keelung, Taiwan
| | - Ting-Wei Lin
- Department of Laboratory Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Jia-Ruei Yu
- Department of Laboratory Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Yi-Ju Tseng
- Department of Computer Science, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
6
|
Li V, Binder MD, Purcell AW, Kilpatrick TJ. Antigen-specific immunotherapy via delivery of tolerogenic dendritic cells for multiple sclerosis. J Neuroimmunol 2024; 390:578347. [PMID: 38663308 DOI: 10.1016/j.jneuroim.2024.578347] [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: 02/12/2024] [Revised: 03/22/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system resulting from loss of immune tolerance. Many disease-modifying therapies for MS have broad immunosuppressive effects on peripheral immune cells, but this can increase risks of infection and attenuate vaccine-elicited immunity. A more targeted approach is to re-establish immune tolerance in an autoantigen-specific manner. This review discusses methods to achieve this, focusing on tolerogenic dendritic cells. Clinical trials in other autoimmune diseases also provide learnings with regards to clinical translation of this approach, including identification of autoantigen(s), selection of appropriate patients and administration route and frequency.
Collapse
Affiliation(s)
- Vivien Li
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia; Department of Neurology, The Royal Melbourne Hospital, Melbourne, Australia.
| | - Michele D Binder
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia
| | - Anthony W Purcell
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Trevor J Kilpatrick
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia; Department of Neurology, The Royal Melbourne Hospital, Melbourne, Australia
| |
Collapse
|
7
|
Shurin MR, Wheeler SE. Clinical Significance of Uncommon, Non-Clinical, and Novel Autoantibodies. Immunotargets Ther 2024; 13:215-234. [PMID: 38686351 PMCID: PMC11057673 DOI: 10.2147/itt.s450184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 04/17/2024] [Indexed: 05/02/2024] Open
Abstract
Autoantibodies are a common mark of autoimmune reaction and their identification in the patients' serum, cerebrospinal fluid, or tissues is generally believed to represent diagnostic or prognostic biomarkers of autoimmune diseases or autoinflammatory conditions. Traditionally, autoantibody testing is an important part of the clinical examination of suspected patients, and in the absence of reliable T cell tests, characterization of autoantibody responses might be suitable in finding causes of specific autoimmune responses, their strength, and sometimes commencement of autoimmune disease. Autoantibodies are also useful for prognostic stratification in clinically diverse groups of patients if checked repeatedly. Antibody discoveries are continuing, with important consequences for verifying autoimmune mechanisms, diagnostic feasibility, and clinical management. Adding newly identified autoantibody-autoantigen pairs to common clinical laboratory panels should help upgrade and harmonize the identification of systemic autoimmune rheumatic disorders and other autoimmune conditions. Herein, we aim to summarize our current knowledge of uncommon and novel autoantibodies in the context of discussing their validation, diagnostic practicability, and clinical relevance. The regular updates within the field are important and well justified.
Collapse
Affiliation(s)
- Michael R Shurin
- Division of Clinical Immunopathology, Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Sarah E Wheeler
- Division of Clinical Immunopathology, Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
8
|
McBride DA, Jones RM, Bottini N, Shah NJ. The therapeutic potential of immunoengineering for systemic autoimmunity. Nat Rev Rheumatol 2024:10.1038/s41584-024-01084-x. [PMID: 38383732 DOI: 10.1038/s41584-024-01084-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2024] [Indexed: 02/23/2024]
Abstract
Disease-modifying drugs have transformed the treatment options for many systemic autoimmune diseases. However, an evolving understanding of disease mechanisms, which might vary between individuals, is paving the way for the development of novel agents that operate in a patient-tailored manner through immunophenotypic regulation of disease-relevant cells and the microenvironment of affected tissue domains. Immunoengineering is a field that is focused on the application of engineering principles to the modulation of the immune system, and it could enable future personalized and immunoregulatory therapies for rheumatic diseases. An important aspect of immunoengineering is the harnessing of material chemistries to design technologies that span immunologically relevant length scales, to enhance or suppress immune responses by re-balancing effector and regulatory mechanisms in innate or adaptive immunity and rescue abnormalities underlying pathogenic inflammation. These materials are endowed with physicochemical properties that enable features such as localization in immune cells and organs, sustained delivery of immunoregulatory agents, and mimicry of key functions of lymphoid tissue. Immunoengineering applications already exist for disease management, and there is potential for this new discipline to improve disease modification in rheumatology.
Collapse
Affiliation(s)
- David A McBride
- Department of NanoEngineering and Chemical Engineering Program, University of California, San Diego, La Jolla, CA, USA
| | - Ryan M Jones
- Department of NanoEngineering and Chemical Engineering Program, University of California, San Diego, La Jolla, CA, USA
| | - Nunzio Bottini
- Kao Autoimmunity Institute and Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Nisarg J Shah
- Department of NanoEngineering and Chemical Engineering Program, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
9
|
Nasonov EL. [Autoimmunity in rheumatology: A review]. TERAPEVT ARKH 2023; 95:1056-1063. [PMID: 38158939 DOI: 10.26442/00403660.2023.12.202501] [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: 12/26/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
Autoimmunity and autoinflammation, co-potentiating pathological processes, are considered within the "immune-inflammatory" continuum (continuity with a variety of elements), reflecting the close relationship between the innate and acquired immune responses. Autoimmunity is the leading pathogenetic mechanism for a specific type of human chronic inflammatory disorders - autoimmune diseases, affecting more than 10% of people in the general population. Advances in molecular biology, pharmacogenetics, and bioinformatics provided the background for individualizing therapy for systemic autoimmune rheumatic diseases within personalized medicine. Studying the immunopathogenesis mechanisms, improving diagnostics, interpreting the molecular taxonomy, and developing approaches to the prevention and personalized therapy of systemic autoimmune rheumatic diseases are the priority issues of modern medicine.
Collapse
Affiliation(s)
- E L Nasonov
- Nasonova Research Institute of Rheumatology
- Sechenov First Moscow State Medical University (Sechenov University)
| |
Collapse
|
10
|
Wang D, Yang D, Yang L, Diao L, Zhang Y, Li Y, Wang H, Ren J, Cheng L, Tan Q, Zhang R, Han X, Zhang X, Wang B, Li D, Chen M, Hermjakob H, Li Y, LaBaer J, Zhou Z, Yu X. Human Autoantigen Atlas: Searching for the Hallmarks of Autoantigens. J Proteome Res 2023. [PMID: 37183442 DOI: 10.1021/acs.jproteome.2c00799] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Understanding autoimmunity to endogenous proteins is crucial in diagnosing and treating autoimmune diseases. In this work, we developed a user-friendly AAgAtlas portal (http://biokb.ncpsb.org.cn/aagatlas_portal/index.php#), which can be used to search for 8045 non-redundant autoantigens (AAgs) and 47 post-translationally modified AAgs against 1073 human diseases that are prioritized by a credential score developed by multisource evidence. Using AAgAtlas, the immunogenic properties of human AAgs was systematically elucidated according to their genetic, biophysical, cytological, expression profile, and evolutionary characteristics. The results indicated that human AAgs are evolutionally conserved in protein sequence and enriched in three hydrophilic and polar amino acid residues (K, D, and E) that are located at the protein surface. AAgs are enriched in proteins that are involved in nucleic acid binding, transferase, and the cytoskeleton. Genome, transcriptome, and proteome analyses further indicated that AAb production is associated with gene variance and abnormal protein expression related to the pathological activities of different tumors. Collectively, our data outlines the hallmarks of human AAgs that facilitate the understanding of humoral autoimmunity and the identification of biomarkers of human diseases.
Collapse
Affiliation(s)
- Dan Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Dong Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Liuhui Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Lihong Diao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yuqi Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yang Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Hongye Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Jing Ren
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Linlin Cheng
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Qiaoyun Tan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ran Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xiaohong Han
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Xiaohan Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
- College of Medicine and Integrated Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Bingwei Wang
- College of Medicine and Integrated Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Dong Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Meng Chen
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Henning Hermjakob
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Yongzhe Li
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Joshua LaBaer
- The Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona 85287, United States
| | - Zhou Zhou
- Department of Laboratory Medicine, National Center for Cardiovascular Diseases and Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100037, China
| | - Xiaobo Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| |
Collapse
|
11
|
Ren J, Wang H, Wei C, Yang X, Yu X. Development of a protein microarray for profiling circulating autoantibodies in human diseases. Proteomics Clin Appl 2022; 16:e2100132. [PMID: 36006834 DOI: 10.1002/prca.202100132] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 12/30/2022]
Abstract
PURPOSE To develop a robust microarray platform to detect thousands of serological autoantibodies (AAbs) simultaneously in different diseases. EXPERIMENTAL DESIGN An AAbMap microarray was prepared by printing a total of 4032 purified His-tagged human proteins and peptide probes on a chemically-modified slide. The sensitivity, dynamic range, and the inter- and intra-array reproducibility of the AAb microarray were then systematically tested and optimized. Finally, the large-scale profiling of AAbs in the serum of patients with different human diseases using the AAbMap microarray was demonstrated. RESULTS The dynamic range of antibody (Ab) detection was 2 to 3 orders of magnitude with the lowest limit of detection (LOD) of 68 pg/mL. The intra-array (r) correlation of duplicate spots was 1.00, whereas the inter-array correlations between different arrays and batches were 0.99 and 0.97 to 0.98, respectively. Notably, 132, 266, 171, and 84 AAbs were detected in pooled serum from healthy controls (HCs) or patients with rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), or lung cancer (LC), respectively. These AAbs included antibodies that target well-known disease biomarkers, such as anti-cyclic citrullinated peptide, anti-ribonucleoprotein, and anti-nucleosome. CONCLUSIONS AND CLINICAL RELEVANCE We developed a microarray platform to measure thousands of serological AAbs simultaneously with high sensitivity and reproducibility. The array can help study autoimmunity and complement genomics, proteomics, and metabolomics data for systematic investigations of human diseases.
Collapse
Affiliation(s)
- Jing Ren
- School of Basic Medicine Sciences, Anhui Medical University, Hefei, Anhui, PR China
| | - Hongye Wang
- National Center for Protein Sciences Beijing (PHOENIX Center), State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, PR China
| | - Chundi Wei
- National Center for Protein Sciences Beijing (PHOENIX Center), State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, PR China
| | - Xiaoming Yang
- School of Basic Medicine Sciences, Anhui Medical University, Hefei, Anhui, PR China.,National Center for Protein Sciences Beijing (PHOENIX Center), State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, PR China
| | - Xiaobo Yu
- National Center for Protein Sciences Beijing (PHOENIX Center), State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, PR China
| |
Collapse
|
12
|
Shome M, Chung Y, Chavan R, Park JG, Qiu J, LaBaer J. Serum autoantibodyome reveals that healthy individuals share common autoantibodies. Cell Rep 2022; 39:110873. [PMID: 35649350 PMCID: PMC9221390 DOI: 10.1016/j.celrep.2022.110873] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 03/02/2022] [Accepted: 05/04/2022] [Indexed: 12/02/2022] Open
Abstract
Autoantibodies are a hallmark of both autoimmune disease and cancer, but
they also occur in healthy individuals. Here, we perform a meta-analysis of nine
datasets and focus on the common autoantibodies shared by healthy individuals.
We report 77 common autoantibodies based on the protein microarray data obtained
from probing 182 healthy individual sera on 7,653 human proteins and an
additional 90 healthy individual sera on 1,666 human proteins. There is no
gender bias; however, the number of autoantibodies increase with age, plateauing
around adolescence. We use a bioinformatics pipeline to determine possible
molecular-mimicry peptides that can contribute to the elicitation of these
common autoantibodies. There is enrichment of intrinsic properties of proteins
like hydrophilicity, basicity, aromaticity, and flexibility for common
autoantigens. Subcellular localization and tissue-expression analysis reveal
that several common autoantigens are sequestered from the circulating
autoantibodies. Shome et al. performed a meta-analysis to discover the common
autoantibodies found in healthy individuals. These common autoantibodies appear
and increase during youth and plateau at adolescence. Bioinformatics techniques
demonstrate the potential role of molecular mimicry in their production as well
as several common intrinsic biochemical properties.
Collapse
Affiliation(s)
- Mahasish Shome
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Yunro Chung
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA; College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Ramani Chavan
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Jin G Park
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Ji Qiu
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Joshua LaBaer
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
13
|
Cinquanta L, Infantino M, Bizzaro N. Detecting Autoantibodies by Multiparametric Assays: Impact on Prevention, Diagnosis, Monitoring, and Personalized Therapy in Autoimmune Diseases. J Appl Lab Med 2022; 7:137-150. [PMID: 34996071 DOI: 10.1093/jalm/jfab132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/07/2021] [Indexed: 11/13/2022]
Abstract
BACKGROUND The introduction of multiparametric autoantibody tests has been proposed to improve the accuracy of the immunological diagnosis of autoimmune diseases (AID) and to accelerate time for completing the diagnostic process. Multiplex tests are capable of detecting many autoantibodies in a single run whereas a traditional immunoassay uses a single antigen to detect only a single specificity of autoantibodies. The reasons why multiplex tests could replace conventional immunoassays lie in the evidence that they allow for more efficient handling of large numbers of samples by the laboratory, while ensuring greater diagnostic sensitivity in AID screening. CONTENT This review aims to highlight the important role that multiparametric tests could assume when designed for defined profiles they are used not only for diagnostic purposes but also to predict the onset of AID to identify clinical phenotypes and to define prognosis. Furthermore, differences in the antibody profile could identify which subjects will be responsive or not to a specific pharmacological treatment. SUMMARY The use of autoantibody profiles, when specifically requested and performed with clinically validated technologies, can represent a significant step toward personalized medicine in autoimmunology.
Collapse
Affiliation(s)
| | - Maria Infantino
- Laboratorio di Immunologia e Allergologia, Ospedale S. Giovanni di Dio, Firenze, Italy
| | - Nicola Bizzaro
- Laboratorio di Patologia Clinica, Ospedale San Antonio, Tolmezzo, Italy.,Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
| |
Collapse
|
14
|
Immune regulations by 14-3-3: A misty terrain. Immunobiology 2021; 226:152145. [PMID: 34628289 DOI: 10.1016/j.imbio.2021.152145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 09/20/2021] [Accepted: 09/28/2021] [Indexed: 11/22/2022]
Abstract
The 14-3-3 proteins are known for their functions related to the cell cycle and play a prominent role in cancer-related diseases. Recent studies show that 14-3-3 proteins are also regulators of immune responses and are involved in the pathogenesis of autoimmune and infectious diseases. This focused review highlights the significant and recent studies on how 14-3-3 proteins influence innate and adaptive immune responses; specifically, their roles as immunogens and cytokine signaling regulators are discussed. These revelations have added numerous questions to the pre-existing list of challenges, including understanding the 14-3-3 proteins' mechanism of immunogenicity to dissecting the isoform-specific immune regulations.
Collapse
|
15
|
Gao X, Zhao C, Zhang N, Cui X, Ren Y, Su C, Wu S, Yao Z, Yang J. Genetic expression and mutational profile analysis in different pathologic stages of hepatocellular carcinoma patients. BMC Cancer 2021; 21:786. [PMID: 34238242 PMCID: PMC8268469 DOI: 10.1186/s12885-021-08442-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/31/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The clinical pathologic stages (stage I, II, III-IV) of hepatocellular carcinoma (HCC) are closely linked to the clinical prognosis of patients. This study aims at investigating the gene expression and mutational profile in different clinical pathologic stages of HCC. METHODS Based on the TCGA-LIHC cohort, we utilized a series of analytical approaches, such as statistical analysis, random forest, decision tree, principal component analysis (PCA), to identify the differential gene expression and mutational profiles. The expression patterns of several targeting genes were also verified by analyzing the Chinese HLivH060PG02 HCC cohort, several GEO datasets, HPA database, and diethylnitrosamine-induced HCC mouse model. RESULTS We identified a series of targeting genes with copy number variation, which is statistically associated with gene expression. Non-synonymous mutations mainly existed in some genes (e.g.,TTN, TP53, CTNNB1). Nevertheless, no association between gene mutation frequency and pathologic stage distribution was detected. The random forest and decision tree modeling analysis data showed a group of genes related to different HCC pathologic stages, including GAS2L3 and SEMA3F. Additionally, our PCA data indicated several genes associated with different pathologic stages, including SNRPA and SNRPD2. Compared with adjacent normal tissues, we observed a highly expressed level of GAS2L3, SNRPA, and SNRPD2 (P = 0.002) genes in HCC tissues of our HLivH060PG02 cohort. We also detected the high expression pattern of GAS2L3, SEMA3F, SNRPA, and SNRPD2 in the datasets of GSE102079, GSE76427, GSE64041, GSE121248, GSE84005, and the qPCR assay using diethylnitrosamine-induced HCC mouse model. Moreover, SEMA3F and SNRPD2 protein were highly stained in the HCC tissues of the HPA database. The high expression level of these four genes was associated with the poor survival prognosis of HCC cases. CONCLUSIONS Our study provides evidence regarding the gene expression and mutational profile in different clinical pathologic stages of TCGA HCC cases. Identifying four targeting genes, including GAS2L3, SNRPA, SNRPD2, and SEMA3F, offers insight into the molecular mechanisms associated with different prognoses of HCC.
Collapse
Affiliation(s)
- Xingjie Gao
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
- Key Laboratory of Immune Microenvironment and Disease, Ministry of Education, Key Laboratory of Cellular and Molecular Immunology in Tianjin, Excellent Talent Project, Tianjin Medical University, Tianjin, China.
| | - Chunyan Zhao
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Key Laboratory of Immune Microenvironment and Disease, Ministry of Education, Key Laboratory of Cellular and Molecular Immunology in Tianjin, Excellent Talent Project, Tianjin Medical University, Tianjin, China
| | - Nan Zhang
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Key Laboratory of Immune Microenvironment and Disease, Ministry of Education, Key Laboratory of Cellular and Molecular Immunology in Tianjin, Excellent Talent Project, Tianjin Medical University, Tianjin, China
| | - Xiaoteng Cui
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Key Laboratory of Immune Microenvironment and Disease, Ministry of Education, Key Laboratory of Cellular and Molecular Immunology in Tianjin, Excellent Talent Project, Tianjin Medical University, Tianjin, China
- Department of Neurosurgery Ministry of Education and Tianjin Municipal Government Laboratory of Neuro-Oncology Key Laboratory of Neurotrauma, Variation, and Regeneration , Tianjin Neurological Institute Tianjin Medical University General Hospital , Tianjin, China
| | - Yuanyuan Ren
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Key Laboratory of Immune Microenvironment and Disease, Ministry of Education, Key Laboratory of Cellular and Molecular Immunology in Tianjin, Excellent Talent Project, Tianjin Medical University, Tianjin, China
| | - Chao Su
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Key Laboratory of Immune Microenvironment and Disease, Ministry of Education, Key Laboratory of Cellular and Molecular Immunology in Tianjin, Excellent Talent Project, Tianjin Medical University, Tianjin, China
| | - Shaoyuan Wu
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Key Laboratory of Immune Microenvironment and Disease, Ministry of Education, Key Laboratory of Cellular and Molecular Immunology in Tianjin, Excellent Talent Project, Tianjin Medical University, Tianjin, China
| | - Zhi Yao
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Key Laboratory of Immune Microenvironment and Disease, Ministry of Education, Key Laboratory of Cellular and Molecular Immunology in Tianjin, Excellent Talent Project, Tianjin Medical University, Tianjin, China
| | - Jie Yang
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
- Key Laboratory of Immune Microenvironment and Disease, Ministry of Education, Key Laboratory of Cellular and Molecular Immunology in Tianjin, Excellent Talent Project, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
16
|
Calabrese L, Winthrop KL. Rheumatology and COVID-19 at 1 year: facing the unknowns. Ann Rheum Dis 2021; 80:679-681. [PMID: 33658236 PMCID: PMC8142424 DOI: 10.1136/annrheumdis-2021-219957] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 02/20/2021] [Accepted: 02/22/2021] [Indexed: 12/15/2022]
Affiliation(s)
| | - Kevin L Winthrop
- School of Public Health, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
17
|
Nasonov EL. 2019 Coronavirus disease (COVID-19): contribution of rheumatology. TERAPEVT ARKH 2021; 93:71504. [DOI: 10.26442/00403660.2021.05.200799] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 06/11/2021] [Indexed: 12/15/2022]
Abstract
The 2019 coronavirus disease (COVID-19) pandemic become a major challenge for humanity and a unique opportunity to get an idea of the real achievements of modern biology and medicine. In the course of the pandemic, a large number of new fundamental and medical issues have been revealed regarding the relationship between viral infection and many common chronic non-infectious diseases, among which immune-mediated rheumatic diseases (IMRD) occupy an important position. It is now well known that SARS-CoV-2 infection is accompanied by a wide range of extrapulmonary clinical and laboratory disorders, some of which are characteristic of IMRD and other autoimmune and autoinflammatory diseases in humans. The most severe consequence of alterations in regulation of the immunity in COVID-19 and IMRD is the so-called cytokine storm syndrome, which is defined as COVID-19-associated hyperinflammatory syndrome in COVID-19, and as macrophage activation syndrome in IMRD. The COVID-19-associated hyperinflammatory syndrome was used as a reason for drug repurposing and off-label use of a wide range of anti-inflammatory drugs, which have been specially developed for the treatment of IMRD over the past 20 years. Common immunopathological mechanisms and approaches to pharmacotherapy in COVID-19 and IMRD determined the unique place of rheumatology among medical specialties contributing to combat the COVID-19 pandemic. The article provides the basic provisions of the International and National Association of Rheumatologists and the Association of Rheumatologists of Russia (ARR) recommendations for management of patients with IMRD during the COVID-19 pandemic.
Collapse
|
18
|
Antiphospholipid Syndrome Alliance for Clinical Trials and International Networking (APS ACTION): 10-Year Update. Curr Rheumatol Rep 2021; 23:45. [PMID: 33932165 PMCID: PMC8088198 DOI: 10.1007/s11926-021-01008-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2021] [Indexed: 12/14/2022]
Abstract
Purpose of Review APS ACTION is an international research network created to design and conduct large-scale, multicenter research in persistently antiphospholipid antibody (aPL)–positive patients. Given the expanding research activities of the network in the last decade since its creation, the purpose of this article is to review the scientific contributions of APS ACTION as well as future directions. Recent Findings APS ACTION has achieved increased international collaboration with internal and external investigators for outcome, interventional, and mechanistic antiphospholipid syndrome (APS) studies. This has been linked to substantial progress in Core laboratory work, which has demonstrated that laboratories can achieve good agreement in performance of aPL assays by use of the same reagents, analyzer type, and protocols. Summary APS ACTION will continue to identify gaps in the existing aPL/APS literature, design mechanistic studies to elucidate underlying mechanisms, and conduct prospective, large-scale clinical studies, all for the ultimate goal of early diagnosis and improved management of aPL-positive patients.
Collapse
|
19
|
Moorman CD, Sohn SJ, Phee H. Emerging Therapeutics for Immune Tolerance: Tolerogenic Vaccines, T cell Therapy, and IL-2 Therapy. Front Immunol 2021; 12:657768. [PMID: 33854514 PMCID: PMC8039385 DOI: 10.3389/fimmu.2021.657768] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 03/04/2021] [Indexed: 12/14/2022] Open
Abstract
Autoimmune diseases affect roughly 5-10% of the total population, with women affected more than men. The standard treatment for autoimmune or autoinflammatory diseases had long been immunosuppressive agents until the advent of immunomodulatory biologic drugs, which aimed at blocking inflammatory mediators, including proinflammatory cytokines. At the frontier of these biologic drugs are TNF-α blockers. These therapies inhibit the proinflammatory action of TNF-α in common autoimmune diseases such as rheumatoid arthritis, psoriasis, ulcerative colitis, and Crohn's disease. TNF-α blockade quickly became the "standard of care" for these autoimmune diseases due to their effectiveness in controlling disease and decreasing patient's adverse risk profiles compared to broad-spectrum immunosuppressive agents. However, anti-TNF-α therapies have limitations, including known adverse safety risk, loss of therapeutic efficacy due to drug resistance, and lack of efficacy in numerous autoimmune diseases, including multiple sclerosis. The next wave of truly transformative therapeutics should aspire to provide a cure by selectively suppressing pathogenic autoantigen-specific immune responses while leaving the rest of the immune system intact to control infectious diseases and malignancies. In this review, we will focus on three main areas of active research in immune tolerance. First, tolerogenic vaccines aiming at robust, lasting autoantigen-specific immune tolerance. Second, T cell therapies using Tregs (either polyclonal, antigen-specific, or genetically engineered to express chimeric antigen receptors) to establish active dominant immune tolerance or T cells (engineered to express chimeric antigen receptors) to delete pathogenic immune cells. Third, IL-2 therapies aiming at expanding immunosuppressive regulatory T cells in vivo.
Collapse
Affiliation(s)
| | | | - Hyewon Phee
- Department of Inflammation and Oncology, Amgen Research, Amgen Inc., South San Francisco, CA, United States
| |
Collapse
|
20
|
The pre-clinical phase of rheumatoid arthritis: From risk factors to prevention of arthritis. Autoimmun Rev 2021; 20:102797. [PMID: 33746022 DOI: 10.1016/j.autrev.2021.102797] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 01/29/2021] [Indexed: 02/06/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease considered as a multistep process spanning from the interaction of genetic (e.g., shared epitope or non-HLA loci), environmental and behavioral risk factors (e.g., smoking) leading to breaking immune tolerance and autoimmune processes such as the production of autoantibodies (e.g., antibodies against citrullinated proteins ACPA or rheumatoid factors, RF), development of the first symptoms without clinical arthritis, and, finally, the manifestation of arthritis. Despite the typical joint involvement in established RA, the pathogenesis of the disease likely begins far from joint structures: in the lungs or periodontium in association with citrullination, intestinal microbiome, or adipose tissue, which supports normal findings in synovial tissue in ACPA+ patients with arthralgia. The presence of ACPA is detectable even years before the first manifestation of RA. The pre-clinical phase of RA is the period preceding clinically apparent RA with ACPA contributing to the symptoms without subclinical inflammation. While the combination of ACPA and RF increases the risk of progression to RA by up to 10 times, increasing numbers of novel autoantibodies are to be investigated to contribute to the increased risk and pathogenesis of RA. With growing knowledge about the course of RA, new aspiration emerges to cure and even prevent RA, shifting the "window of opportunity" to the pre-clinical phases of RA. The clinical definition of individuals at risk of developing RA (clinically suspect arthralgia, CSA) makes it possible to unify these at-risk individuals' clinical characteristics for "preventive" treatment in ongoing clinical trials using mostly biological or conventional synthetic disease-modifying drugs. However, the combination of symptoms, laboratory, and imaging biomarkers may be the best approach to select the correct target at-risk population. The current review aims to explore different phases of RA and discuss the potential of (non)pharmacological intervention aiming to prevent RA.
Collapse
|
21
|
Perniola R, Fierabracci A, Falorni A. Autoimmune Addison's Disease as Part of the Autoimmune Polyglandular Syndrome Type 1: Historical Overview and Current Evidence. Front Immunol 2021; 12:606860. [PMID: 33717087 PMCID: PMC7953157 DOI: 10.3389/fimmu.2021.606860] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/25/2021] [Indexed: 12/11/2022] Open
Abstract
The autoimmune polyglandular syndrome type 1 (APS1) is caused by pathogenic variants of the autoimmune regulator (AIRE) gene, located in the chromosomal region 21q22.3. The related protein, AIRE, enhances thymic self-representation and immune self-tolerance by localization to chromatin and anchorage to multimolecular complexes involved in the initiation and post-initiation events of tissue-specific antigen-encoding gene transcription. Once synthesized, the self-antigens are presented to, and cause deletion of, the self-reactive thymocyte clones. The clinical diagnosis of APS1 is based on the classic triad idiopathic hypoparathyroidism (HPT)-chronic mucocutaneous candidiasis-autoimmune Addison's disease (AAD), though new criteria based on early non-endocrine manifestations have been proposed. HPT is in most cases the first endocrine component of the syndrome; however, APS1-associated AAD has received the most accurate biochemical, clinical, and immunological characterization. Here is a comprehensive review of the studies on APS1-associated AAD from initial case reports to the most recent scientific findings.
Collapse
Affiliation(s)
- Roberto Perniola
- Department of Pediatrics-Neonatal Intensive Care, V. Fazzi Hospital, ASL LE, Lecce, Italy
| | - Alessandra Fierabracci
- Infectivology and Clinical Trials Research Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Alberto Falorni
- Section of Internal Medicine and Endocrinological and Metabolic Sciences, Department of Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
22
|
Moritz CP, Tholance Y, Stoevesandt O, Ferraud K, Camdessanché JP, Antoine JC. CIDP Antibodies Target Junction Proteins and Identify Patient Subgroups: An Autoantigenomic Approach. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/2/e944. [PMID: 33408168 PMCID: PMC7862091 DOI: 10.1212/nxi.0000000000000944] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 10/29/2020] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To discover systemic characteristics in the repertoires of targeted autoantigens in chronic inflammatory demyelinating polyneuropathy (CIDP), we detected the entire autoantigen repertoire of patients and controls and analyzed them systematically. METHODS We screened 43 human serum samples, of which 22 were from patients with CIDP, 12 from patients with other neuropathies, and 9 from healthy controls via HuProt Human Proteome microarrays testing about 16,000 distinct human bait proteins. Autoantigen repertoires were analyzed via bioinformatical autoantigenomic approaches: principal component analysis, analysis of the repertoire sizes in disease groups and clinical subgroups, and overrepresentation analyses using Gene Ontology and PantherDB. RESULTS The autoantigen repertoires enabled the identification of a subgroup of 10/22 patients with CIDP with a younger age at onset and a higher frequency of mixed motor and sensory CIDP. IV immunoglobulin therapy responders targeted 3 times more autoantigens than nonresponders. No CIDP-specific autoantibody is present in all patients; however, anchoring junction components were significantly targeted by 86.4% of patients with CIDP. There are potential novel CIDP-specific autoantigens such as the myelination- or axo-glial structure-related proteins actin-related protein 2/3 complex subunit 1B, band 4.1-like protein 2, cadherin-15, cytohesin-1, epidermal growth factor receptor, ezrin, and radixin. CONCLUSIONS The repertoire of targeted autoantigens of patients with CIDP differs in a systematic degree from those of controls. Systematic autoantigenomic approaches can help to understand the disease and to discover novel bioinformatical tools and novel autoantigen panels to improve diagnosis, treatment, prognosis, or patient stratification.
Collapse
Affiliation(s)
- Christian P Moritz
- From the Department of Neurology (C.P.M., K.F., J.-P.C., J.-C.A.), and Department of Biochemistry (Y.T.), University Hospital of Saint-Etienne; Synaptopathies and Autoantibodies (C.P.M., Y.T., J.-P.C., J.-C.A.), Institut NeuroMyoGène, INSERM U1217/CNRS UMR 5310, University of Lyon, University Jean-Monnet, Saint-Étienne, France; and Cambridge Protein Arrays Ltd. (O.S.), Babraham Research Campus, United Kingdom.
| | - Yannick Tholance
- From the Department of Neurology (C.P.M., K.F., J.-P.C., J.-C.A.), and Department of Biochemistry (Y.T.), University Hospital of Saint-Etienne; Synaptopathies and Autoantibodies (C.P.M., Y.T., J.-P.C., J.-C.A.), Institut NeuroMyoGène, INSERM U1217/CNRS UMR 5310, University of Lyon, University Jean-Monnet, Saint-Étienne, France; and Cambridge Protein Arrays Ltd. (O.S.), Babraham Research Campus, United Kingdom
| | - Oda Stoevesandt
- From the Department of Neurology (C.P.M., K.F., J.-P.C., J.-C.A.), and Department of Biochemistry (Y.T.), University Hospital of Saint-Etienne; Synaptopathies and Autoantibodies (C.P.M., Y.T., J.-P.C., J.-C.A.), Institut NeuroMyoGène, INSERM U1217/CNRS UMR 5310, University of Lyon, University Jean-Monnet, Saint-Étienne, France; and Cambridge Protein Arrays Ltd. (O.S.), Babraham Research Campus, United Kingdom
| | - Karine Ferraud
- From the Department of Neurology (C.P.M., K.F., J.-P.C., J.-C.A.), and Department of Biochemistry (Y.T.), University Hospital of Saint-Etienne; Synaptopathies and Autoantibodies (C.P.M., Y.T., J.-P.C., J.-C.A.), Institut NeuroMyoGène, INSERM U1217/CNRS UMR 5310, University of Lyon, University Jean-Monnet, Saint-Étienne, France; and Cambridge Protein Arrays Ltd. (O.S.), Babraham Research Campus, United Kingdom
| | - Jean-Philippe Camdessanché
- From the Department of Neurology (C.P.M., K.F., J.-P.C., J.-C.A.), and Department of Biochemistry (Y.T.), University Hospital of Saint-Etienne; Synaptopathies and Autoantibodies (C.P.M., Y.T., J.-P.C., J.-C.A.), Institut NeuroMyoGène, INSERM U1217/CNRS UMR 5310, University of Lyon, University Jean-Monnet, Saint-Étienne, France; and Cambridge Protein Arrays Ltd. (O.S.), Babraham Research Campus, United Kingdom
| | - Jean-Christophe Antoine
- From the Department of Neurology (C.P.M., K.F., J.-P.C., J.-C.A.), and Department of Biochemistry (Y.T.), University Hospital of Saint-Etienne; Synaptopathies and Autoantibodies (C.P.M., Y.T., J.-P.C., J.-C.A.), Institut NeuroMyoGène, INSERM U1217/CNRS UMR 5310, University of Lyon, University Jean-Monnet, Saint-Étienne, France; and Cambridge Protein Arrays Ltd. (O.S.), Babraham Research Campus, United Kingdom
| |
Collapse
|
23
|
Pyroptosis by caspase-11 inflammasome-Gasdermin D pathway in autoimmune diseases. Pharmacol Res 2021; 165:105408. [PMID: 33412278 DOI: 10.1016/j.phrs.2020.105408] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/28/2020] [Accepted: 12/28/2020] [Indexed: 02/08/2023]
Abstract
Inflammasomes are a group of supramolecular complexes primarily comprise a sensor, adaptor protein and an effector. Among them, canonical inflammasomes are assembled by one specific pattern recognition receptor, the adaptor protein apoptosis-associated speck-like protein containing a CARD and procaspase-1. Murine caspase-11 and its human ortholog caspase-4/5 are identified as cytosolic sensors which directly responds to LPS. Once gaining access to cytosol, LPS further trigger inflammasome activation in noncanonical way. Downstream pore-forming Gasdermin D is a pyroptosis executioner. Emerging evidence announced in recent years demonstrate the vital role played by caspase-11 non-canonical inflammasome in a range of autoimmune diseases. Pharmacological ablation of caspase-11 and its related effector results in potent therapeutic effects. Though recent advances have highlighted the potential of caspase-11 as a drug target, the understanding of caspase-11 molecular activation and regulation mechanism remains to be limited and thus hampered the discovery and progression of novel inhibitors. Here in this timeline review, we explored how caspase-11 get involved in the pathogenesis of autoimmune diseases, we also collected the reported small-molecular caspase-11 inhibitors. Moreover, the clinical implications and therapeutic potential of caspase-11 inhibitors are discussed. Targeting non-canonical inflammasomes is a promising strategy for autoimmune diseases treatment, while information about the toxicity and physiological disposition of the promising caspase-11 inhibitors need to be supplemented before they can be translated from bench to bedside.
Collapse
|
24
|
An updated advance of autoantibodies in autoimmune diseases. Autoimmun Rev 2020; 20:102743. [PMID: 33333232 DOI: 10.1016/j.autrev.2020.102743] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 08/06/2020] [Indexed: 12/18/2022]
Abstract
Autoantibodies are abnormal antibodies which are generated by pathogenic B cells when targeting an individual's own tissue. Autoantibodies have been identified as a symbol of autoimmune disorders and are frequently considered a clinical marker of these disorders. Autoimmune diseases, including system lupus erythematosus and rheumatoid arthritis, consist of a series of disorders that share some similarities and differences. They are characterized by chronic, systemic, excessive immune activation and inflammation and involve in almost all body tissues. Autoimmune diseases occur more frequently in women than men due to hormonal impacts. In this review we systemically introduce and summarize the latest advances of various autoantibodies in multiple autoimmune diseases.
Collapse
|
25
|
Zeneyedpour L, Sten-van `t Hoff J, Luider T. Using phosphoproteomics and next generation sequencing to discover novel therapeutic targets in patient antibodies. Expert Rev Proteomics 2020; 17:675-684. [DOI: 10.1080/14789450.2020.1845147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Lona Zeneyedpour
- Department of Neurology, Erasmus MC, Laboratory of Neuro-Oncology/Clinical & Cancer Proteomics, Rotterdam, The Netherlands
| | - Jenny Sten-van `t Hoff
- Department of Neurology, Erasmus MC, Laboratory of Neuro-Oncology/Clinical & Cancer Proteomics, Rotterdam, The Netherlands
| | - Theo Luider
- Department of Neurology, Erasmus MC, Laboratory of Neuro-Oncology/Clinical & Cancer Proteomics, Rotterdam, The Netherlands
| |
Collapse
|
26
|
Moritz CP, Stoevesandt O, Tholance Y, Camdessanché JP, Antoine JC. Proper definition of the set of autoantibody-targeted antigens relies on appropriate reference group selection. N Biotechnol 2020; 60:168-172. [PMID: 33045420 DOI: 10.1016/j.nbt.2020.08.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 08/04/2020] [Accepted: 08/30/2020] [Indexed: 02/06/2023]
Abstract
Autoimmune diseases are frequently associated with autoantibodies. Recently, large sets of autoantibody-targeted antigens ("autoantigen-omes") of patient and control sera have been revealed, enabling autoantigen-omic approaches. However, statistical standards for defining such autoantigen-omes are lacking. The z-score indicates how many standard deviations an antigen reactivity of a given sample is from the mean reactivity of the corresponding antigen in a reference group. Hence, it is a common measure to define significantly positive reactivity in autoantigen profiling approaches. Here, we address the risk of biased analyses resulting from unbalanced selection of the reference group. Three study groups were selected. Patients-of-interest were chronic inflammatory demyelinating polyneuropathy (CIDP); controls were other neuropathies (ONP); and healthy controls (HC). Each serum was screened for significant autoantigen reactivity using HuProt™ protein arrays. We compared three possible selections of reference groups for statistical z-score calculations: method#1, the control groups (ONP + HC); method #2, all groups together; method #3, the respective other groups (e.g., CIDP + HC for the ONP autoantigen-ome). The method selection seriously affected the size of the autoantigen-omes. Method #1 introduced a bias favoring significantly more antigens per patient in the CIDP group (for z >4: 19 ± 3 antigens) than in the control groups (ONP: 2 ± 1; HC: 0 ± 0). The more balanced methods #2 and #3 did not result in significant differences. This contribution may help to avoid interpretation biases and to develop guidelines for population studies revealing autoantigen-omes via high throughput studies such as protein microarrays, immunoprecipitation with mass spectrometry, or phage display assays.
Collapse
Affiliation(s)
- Christian P Moritz
- Synaptopathies and Autoantibodies, Institut NeuroMyoGene INSERM U1217/CNRS UMR, Faculty of Medicine Jacques Lisfranc, University Jean Monnet, University of Lyon, Saint-Étienne, France; Department of Neurology, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Étienne, France.
| | - Oda Stoevesandt
- Cambridge Protein Arrays Ltd., Babraham Research Campus, Cambridge, UK
| | - Yannick Tholance
- Synaptopathies and Autoantibodies, Institut NeuroMyoGene INSERM U1217/CNRS UMR, Faculty of Medicine Jacques Lisfranc, University Jean Monnet, University of Lyon, Saint-Étienne, France; Department of Biochemistry, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Étienne, France
| | - Jean-Philippe Camdessanché
- Synaptopathies and Autoantibodies, Institut NeuroMyoGene INSERM U1217/CNRS UMR, Faculty of Medicine Jacques Lisfranc, University Jean Monnet, University of Lyon, Saint-Étienne, France; Department of Neurology, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Étienne, France
| | - Jean-Christophe Antoine
- Synaptopathies and Autoantibodies, Institut NeuroMyoGene INSERM U1217/CNRS UMR, Faculty of Medicine Jacques Lisfranc, University Jean Monnet, University of Lyon, Saint-Étienne, France; Department of Neurology, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Étienne, France
| |
Collapse
|
27
|
Roccatello D, Fenoglio R, Baldovino S, Naretto C, Ferro M, Barreca A, Rossi D, Sciascia S. Towards a novel target therapy for renal diseases related to plasma cell dyscrasias: The example of AL amyloidosis. Autoimmun Rev 2020; 19:102622. [PMID: 32663622 DOI: 10.1016/j.autrev.2020.102622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 04/11/2020] [Indexed: 11/24/2022]
Abstract
Immunoglobulin light chain amyloidosis is a rare systemic disease caused by monoclonal light chains (LCs) depositing in tissue as insoluble fibrils resulting in irreversible damage of vital organs. The mechanisms involved in aggregation and deposition of LCs are not fully understood, but CD138/38 plasma cells are undoubtedly involved in monoclonal LC production. We are reporting favorable effects on AL amyloidosis patients with renal involvement using the anti-CD38 monoclonal antibody Daratumumab. We speculate that research for the near future should be devoted to design similar therapeutic approaches for other diseases attributable to a plasma cell dyscrasia.
Collapse
Affiliation(s)
- Dario Roccatello
- Nephrology and Dialysis Unit-CMID, Center of Research of Immunopathology and Rare Diseases, Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hospital of Turin, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy.
| | - Roberta Fenoglio
- Nephrology and Dialysis Unit-CMID, Center of Research of Immunopathology and Rare Diseases, Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hospital of Turin, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Simone Baldovino
- Nephrology and Dialysis Unit-CMID, Center of Research of Immunopathology and Rare Diseases, Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hospital of Turin, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Carla Naretto
- Nephrology and Dialysis Unit-CMID, Center of Research of Immunopathology and Rare Diseases, Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hospital of Turin, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Michela Ferro
- Nephrology and Dialysis Unit-CMID, Center of Research of Immunopathology and Rare Diseases, Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hospital of Turin, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Antonella Barreca
- Pathology Division, Department of Oncology, University of Turin, Italy
| | - Daniela Rossi
- Nephrology and Dialysis Unit-CMID, Center of Research of Immunopathology and Rare Diseases, Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hospital of Turin, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Savino Sciascia
- Nephrology and Dialysis Unit-CMID, Center of Research of Immunopathology and Rare Diseases, Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hospital of Turin, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| |
Collapse
|