1
|
Steri M, Orrù V, Sidore C, Mulas A, Pitzalis M, Busonero F, Maschio A, Serra V, Dei M, Lai S, Virdis F, Lobina M, Loizedda A, Marongiu M, Masala M, Floris M, Curreli N, Balaci L, Loi F, Pilia MG, Delitala A, Fiorillo E, Schlessinger D, Zoledziewska M. TYK2 :p.Pro1104Ala Variant Protects Against Autoimmunity by Modulating Immune Cell Levels. Immunology 2025; 174:462-469. [PMID: 39835539 PMCID: PMC11885862 DOI: 10.1111/imm.13902] [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: 09/26/2024] [Revised: 12/23/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
The TYK2:p.Pro1104Ala (rs34536443) hypomorph variant has been associated with protection against numerous autoimmune disorders. Thus, its mechanism of action becomes of great interest. Here, consistent with the participation of activated immune cells in autoimmunity, we show that the variant regulates the levels of immune cells at a human, general population level and is associated particularly with higher levels of T and B lymphocytes, especially the naïve (non-activated) compartment. Also, consistent with a protective function in autoimmunity, the level of regulatory CD4+ T cells was increased. Thus, this variant decreases immune activation thereby protecting from autoimmunity. Our work links the cellular mechanism regulated by the TYK2:p.Pro1104Ala variant to autoimmunity protection and supports TYK2 as a therapeutic target in autoimmunity.
Collapse
Affiliation(s)
- Maristella Steri
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Valeria Orrù
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Carlo Sidore
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Antonella Mulas
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Maristella Pitzalis
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Fabio Busonero
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Andrea Maschio
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Valentina Serra
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Mariano Dei
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Sandra Lai
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Francesca Virdis
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Monia Lobina
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Annalisa Loizedda
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Michele Marongiu
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Marco Masala
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Matteo Floris
- Department of Biomedical SciencesUniversity of SassariSassariItaly
| | - Nicolò Curreli
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Lenuta Balaci
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Francesco Loi
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Maria Grazia Pilia
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - Alessandro Delitala
- Department of Medicine, Surgery and PharmacyUniversity of SassariSassariItaly
| | - Edoardo Fiorillo
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| | - David Schlessinger
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA)National Institutes of Health (NIH)BaltimoreMarylandUSA
| | - Magdalena Zoledziewska
- Institute of Genetic and Biomedical Research (IRGB)Italian National Research Council (CNR)MonserratoSardiniaItaly
| |
Collapse
|
2
|
Royo M, Joseph-Mullol B, Sandoval S, Moliné T, Solé C, Cortés-Hernández J. Integrative miRNA-mRNA profiling uncovers mechanisms of belimumab action in systemic lupus erythematosus. Front Immunol 2025; 16:1553971. [PMID: 40160819 PMCID: PMC11949941 DOI: 10.3389/fimmu.2025.1553971] [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: 12/31/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disorder driven by autoreactive B cells and characterized by the production of pathogenic autoantibodies. Belimumab, an anti-BAFF monoclonal antibody, has demonstrated efficacy in reducing disease activity and corticosteroid use in SLE patients, although responses remain variable. B-cell activating factor (BAFF) is essential for B cell survival and autoantibody production, positioning it as a key target in SLE pathogenesis. MicroRNAs (miRNAs), critical regulators of gene expression and immune homeostasis, have an emerging role in SLE pathophysiology. However, their regulation in response to anti-BAFF therapies, such as belimumab, remains unexplored. This study investigates miRNA-mRNA interactions in T cells, B cells, and myeloid cells from SLE patients before and after belimumab treatment. A total of 79 miRNAs associated with treatment response and 525 miRNA-gene interactions were identified. Validation in 18 SLE responders revealed significant changes in miRNA expression in T and myeloid cells, but not in B cells. Belimumab was found to modulate B cell development by regulating genes such as BLNK, BANK1, and MEF2C, as well as the CD40/CD40L axis. In T cells, miRNAs influenced interferon signaling and inflammatory cytokines via NF-κB activation. Changes in myeloid cells, characterized by the downregulation of KLF13, CCL5, and IL4, appear to be secondary to T cell modulation. These findings provide novel insights into the miRNA-mediated regulatory networks underlying belimumab's immunomodulatory effects in SLE. Further research is required to validate these findings and through in vitro experiments to better understand the role of miRNAs in guiding treatment responses.
Collapse
Affiliation(s)
- Maria Royo
- Rheumatology Research Group, Lupus Unit, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Blanca Joseph-Mullol
- Rheumatology Research Group, Lupus Unit, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sebastian Sandoval
- Rheumatology Research Group, Lupus Unit, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Teresa Moliné
- Department of Pathology, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cristina Solé
- Rheumatology Research Group, Lupus Unit, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Josefina Cortés-Hernández
- Rheumatology Research Group, Lupus Unit, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
3
|
Di Sabatino E, Ferraro D, Gaetani L, Emiliano E, Parnetti L, Di Filippo M. CSF biomarkers of B-cell activation in multiple sclerosis: a clinical perspective. J Neurol 2025; 272:211. [PMID: 39960641 PMCID: PMC11832686 DOI: 10.1007/s00415-025-12907-6] [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: 10/27/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 02/20/2025]
Abstract
The role of B cells in the pathophysiology of multiple sclerosis (MS) extends beyond antibody synthesis, also involving the modulation of T lymphocytes and myeloid cells. B-cell activation within the Central Nervous System is associated with the release of various antibodies, cytokines, and chemokines, measurable in biofluids, thereby serving as biomarkers of the immune processes responsible for MS. To this purpose, a biomarker-based characterization of the disease through the combination of well-established markers, e.g., immunoglobulin (Ig) G index, IgG oligoclonal bands, Ig free light chains, with new promising markers, namely chemokine (C-X-C motif) ligand 13, and B-cell activating factor/A proliferation-inducing ligand, might represent a significant improvement in the management of people with MS.
Collapse
Affiliation(s)
- Elena Di Sabatino
- Clinica Neurologica, Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Umbria, Italy
| | - Diana Ferraro
- Dipartimento di Neuroscienze, Ospedale Civile di Baggiovara, Azienda Ospedaliera-Università di Modena, Modena, Italy
| | - Lorenzo Gaetani
- Clinica Neurologica, Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Umbria, Italy
| | - Edoardo Emiliano
- Clinica Neurologica, Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Umbria, Italy
| | - Lucilla Parnetti
- Clinica Neurologica, Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Umbria, Italy
| | - Massimiliano Di Filippo
- Clinica Neurologica, Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Umbria, Italy.
| |
Collapse
|
4
|
Jägerback S, Gomez A, Parodis I. Predictors of renal flares in systemic lupus erythematosus: a post-hoc analysis of four phase III clinical trials of belimumab. Rheumatology (Oxford) 2025; 64:623-631. [PMID: 38216728 PMCID: PMC11781576 DOI: 10.1093/rheumatology/keae023] [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: 03/21/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 01/14/2024] Open
Abstract
OBJECTIVE The objective of this study was to identify predictors of renal flares in patients with SLE treated for active extra-renal disease. METHODS Data from four clinical trials of belimumab in SLE (BLISS-52, NCT00424476; BLISS-76, NCT00410384; BLISS-NEA, NCT01345253; BLISS-SC, NCT01484496) were used. Patients were assigned to belimumab or placebo on top of standard therapy. We investigated the performance of predictors of renal flares through weeks 52-76 using proportional hazards regression analysis. RESULTS Of 3225 participants, 192 developed at least one renal flare during follow-up, with the first occurring after a median time of 197 days. Current/former renal involvement [hazards ratio (HR): 15.4; 95% CI: 8.3-28.2; P < 0.001], low serum albumin levels (HR 0.9; 95% CI: 0.8-0.9; P < 0.001), proteinuria (HR: 1.6; 95% CI: 1.5-1.7; P < 0.001), and low C3 levels (HR: 2.9; 95% CI: 2.1-4.1; P < 0.001) at baseline appeared robust determinants of impending renal flares. Anti-dsDNA positivity yielded an increased hazard for renal flares (HR: 2.1; 95% CI: 1.4-3.2; P < 0.001), which attenuated after adjustments. Anti-Sm positivity was associated with renal flares in the placebo (HR: 3.7; 95% CI: 2.0-6.9; P < 0.001) but not in the belimumab subgroup, whereas anti-ribosomal P positivity was associated with renal flares in the belimumab subgroup only (HR: 2.8; 95% CI: 1.5-5.0; P = 0.001). CONCLUSION A history of renal involvement, high baseline proteinuria, hypoalbuminaemia, and C3 consumption were robust determinants of impending renal flares. In addition to anti-dsDNA, anti-Sm and anti-ribosomal P protein antibody positivity may have value in surveillance of renal SLE.
Collapse
Affiliation(s)
- Sandra Jägerback
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Division of Rheumatology, Department of Medicine, Danderyd University Hospital, Danderyd, Sweden
| | - Alvaro Gomez
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
5
|
Hu L, Zhang X, Zhang W, Jin S, Zhao J, Zheng J, Song W, Shen Z. Targeting TCMR-associated cytokine genes for drug screening identifies PPARγ agonists as novel immunomodulatory agents in transplantation. Front Immunol 2025; 16:1539645. [PMID: 39911401 PMCID: PMC11794815 DOI: 10.3389/fimmu.2025.1539645] [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: 12/04/2024] [Accepted: 01/06/2025] [Indexed: 02/07/2025] Open
Abstract
Objective T cell-mediated rejection (TCMR) remains a significant challenge in organ transplantation. This study aimed to define a TCMR-associated cytokine gene set and identify drugs to prevent TCMR through drug repurposing. Methods Gene expression profiles from kidney, heart, and lung transplant biopsies were obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) between TCMR and non-TCMR groups were identified, and their intersection with cytokine-related genes yielded an 11-gene TCMR-associated cytokine gene set (TCMR-Cs). To evaluate the effectiveness of this gene set, a diagnostic predictive model was constructed using Lasso regression and multivariate logistic regression, with validation in independent datasets. Connectivity Map (CMap) analysis was employed to screen drugs targeting TCMR-Cs. Experimental validation of the identified drug was performed in vitro using T cell activation and Th1 differentiation assays, and in vivo in a mouse skin transplant model with survival analysis. Results The TCMR-Cs exhibited outstanding predictive performance for TCMR, achieving an AUC of 0.99 in the training cohorts and maintaining strong performance in the test cohorts. CMap analysis identified peroxisome proliferator-activated receptor gamma (PPARγ) agonists as potential therapeutic candidates. Experimental validation showed that the PPARγ agonist rosiglitazone significantly suppressed T cell activation and reduced Th1 differentiation in vitro without cytotoxic effects. The combination of rosiglitazone and rapamycin significantly prolonged graft survival. Conclusions This study defined a novel TCMR-associated cytokine gene set that effectively predicts TCMR and identified PPARγ agonists, which prevent TCMR and improve graft survival when combined with rapamycin.
Collapse
Affiliation(s)
- Lu Hu
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
- Research Institute of Transplant Medicine, School of Medicine, Nankai University, Tianjin, China
| | - Xiaohan Zhang
- Research Institute of Transplant Medicine, School of Medicine, Nankai University, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| | - Weiqi Zhang
- Research Institute of Transplant Medicine, School of Medicine, Nankai University, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| | - Shuai Jin
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
- Research Institute of Transplant Medicine, School of Medicine, Nankai University, Tianjin, China
| | - Jie Zhao
- Department of Renal Transplantation, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Jianming Zheng
- Department of Renal Transplantation, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Wenli Song
- Department of Renal Transplantation, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Zhongyang Shen
- Research Institute of Transplant Medicine, School of Medicine, Nankai University, Tianjin, China
- NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin, China
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| |
Collapse
|
6
|
Paul SK, Saddam M, Tabassum N, Hasan M. Molecular dynamics simulation of wild and mutant proteasome subunit beta type 8 (PSMB8) protein: Implications for restoration of inflammation in experimental autoimmune encephalomyelitis pathogenesis. Heliyon 2025; 11:e41166. [PMID: 39802026 PMCID: PMC11719297 DOI: 10.1016/j.heliyon.2024.e41166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025] Open
Abstract
Multiple Sclerosis (MS) is an autoimmune and chronic disease in the brain and spinal cord. MS has inflammatory progression characterized by its hallmark inflammatory plaques. The histological and clinical characteristics of MS are shared by Experimental Autoimmune Encephalomyelitis (EAE). Genetic and environmental factors contribute to the development of MS. In EAE-MS disease, the level of proteasome subunit beta type-8 (PSMB8), encoded by the PSMB8 gene, is increased and regulates the inflammatory response in this disease. In humans, the Nakajo-Nishimura Syndrome is caused by a mutation in the gene PSMB8, a part of the immunoproteasome subunit. Therefore, special attention to wild and mutant (G210V) PSMB8 protein is imperative. In this study, we performed a 100 ns molecular dynamics (MD) simulation for wild-type PSMB8 and the mutant G210V. Then, we analyzed the fundamental and essential simulation results using another Google Colab system. The energy analysis ensures the structural deviation due to point mutation. The trajectory of the fundamental simulation (RMSD, RMSF, and Rg) describes that the G210V mutated protein is more flexible and less stable than the wild type. We observed the conformational changes due to mutation by analyzing the RMSD average linkage hierarchical clustering, total SASA, and SASA autocorrelation. The differences in the protein's overall motion and the atoms' precise location are identified by the principal component analysis, showing that the overall motion and location of the atoms are different. Our study provides valuable insights into the dynamics and structure of this protein, which can aid in further understanding its biological functions and potential implications for disease.
Collapse
Affiliation(s)
- Shamrat Kumar Paul
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Md Saddam
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Nisat Tabassum
- Department of Biotechnology and Genetic Engineering, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Mahbub Hasan
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| |
Collapse
|
7
|
Diehl C, Soberón V, Baygün S, Chu Y, Mandelbaum J, Kraus L, Engleitner T, Rudelius M, Fangazio M, Daniel C, Bortoluzzi S, Helmrath S, Singroul P, Gölling V, Osorio Barrios F, Seyhan G, Oßwald L, Kober-Hasslacher M, Zeng T, Öllinger R, Afzali AM, Korn T, Honarpisheh M, Lech M, Ul Ain Q, Pircher J, Imširović V, Jelenčić V, Wensveen FM, Passerini V, Bärthel S, Bhagat G, Dominguez-Sola D, Saur D, Steiger K, Rad R, Pasqualucci L, Weigert O, Schmidt-Supprian M. Hyperreactive B cells instruct their elimination by T cells to curb autoinflammation and lymphomagenesis. Immunity 2025; 58:124-142.e15. [PMID: 39729992 DOI: 10.1016/j.immuni.2024.11.023] [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: 02/21/2024] [Revised: 08/14/2024] [Accepted: 11/26/2024] [Indexed: 12/29/2024]
Abstract
B cell immunity carries the inherent risk of deviating into autoimmunity and malignancy, which are both strongly associated with genetic variants or alterations that increase immune signaling. Here, we investigated the interplay of autoimmunity and lymphoma risk factors centered around the archetypal negative immune regulator TNFAIP3/A20 in mice. Counterintuitively, B cells with moderately elevated sensitivity to stimulation caused fatal autoimmune pathology, while those with high sensitivity did not. We resolved this apparent paradox by identifying a rheostat-like cytotoxic T cell checkpoint. Cytotoxicity was instructed by and directed against B cells with high intrinsic hyperresponsiveness, while less reactive cells were spared. Removing T cell control restored a linear relationship between intrinsic B cell reactivity and lethal lymphoproliferation, lymphomagenesis, and autoinflammation. We thus identify powerful T cell-mediated negative feedback control of inherited and acquired B cell pathogenicity and define a permissive window for autoimmunity to emerge.
Collapse
Affiliation(s)
- Carina Diehl
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Valeria Soberón
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Max-Planck Institute of Biochemistry, 82152 Planegg, Germany
| | - Seren Baygün
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Yuanyuan Chu
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Max-Planck Institute of Biochemistry, 82152 Planegg, Germany
| | - Jonathan Mandelbaum
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Laura Kraus
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Thomas Engleitner
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Martina Rudelius
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University, 80337 Munich, Germany
| | - Marco Fangazio
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Christoph Daniel
- Department of Nephropathology, Faculty of Medicine, Friedrich-Alexander University (FAU) Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Sabrina Bortoluzzi
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Sabine Helmrath
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Pankaj Singroul
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Vanessa Gölling
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Francisco Osorio Barrios
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Gönül Seyhan
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Lena Oßwald
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; Department of Medicine III, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Maike Kober-Hasslacher
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Max-Planck Institute of Biochemistry, 82152 Planegg, Germany
| | - Theodor Zeng
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Rupert Öllinger
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Ali M Afzali
- Institute for Experimental Neuroimmunology, Department of Neurology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Thomas Korn
- Institute for Experimental Neuroimmunology, Department of Neurology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Mohsen Honarpisheh
- Renal Division, Department of Medicine IV, Faculty of Medicine, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Maciej Lech
- Renal Division, Department of Medicine IV, Faculty of Medicine, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Qurrat Ul Ain
- Department of Medicine I, Faculty of Medicine, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Joachim Pircher
- Department of Medicine I, Faculty of Medicine, Ludwig-Maximilians-University, 81377 Munich, Germany; Partner site Munich Heart Alliance, DZHK (German Centre for Cardiovascular Research), 80802 Munich, Germany
| | - Vanna Imširović
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Vedrana Jelenčić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Felix M Wensveen
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Verena Passerini
- Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Faculty of Medicine, Department of Medicine III, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Stefanie Bärthel
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Chair of Translational Cancer Research and Institute of Experimental Cancer Therapy, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Govind Bhagat
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | - David Dominguez-Sola
- Departments of Oncological Sciences and Pathology, Tisch Cancer Institute, Lipschultz Precision Immunology Institute, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dieter Saur
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Chair of Translational Cancer Research and Institute of Experimental Cancer Therapy, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Katja Steiger
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Institute of Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Roland Rad
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Laura Pasqualucci
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | - Oliver Weigert
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Faculty of Medicine, Department of Medicine III, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Marc Schmidt-Supprian
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Max-Planck Institute of Biochemistry, 82152 Planegg, Germany.
| |
Collapse
|
8
|
Kacar M, Al-Hakim A, Savic S. Sequelae of B-Cell Depleting Therapy: An Immunologist's Perspective. BioDrugs 2025; 39:103-130. [PMID: 39680306 DOI: 10.1007/s40259-024-00696-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2024] [Indexed: 12/17/2024]
Abstract
B-cell depleting therapy (BCDT) has revolutionised the treatment of B-cell malignancies and autoimmune diseases by targeting specific B-cell surface antigens, receptors, ligands, and signalling pathways. This narrative review explores the mechanisms, applications, and complications of BCDT, focusing on the therapeutic advancements since the introduction of rituximab in 1997. Various monoclonal antibodies and kinase inhibitors are examined for their roles in depleting B cells through antibody-dependent and independent mechanisms. The off-target effects, such as hypogammaglobulinemia, infections, and cytokine release syndrome, are discussed, emphasising the need for immunologists to identify and help manage these complications. The increasing prevalence of BCDT has necessitated the involvement of clinical immunologists in addressing treatment-associated immunological abnormalities, including persistent hypogammaglobulinemia and neutropenia. We highlight the importance of considering underlying inborn errors of immunity (IEI) in patients presenting with these complications. Furthermore, we discuss the impact of BCDT on other immune cell populations and the challenges in predicting and managing long-term immunological sequelae. The potential for novel BCDT agents targeting the BAFF/APRIL-TACI/BCMA axis and B-cell receptor signalling pathways to treat autoimmune disorders is also explored, underscoring the rapidly evolving landscape of B-cell targeted therapies.
Collapse
Affiliation(s)
- Mark Kacar
- Department of Allergy, University Clinic Golnik, Golnik, Slovenia
- Department of Allergy and Clinical Immunology, St James' University Hospital, Leeds, UK
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Adam Al-Hakim
- Department of Allergy and Clinical Immunology, St James' University Hospital, Leeds, UK
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Sinisa Savic
- Department of Allergy and Clinical Immunology, St James' University Hospital, Leeds, UK.
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.
- NIHR Leeds Biomedical Research Centre, Leeds, UK.
| |
Collapse
|
9
|
Keritam O, Vincent A, Zimprich F, Cetin H. A clinical perspective on muscle specific kinase antibody positive myasthenia gravis. Front Immunol 2024; 15:1502480. [PMID: 39703505 PMCID: PMC11655327 DOI: 10.3389/fimmu.2024.1502480] [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: 09/26/2024] [Accepted: 11/20/2024] [Indexed: 12/21/2024] Open
Abstract
The discovery of autoantibodies directed against muscle-specific kinase (MuSK) in "seronegative" myasthenia gravis (MG) patients marked a milestone in MG research. In healthy muscle, MuSK regulates a phosphorylation pathway, which is essential for the development and maintenance of acetylcholine receptor (AChR) clusters at the neuromuscular junction. Autoantibodies directed against MuSK are predominantly of the IgG4 subclass, but there is increasing evidence that IgG1-3 could also contribute to the pathology underlying MuSK-MG. MuSK-IgG4 are monovalent and block the binding site for LRP4 on MuSK, thereby inhibiting the downstream phosphorylation pathway and compromising the formation of AChR clusters. Clinically, MuSK-MG is commonly associated with the predominant involvement of bulbar, facial, shoulder and neck muscles. Cholinesterase inhibitors should be avoided in MuSK-MG due to the risk of clinical impairment and cholinergic crisis. Corticosteroids and other non-steroidal immunosuppressants are less effective with the need for higher doses and prolonged treatment. Rituximab, by contrast, has been shown to be particularly effective and is now often used early in the disease course. Its use is associated with a significant improvement in the clinical outcome of MuSK-MG patients over time. This review aims to describe the pathophysiology underlying MuSK-MG and provide a comprehensive overview of the clinical features and therapeutic options.
Collapse
Affiliation(s)
- Omar Keritam
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Vienna, Austria
| | - Angela Vincent
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Fritz Zimprich
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Vienna, Austria
| | - Hakan Cetin
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
10
|
Li Z, Chen P, Zhang Y, Chen J, Zheng S, Li W, Tang L, Liu Y, Zhao N. Serum BAFF levels are associated with the prognosis of idiopathic membranous nephropathy. Ren Fail 2024; 46:2391069. [PMID: 39143819 PMCID: PMC11328817 DOI: 10.1080/0886022x.2024.2391069] [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: 03/26/2024] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024] Open
Abstract
OBJECTIVE High serum levels of B-cell activation factor (BAFF) and a proliferation-inducing ligand (APRIL) have been observed in patients with idiopathic membranous nephropathy (iMN); however, their relationships with disease severity and progression remain unclear. METHODS Patients with iMN diagnosed via renal biopsy were enrolled in this study. The concentrations of BAFF and APRIL were determined using ELISA kits. Proteinuria remission, including complete remission (CR) and partial remission (PR), and renal function deterioration were defined as clinical events. The Cox proportional hazards method was used to analyze the relationship between cytokine levels and disease progression. RESULTS Seventy iMN patients were enrolled in this study, with a median follow-up time of 24 months (range 6-72 months). The serum levels of BAFF and APRIL were higher in iMN patients than in healthy controls but lower than those in minimal change disease (MCD) patients. The serum BAFF level was positively correlated with the serum APRIL level, serum anti-phospholipase A2 receptor (anti-PLA2R) antibody level, and 24-h proteinuria and negatively correlated with the serum albumin (ALB) level. However, no significant correlation was observed between the serum APRIL level and clinical parameters. According to the multivariate Cox proportional hazards regression model adjusted for sex, age, systolic blood pressure (SBP), estimated glomerular filtration rate (eGFR), immunosuppressive agent use, 24-h proteinuria, APRIL level, and anti-PLA2R antibody, only the serum BAFF level was identified as an independent predictor of PR (HR, 0.613; 95% CI, 0.405-0.927; p = 0.021) and CR of proteinuria (HR, 0.362; 95% CI, 0.202-0.648; p < 0.001). CONCLUSIONS A high serum BAFF level is associated with severe clinical manifestations and poor disease progression in patients with iMN.
Collapse
Affiliation(s)
- Zhaohui Li
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, P.R. China
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, P.R. China
| | - Ping Chen
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, P.R. China
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, P.R. China
- Shandong Institute of Nephrology, Jinan, Shandong, P.R. China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| | - Ying Zhang
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, P.R. China
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, P.R. China
- Shandong Institute of Nephrology, Jinan, Shandong, P.R. China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| | - Juan Chen
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, P.R. China
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, P.R. China
- Shandong Institute of Nephrology, Jinan, Shandong, P.R. China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| | - Shanshan Zheng
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, P.R. China
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, P.R. China
- Shandong Institute of Nephrology, Jinan, Shandong, P.R. China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| | - Wenbin Li
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, P.R. China
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, P.R. China
- Shandong Institute of Nephrology, Jinan, Shandong, P.R. China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| | - Lijun Tang
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, P.R. China
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, P.R. China
- Shandong Institute of Nephrology, Jinan, Shandong, P.R. China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| | - Yipeng Liu
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, P.R. China
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, P.R. China
- Shandong Institute of Nephrology, Jinan, Shandong, P.R. China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| | - Na Zhao
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, P.R. China
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, P.R. China
- Shandong Institute of Nephrology, Jinan, Shandong, P.R. China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| |
Collapse
|
11
|
Cauble EL, Reynolds P, Epeldegui M, Andra SS, Magpantay L, Narasimhan S, Pulivarthi D, Von Behren J, Martinez-Maza O, Goldberg D, Spielfogel ES, Lacey JV, Wang SS. Associations between per- and poly-fluoroalkyl substance (PFAS) exposure and immune responses among women in the California Teachers study: A cross-sectional evaluation. Cytokine 2024; 184:156753. [PMID: 39299102 DOI: 10.1016/j.cyto.2024.156753] [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: 04/22/2024] [Revised: 08/19/2024] [Accepted: 09/06/2024] [Indexed: 09/22/2024]
Abstract
INTRODUCTION Per- and polyfluoroalkyl substances (PFAS) are persistent environmental contaminants that have been linked to a number of health outcomes, including those related to immune dysfunction. However, there are limited numbers of epidemiological-based studies that directly examine the association between PFAS exposure and immune responses. METHODS In this cross-sectional study nested in the California Teachers Study cohort, we measured nine PFAS analytes in serum. Of the 9 analytes, we further evaluated four (PFHxS [perfluorohexane sulfonate], PFNA [perfluorononanoic acid], PFOA [perfluorooctanoic acid], PFOS [perfluorooctanesulfonic acid]) that had detection levels of > 80 %, in relation to 16 systemic inflammatory/immune markers and corresponding immune pathways (Th1 [pro-inflammatory/macrophage activation], B-cell activation, and T-cell activation). Study participants (n = 722) were female, completed a questionnaire regarding various health measures and behaviors, and donated a blood sample between 2013-2016. The association between PFAS analytes and individual immune markers and pathways were evaluated by calculating odds ratios (OR) and 95 % confidence intervals (CI) in a logistic regression model. PFAS analytes were evaluated both as a dichotomous exposure (above or below the respective median) and as a continuous variable (per 1 unit increase [ng/mL]). RESULTS The prevalence of detecting any PFAS analyte rose with increasing age, with the highest PFAS prevalence observed among those aged 75 + years and the lowest PFAS prevalence observed among those aged 40-49 years (study participant age range: 40-95 years). Significant associations with BAFF (B-cell activating factor) levels above the median were observed among participants with elevated (defined as above the median) levels of PFHxS (OR=1.53), PFOA (OR=1.43), and PFOS (OR=1.40). Similarly, there were statistically significant associations between elevated levels of PFHxS and TNFRII (tumor necrosis factor receptor 2) levels (OR=1.78) and IL2Rα (interleukin 2 receptor subunit alpha) levels (OR=1.48). We also observed significant inverse associations between elevated PFNA and sCD14 (soluble cluster of differentiation 14) (OR=0.73). No significant associations were observed between elevated PFNA and any immune marker. Evaluation of PFAS exposures as continuous exposures in association with dichotomized cytokines were generally consistent with the dichotomized associations. CONCLUSIONS PFAS exposure was associated with altered levels of circulating inflammatory/immune markers; the associations were specific to PFAS analyte and immune marker. If validated, our results may suggest potential immune mechanisms underlying associations between the different PFAS analytes and adverse health outcomes.
Collapse
Affiliation(s)
- Emily L Cauble
- Division of Health Analytics, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Peggy Reynolds
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Marta Epeldegui
- UCLA AIDS Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Syam S Andra
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Srinivasan Narasimhan
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Divya Pulivarthi
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julie Von Behren
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | | | - Debbie Goldberg
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Emma S Spielfogel
- Division of Health Analytics, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - James V Lacey
- Division of Health Analytics, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Sophia S Wang
- Division of Health Analytics, Beckman Research Institute, City of Hope, Duarte, CA, USA.
| |
Collapse
|
12
|
Fu Y, Feng C, Qin S, Xing Z, Liu C, Liu Z, Yu H. Breaking barriers: advancing cellular therapies in autoimmune disease management. Front Immunol 2024; 15:1503099. [PMID: 39676874 PMCID: PMC11638217 DOI: 10.3389/fimmu.2024.1503099] [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: 09/28/2024] [Accepted: 11/11/2024] [Indexed: 12/17/2024] Open
Abstract
Autoimmune diseases occur due to a dysregulation within the immune system, leading to an aberrant assault on the organism's own tissues. The pathogenesis of these conditions is multifactorial, encompassing intricate interplays among genetic predispositions, environmental determinants, and hormonal fluctuations. The spectrum of autoimmune diseases is broad, impacting a multitude of organ systems, with notable examples such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), multiple sclerosis (MS), psoriasis, and vitiligo. Despite substantial progress in therapeutic interventions over recent years, a definitive cure for autoimmune diseases has yet to be realized, with existing modalities largely providing palliative care. Cellular therapy is considered the fourth pillar in the management of oncological disorders subsequent to surgical resection, radiotherapy, and chemotherapy. Cellular therapies have shown potential in augmenting immune competence and eliminating of targeted neoplastic cells in a spectrum of cancers. As targeting specific molecules on the surface of autoreactive B and T cells, such as CD19, BCMA, CD20, and CTLA-4, cellular therapies are emerging as promising approaches for the treatment of autoimmune diseases. This review delineates the advancements in the application of cellular therapies applied recently for autoimmune diseases and proposes considerations for the advancement of novel therapeutic strategies.
Collapse
Affiliation(s)
- Yanhong Fu
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
- Tianjin University and Health-Biotech United Group Joint Laboratory of Innovative Drug Development and Translational Medicine, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
| | - Chunjing Feng
- Tianjin University and Health-Biotech United Group Joint Laboratory of Innovative Drug Development and Translational Medicine, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
- Jiangxi Engineering Research Center for Stem Cell, Jiangxi Health-Biotech Stem Cell Technology Co., Ltd., Shangrao, Jiangxi, China
| | - Shan Qin
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
- Tianjin University and Health-Biotech United Group Joint Laboratory of Innovative Drug Development and Translational Medicine, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
| | - Zhiyao Xing
- Tianjin University and Health-Biotech United Group Joint Laboratory of Innovative Drug Development and Translational Medicine, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
- Jinnan Hospital, Faculty of Medicine, Tianjin Jinnan Hospital, Tianjin University, Tianjin, China
| | - Chong Liu
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
- Tianjin University and Health-Biotech United Group Joint Laboratory of Innovative Drug Development and Translational Medicine, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
| | - Zichuan Liu
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, China
| | - Hongjian Yu
- Jinnan Hospital, Faculty of Medicine, Tianjin Jinnan Hospital, Tianjin University, Tianjin, China
| |
Collapse
|
13
|
Wu H, Liao K, Li Y, Tan Z, Zhou Z, Zeng C, Gong J, Wang H, Xu H, Hu Y. Identifying the genetic association between severe autoimmune type 2 diabetes and the risk of focal epilepsy. Front Endocrinol (Lausanne) 2024; 15:1396912. [PMID: 39568813 PMCID: PMC11576724 DOI: 10.3389/fendo.2024.1396912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 10/14/2024] [Indexed: 11/22/2024] Open
Abstract
Background Observational studies suggested a bidirectional relationship between severe autoimmune type 2 diabetes and focal epilepsy. However, it remains debated whether and in which direction a causal association exists. This genetics-based study aimed to explore the relationships of severe autoimmune type 2 diabetes (T2DM) and focal epilepsy outcomes with two sample Mendelian randomization (TSMR) method. Methods Genetic instruments were obtained from large-scale genome-wide meta-analysis of severe autoimmune T2DM (Ncase = 452, Ncontrol = 2,744), and focal epilepsy (Ncase = 929, Ncontrol = 212,532) of European ancestry. A series of analyses were performed to select eligible genetic instruments robustly associated with each of the traits using summary-level statistics. Inverse variance weighted was used for primary analysis, with alternative 11 MR methods. A scatter plot was utilized to illustrate the association between single nucleotide polymorphism (SNP) effects on the exposure and SNP effects on the outcome. The Wald ratio for individual SNPs and their cumulative effects was depicted using a forest plot. And diagnostics and sensitivity analyses were used to evaluate if the causal estimates are robust to violations of MR underlying assumptions, including pleiotropy, heterogeneity assessment, and leave-one-out analysis. Then the results were validated using CURATED database of DisGeNET platform. Results For forward analysis, genetic predisposition to severe autoimmune T2DM was associated with an increased risk of focal epilepsy (Inverse variance weighted (IVW) method: OR = 1.11, 95% CI = 1.03-1.18, p = 0.012). For reverse analysis, there was no enough instrument variables of focal epilepsy on severe autoimmune T2DM. Further, the interrelation between severe autoimmune T2DM and focal epilepsy was demonstrated via variant-disease association network analysis using the instrument SNPs. Discussion This MR study supports a causal link between severe autoimmune T2DM and focal epilepsy. More effort should be made to screen seizure in severe autoimmune T2DM, unravel its clinical implications, and explore its role as a putative modifiable risk factor.
Collapse
Affiliation(s)
- Huanhua Wu
- Central Laboratory, The Affiliated Shunde Hospital of Jinan University, Foshan, Guangdong, China
| | - Kai Liao
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University and Institute of Molecular and Functional Imaging, Jinan University, Guangzhou, Guangdong, China
| | - Ying Li
- Department of Pharmacology, Medical College of Jinan University, Guangzhou, Guangdong, China
| | - Zhiqiang Tan
- Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University and Institute of Molecular and Functional Imaging, Jinan University, Guangzhou, Guangdong, China
| | - Ziqing Zhou
- Department of Nuclear Medicine, Nanhai District People's Hospital of Foshan, Foshan, Guangdong, China
| | - Chunyuan Zeng
- Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University and Institute of Molecular and Functional Imaging, Jinan University, Guangzhou, Guangdong, China
| | - Jian Gong
- Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University and Institute of Molecular and Functional Imaging, Jinan University, Guangzhou, Guangdong, China
| | - Huadong Wang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Hao Xu
- Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University and Institute of Molecular and Functional Imaging, Jinan University, Guangzhou, Guangdong, China
| | - Youzhu Hu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Veerasubramanian PK, Wynn TA, Quan J, Karlsson FJ. Targeting TNF/TNFR superfamilies in immune-mediated inflammatory diseases. J Exp Med 2024; 221:e20240806. [PMID: 39297883 PMCID: PMC11413425 DOI: 10.1084/jem.20240806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/19/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Dysregulated signaling from TNF and TNFR proteins is implicated in several immune-mediated inflammatory diseases (IMIDs). This review centers around seven IMIDs (rheumatoid arthritis, systemic lupus erythematosus, Crohn's disease, ulcerative colitis, psoriasis, atopic dermatitis, and asthma) with substantial unmet medical needs and sheds light on the signaling mechanisms, disease relevance, and evolving drug development activities for five TNF/TNFR signaling axes that garner substantial drug development interest in these focus conditions. The review also explores the current landscape of therapeutics, emphasizing the limitations of the approved biologics, and the opportunities presented by small-molecule inhibitors and combination antagonists of TNF/TNFR signaling.
Collapse
Affiliation(s)
| | - Thomas A. Wynn
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA, USA
| | - Jie Quan
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA, USA
| | | |
Collapse
|
15
|
Sharma Y, Gupta JK, Babu MA, Singh S, Sindhu RK. Signaling Pathways Concerning Mitochondrial Dysfunction: Implications in Neurodegeneration and Possible Molecular Targets. J Mol Neurosci 2024; 74:101. [PMID: 39466510 DOI: 10.1007/s12031-024-02269-5] [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: 06/03/2024] [Accepted: 09/16/2024] [Indexed: 10/30/2024]
Abstract
Mitochondrion is an important organelle present in our cells responsible for meeting energy requirements. All higher organisms rely on efficient mitochondrial bioenergetic machinery to sustain life. No other respiratory process can produce as much power as generated by mitochondria in the form of ATPs. This review is written in order to get an insight into the magnificent working of mitochondrion and its implications in cellular homeostasis, bioenergetics, redox, calcium signaling, and cell death. However, if this machinery gets faulty, it may lead to several disease states. Mitochondrial dysfunctioning is of growing concern today as it is seen in the pathogenesis of several diseases which includes neurodegenerative disorders, cardiovascular disorders, diabetes mellitus, skeletal muscle defects, liver diseases, and so on. To cover all these aspects is beyond the scope of this article; hence, our study is restricted to neurodegenerative disorders only. Moreover, faulty functioning of this organelle can be one of the causes of early ageing in individuals. This review emphasizes mutations in the mitochondrial DNA, defects in oxidative phosphorylation, generation of ROS, and apoptosis. Researchers have looked into new approaches that might be able to control mitochondrial failure and show a lot of promise as treatments.
Collapse
Affiliation(s)
- Yati Sharma
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India
| | - Jeetendra Kumar Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India
| | - Sumitra Singh
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, 125001, India
| | - Rakesh K Sindhu
- School of Pharmacy, Sharda University, Gautam Buddha Nagar, Greater Noida, Uttar Paresdh, 201310, India.
| |
Collapse
|
16
|
Vukovic J, Abazovic D, Vucetic D, Medenica S. CAR-engineered T cell therapy as an emerging strategy for treating autoimmune diseases. Front Med (Lausanne) 2024; 11:1447147. [PMID: 39450112 PMCID: PMC11500465 DOI: 10.3389/fmed.2024.1447147] [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: 06/11/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024] Open
Abstract
CAR-T therapy has demonstrated great success in treating hematological malignancies, which has led to further research into its potential in treating other diseases. Autoimmune diseases have great potential to be treated with this therapy due to the possibility of specific targeting of pathological immune cells and cells that produce autoantibodies, which could lead to permanent healing and restoration of immunological tolerance. Several approaches are currently under investigation, including targeting and depleting B cells via CD19 in the early stages of the disease, simultaneously targeting B cells and memory plasma cells in later stages and refractory states, as well as targeting specific autoantigens through the chimeric autoantibody receptor (CAAR). Additionally, CAR-engineered T regulatory cells can be modified to specifically target the autoimmune niche and modulate the pathological immune response. The encouraging results from preclinical studies have led to the first successful use of CAR-T therapy in humans to treat autoimmunity. This paved the way for further clinical studies, aiming to evaluate the long-term safety and efficacy of these therapies, potentially revolutionizing clinical use.
Collapse
Affiliation(s)
- Jovana Vukovic
- Institute for the Application of Nuclear Energy - INEP, University of Belgrade, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dzihan Abazovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dusan Vucetic
- Institute for Transfusiology and Hemobiology, Military Medical Academy, Belgrade, Serbia
| | - Sanja Medenica
- Faculty of Medicine, University of Montenegro, Podgorica, Montenegro
- Department of Endocrinology, Internal Medicine Clinic, Clinical Center of Montenegro, Podgorica, Montenegro
| |
Collapse
|
17
|
Lodde V, Zarbo IR, Farina G, Masia A, Solla P, Campesi I, Delogu G, Muroni MR, Tsitsipatis D, Gorospe M, Floris M, Idda ML. Identification of hsa_circ_0018905 as a New Potential Biomarker for Multiple Sclerosis. Cells 2024; 13:1668. [PMID: 39404430 PMCID: PMC11475351 DOI: 10.3390/cells13191668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Multiple sclerosis (MS) is a demyelinating autoimmune disease characterized by early onset, for which the interaction of genetic and environmental factors is crucial. Dysregulation of the immune system as well as myelinization-de-myelinization has been shown to correlate with changes in RNA, including non-coding RNAs. Recently, circular RNAs (circRNAs) have emerged as a key player in the complex network of gene dysregulation associated with MS. Despite several efforts, the mechanisms driving circRNA regulation and dysregulation in MS still need to be properly elucidated. Here, we explore the panorama of circRNA expression in PBMCs purified from five newly diagnosed MS patients and five healthy controls (HCs) using the Arraystar Human circRNAs microarray. Experimental validation was then carried out in a validation cohort, and a possible correlation with disease severity was tested. We identified 64 differentially expressed circRNAs, 53 of which were downregulated in PBMCs purified from MS compared to the HCs. The discovery dataset was subsequently validated using qRT-PCR with an independent cohort of 20 RRMS patients and 20 HCs. We validated seven circRNAs differentially expressed in the RRMS group versus the HC group. hsa_circ_0000518, hsa_circ_0000517, hsa_circ_0000514, and hsa_circ_0000511 were significantly upregulated in the MS group, while hsa_circ_0018905, hsa_circ_0048764, and hsa_circ_0003445 were significantly downregulated; Among them, the expression level of hsa_circ_0018905 was significantly decreased in patients showing a higher level of disability and in progressive forms of MS. We described the circRNAs expression profile of PBMCs in newly diagnosed MS patients and proposed hsa_circ_0018905 as potential MS biomarker.
Collapse
Affiliation(s)
- Valeria Lodde
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy; (V.L.); (I.C.); (G.D.); (M.F.)
| | - Ignazio Roberto Zarbo
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari 07100, Italy; (I.R.Z.); (A.M.); (P.S.); (M.R.M.)
- Unit of Clinical Neurology, AOU, Sassari 07100, Italy;
| | | | - Aurora Masia
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari 07100, Italy; (I.R.Z.); (A.M.); (P.S.); (M.R.M.)
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari 09042, Italy
| | - Paolo Solla
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari 07100, Italy; (I.R.Z.); (A.M.); (P.S.); (M.R.M.)
- Unit of Clinical Neurology, AOU, Sassari 07100, Italy;
| | - Ilaria Campesi
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy; (V.L.); (I.C.); (G.D.); (M.F.)
| | - Giuseppe Delogu
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy; (V.L.); (I.C.); (G.D.); (M.F.)
| | - Maria Rosaria Muroni
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari 07100, Italy; (I.R.Z.); (A.M.); (P.S.); (M.R.M.)
| | - Dimitrios Tsitsipatis
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA; (D.T.); (M.G.)
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA; (D.T.); (M.G.)
| | - Matteo Floris
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy; (V.L.); (I.C.); (G.D.); (M.F.)
| | - Maria Laura Idda
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy; (V.L.); (I.C.); (G.D.); (M.F.)
| |
Collapse
|
18
|
Boukouaci W, Lajnef M, Wu CL, Bouassida J, Saitoh K, Sugunasabesan S, Richard JR, Apavou M, Lamy A, Henensal A, Nkam I, Hasty L, Sayous R, Bengoufa D, Barau C, Le Corvoisier P, Honnorat J, Maskos U, Yolken R, Leboyer M, Tamouza R. B Cell-activating factor (BAFF): A promising trans-nosographic biomarker of inflammation and autoimmunity in bipolar disorder and schizophrenia. Brain Behav Immun 2024; 121:178-188. [PMID: 39047848 DOI: 10.1016/j.bbi.2024.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/27/2024] [Accepted: 07/20/2024] [Indexed: 07/27/2024] Open
Abstract
Immune dysregulation is an important aspect of schizophrenia (SZ) and bipolar disorders (BD) pathophysiology, including not only inflammatory but also autoimmune process reflective of abnormal humoral immune responses. Given that B cell-activating factor (BAFF) is an integral aspect of B lymphocyte regulation, the current study investigated BAFF in SZ and BD. 255 SZ patients, 407 BD patients and 185 healthy controls (HC) were investigated across three aspects of soluble BAFF (sBAFF) by (i) comparing sBAFF circulatory levels across SZ, BD and HC, (ii) determining potential correlations between the circulating levels of sBAFF and the genotype distribution of a functionally relevant polymorphism, namely the TNFSF13B 3'UTR insertion-deletion polymorphism (GCTGT>A), (iii) analyzing relationships between both sBAFF levels and 3'UTR insertion-deletion genotypes and disease risk, patients clinical characteristics and circulating levels of potent inflammatory molecules. In addition, in subsets of patients, we also searched for possible correlations between sBAFF levels and stigma of past infectious events as well as positivity for circulating systemic autoantibodies or those directed against central nervous system (CNS) structures. Studying blood derived serum and DNA, weobserved that circulating sBAFF levels were significantly higher in SZ and BD patients, versus HC (p = 5.3*10-10and p = 4.4*10-09). Patients experiencing acute episodes, versus stable patients, in between acute episodes, exhibited higher sBAFF levels (p = 0.017).In SZ patients, positive correlations were observed between elevated sBAFF levels and: (i) elevated positive psychotic symptoms (PANSS pos), (ii) history of childhood trauma (physical abuse), and (iii) low scores on global functioning (GAF) (p = 0.024, p = 0.024, and p = 0.041).We also found that the distribution of the BAFF Ins/Del genotypes was significantly correlated with circulating sBAFF levels in SZ and BD patients (p = 0.0004). Elevated sBAFF levels were also correlated with increased levels of pro-inflammatory markers in both SZ and BD cohorts (p < 0.001). Regarding infectious stigma, only patients seropositive, versus seronegative, for herpes simplex virus (HSV)1 immunoglobulin (Ig)G antibodies exhibited a significant association with high sBAFF levels (p = 0.013). In contrast, positivity for systemic or CNS autoantibodies was significantly associated with reduced sBAFF levels, compared to patients without autoantibodies (p = 0.0017). Overall, our findings indicate that BAFF may be a promising trans-nosographic biomarker of inflammation that is likely to offer predictive, diagnostic, and prognostic tools for the management of SZ and BD. The results therefore have practicable clinical utility given the availability of immunotherapeutic treatment options including targeted monoclonal antibodies against BAFF.
Collapse
Affiliation(s)
- Wahid Boukouaci
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Mohamed Lajnef
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Ching-Lien Wu
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Jihène Bouassida
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Kaori Saitoh
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Sobika Sugunasabesan
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Jean-Romain Richard
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Maud Apavou
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Anais Lamy
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Adèle Henensal
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Irène Nkam
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Lauren Hasty
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Romain Sayous
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Djaouida Bengoufa
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Caroline Barau
- Plateforme de Ressources Biologiques, HU Henri Mondor, AP-HP, Créteil F94010, France
| | - Philippe Le Corvoisier
- Inserm, Centre d'Investigation Clinique 1430 et AP-HP, Hôpitaux Universitaires Henri Mondor, Univ Paris Est Creteil, Créteil F-94010, France
| | - Jérome Honnorat
- French Reference Centre on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, MeLiS-UCBL-CNRS UMR 5284. INSERM U1314, University Claude Bernard Lyon 1, Lyon, France
| | - Uwe Maskos
- Institut Pasteur, Université de Paris, Integrative Neurobiology of Cholinergic Systems, CNRS UMR 3571, Paris, France
| | - Robert Yolken
- Johns Hopkins school of medicine, Baltimore, MD, USA
| | - Marion Leboyer
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Ryad Tamouza
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France.
| |
Collapse
|
19
|
Enomoto N, Nakai S, Yazawa S, Mochizuka Y, Fukada A, Tanaka Y, Naoi H, Inoue Y, Yasui H, Karayama M, Suzuki Y, Hozumi H, Furuhashi K, Toyoshima M, Kono M, Imokawa S, Fujii M, Akamatsu T, Koshimizu N, Yokomura K, Matsuda H, Kaida Y, Nakamura Y, Shirai M, Mori K, Masuda M, Fujisawa T, Inui N, Sugiura H, Sumikawa H, Kitani M, Tabata K, Ogawa N, Suda T. CXCL10 predicts autoimmune features and a favorable clinical course in patients with IIP: post hoc analysis of a prospective and multicenter cohort study. Respir Res 2024; 25:346. [PMID: 39342309 PMCID: PMC11439282 DOI: 10.1186/s12931-024-02982-0] [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: 07/16/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Interstitial pneumonia with autoimmune features (IPAF), which does not meet any of the criteria for connective tissue diseases (CTD), has been attracting an attention in patients with idiopathic interstitial pneumonia (IIP). However, the biomarkers that reflect the clinical course of these patients have not been fully elucidated. OBJECTIVE To identify useful serum biomarkers reflecting CTD-related features and favorable prognoses in patients with IIP. METHODS This was a post hoc analysis of a prospective and multicenter cohort study between 2015 and 2020. Newly diagnosed patients with IIP were consecutively enrolled, and 74 autoimmune features and autoantibodies were comprehensively checked during IIP diagnosis. Serum levels of CXCL10, CXCL1, CCL2, BAFF, angiopoietin-2, and leptin were evaluated at the time of IIP diagnosis. RESULTS Two hundred twenty-two patients (159 men and 63 women) with IIP were enrolled. The median observation duration was 36 months. The median age was 71 years old, and median %forced vital capacity (FVC) was 84.1% at the time of IIP diagnosis. The proportion of patients who met the classification criteria for IPAF was 11.7%. In patients with high serum CXCL10, changes in both %FVC and %diffusion lung capacity for carbon monoxide at one year were significantly higher than those in patients with low CXCL10 (p = 0.014 and p = 0.009, respectively), whereas these changes were not significant for other chemokines and cytokines. High CXCL10 levels were associated with acute/subacute onset (p < 0.001) and the diagnosis of nonspecific interstitial pneumonia with organizing pneumonia overlap (p = 0.003). High CXCL10 levels were related to a higher classification of IPAF (relative risk for IPAF was 3.320, 95%CI: 1.571-7.019, p = 0.003) and lower classification of progressive pulmonary fibrosis (PPF; relative risk for PPF was 0.309, 95%CI: 0.100-0.953, p = 0.027) compared to those with low CXCL10. Finally, survival was higher in patients with IPF and high CXCL10 (p = 0.044), and high CXCL10 was a significant prognostic factor in multivariate Cox proportional hazards models (hazard ratio 0.368, p = 0.005). CONCLUSIONS High serum levels of CXCL10 are associated with CTD-related features, the favorable clinical course, and survival in patients with IIP, especially IPF. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.
- Health Administration Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan.
| | - Shogo Nakai
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Shusuke Yazawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yasutaka Mochizuka
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Atsuki Fukada
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuko Tanaka
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hyogo Naoi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yusuke Inoue
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hideki Yasui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masato Karayama
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuzo Suzuki
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hironao Hozumi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuki Furuhashi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Mikio Toyoshima
- Department of Respiratory Medicine, Hamamatsu Rosai Hospital, Hamamatsu, Japan
| | - Masato Kono
- Department of Respiratory Medicine, Seirei Hamamatsu General Hospital, Hamamatsu, Japan
| | - Shiro Imokawa
- Department of Respiratory Medicine, Iwata City Hospital, Iwata, Japan
| | - Masato Fujii
- Department of Respiratory Medicine, Shizuoka City Shizuoka Hospital, Shizuoka, Japan
| | - Taisuke Akamatsu
- Department of Respiratory Medicine, Shizuoka General Hospital, Shizuoka, Japan
| | - Naoki Koshimizu
- Department of Respiratory Medicine, Fujieda Municipal General Hospital, Fujieda, Japan
| | - Koshi Yokomura
- Department of Respiratory Medicine, Respiratory Disease Center, Seirei Mikatahara General Hospital, Hamamatsu, Japan
| | - Hiroyuki Matsuda
- Department of Respiratory Medicine, Japanese Red Cross Shizuoka Hospital, Shizuoka, Japan
| | - Yusuke Kaida
- Department of Respiratory Medicine, Enshu Hospital, Hamamatsu, Japan
| | - Yutaro Nakamura
- Respiratory and Allergy Medicine, National Hospital Organization Tenryu Hospital, Hamamatsu, Japan
| | - Masahiro Shirai
- Respiratory and Allergy Medicine, National Hospital Organization Tenryu Hospital, Hamamatsu, Japan
| | - Kazutaka Mori
- Respiratory Medicine, Shizuoka City Shimizu Hospital, Shizuoka, Japan
| | - Masafumi Masuda
- Respiratory Medicine, Shizuoka City Shimizu Hospital, Shizuoka, Japan
| | - Tomoyuki Fujisawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Naoki Inui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hiroaki Sugiura
- Department of Radiology, National Defense Medical College, Saitama, Japan
| | - Hiromitsu Sumikawa
- Department of Radiology, National Hospital Organization Kinki-Chuo Chest Medical Center, Osaka, Japan
| | - Masashi Kitani
- Department of Pathology, NHO Tokyo National Hospital, Tokyo, Japan
| | - Kazuhiro Tabata
- Department of Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Noriyoshi Ogawa
- Division of Immunology and Rheumatology, Department of Internal Medicine 3, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
20
|
Hagadorn KA, Peterson ME, Kole H, Scott B, Skinner J, Diouf A, Takashima E, Ongoiba A, Doumbo S, Doumtabe D, Li S, Sekar P, Yan M, Zhu C, Nagaoka H, Kanoi BN, Li QZ, Long C, Long EO, Kayentao K, Jenks SA, Sanz I, Tsuboi T, Traore B, Bolland S, Miura K, Crompton PD, Hopp CS. Autoantibodies inhibit Plasmodium falciparum growth and are associated with protection from clinical malaria. Immunity 2024; 57:1769-1779.e4. [PMID: 38901428 PMCID: PMC11324401 DOI: 10.1016/j.immuni.2024.05.024] [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: 10/24/2023] [Revised: 02/23/2024] [Accepted: 05/30/2024] [Indexed: 06/22/2024]
Abstract
Many infections, including malaria, are associated with an increase in autoantibodies (AAbs). Prior studies have reported an association between genetic markers of susceptibility to autoimmune disease and resistance to malaria, but the underlying mechanisms are unclear. Here, we performed a longitudinal study of children and adults (n = 602) in Mali and found that high levels of plasma AAbs before the malaria season independently predicted a reduced risk of clinical malaria in children during the ensuing malaria season. Baseline AAb seroprevalence increased with age and asymptomatic Plasmodium falciparum infection. We found that AAbs purified from the plasma of protected individuals inhibit the growth of blood-stage parasites and bind P. falciparum proteins that mediate parasite invasion. Protected individuals had higher plasma immunoglobulin G (IgG) reactivity against 33 of the 123 antigens assessed in an autoantigen microarray. This study provides evidence in support of the hypothesis that a propensity toward autoimmunity offers a survival advantage against malaria.
Collapse
Affiliation(s)
- Kelly A Hagadorn
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, NIAID, NIH, Rockville, MD, USA; Yale School of Public Health, Department of Epidemiology of Microbial Diseases, New Haven, CT, USA
| | - Mary E Peterson
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, NIAID, NIH, Rockville, MD, USA
| | - Hemanta Kole
- Autoimmunity and Functional Genomics Section, Laboratory of Immunogenetics, NIAID, NIH, Rockville, MD, USA
| | - Bethany Scott
- Autoimmunity and Functional Genomics Section, Laboratory of Immunogenetics, NIAID, NIH, Rockville, MD, USA
| | - Jeff Skinner
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, NIAID, NIH, Rockville, MD, USA
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, NIAID, NIH, Rockville, MD, USA
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Aissata Ongoiba
- Malaria Research and Training Centre, Department of Epidemiology of Parasitic Diseases, International Center of Excellence in Research, University of Sciences, Technique and Technology of Bamako, Bamako, Mali
| | - Safiatou Doumbo
- Malaria Research and Training Centre, Department of Epidemiology of Parasitic Diseases, International Center of Excellence in Research, University of Sciences, Technique and Technology of Bamako, Bamako, Mali
| | - Didier Doumtabe
- Malaria Research and Training Centre, Department of Epidemiology of Parasitic Diseases, International Center of Excellence in Research, University of Sciences, Technique and Technology of Bamako, Bamako, Mali
| | - Shanping Li
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, NIAID, NIH, Rockville, MD, USA
| | - Padmapriya Sekar
- Molecular and Cellular Immunology Section, Laboratory of Immunogenetics, NIAID, NIH, Rockville, MD, USA
| | - Mei Yan
- Department of Immunology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chengsong Zhu
- Department of Immunology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Bernard N Kanoi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan; Centre for Malaria Elimination, Institute of Tropical Medicine, Mount Kenya University, Thika, Kenya
| | - Quan-Zhen Li
- Department of Immunology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA; Genecopoeia Inc, Rockville, MD, USA
| | - Carole Long
- Laboratory of Malaria and Vector Research, NIAID, NIH, Rockville, MD, USA
| | - Eric O Long
- Molecular and Cellular Immunology Section, Laboratory of Immunogenetics, NIAID, NIH, Rockville, MD, USA
| | - Kassoum Kayentao
- Malaria Research and Training Centre, Department of Epidemiology of Parasitic Diseases, International Center of Excellence in Research, University of Sciences, Technique and Technology of Bamako, Bamako, Mali
| | - Scott A Jenks
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology and Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Ignacio Sanz
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology and Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Takafumi Tsuboi
- Division of Cell-Free Sciences, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Boubacar Traore
- Malaria Research and Training Centre, Department of Epidemiology of Parasitic Diseases, International Center of Excellence in Research, University of Sciences, Technique and Technology of Bamako, Bamako, Mali
| | - Silvia Bolland
- Autoimmunity and Functional Genomics Section, Laboratory of Immunogenetics, NIAID, NIH, Rockville, MD, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, NIAID, NIH, Rockville, MD, USA
| | - Peter D Crompton
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, NIAID, NIH, Rockville, MD, USA.
| | - Christine S Hopp
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, NIAID, NIH, Rockville, MD, USA; Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
| |
Collapse
|
21
|
Cruciani C, Gatto M, Iaccarino L, Doria A, Zen M. Monoclonal antibodies targeting interleukins for systemic lupus erythematosus: updates in early clinical drug development. Expert Opin Investig Drugs 2024; 33:801-814. [PMID: 38958085 DOI: 10.1080/13543784.2024.2376566] [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: 01/21/2024] [Accepted: 07/02/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION The advent of biological therapies has already revolutionized treatment strategies and disease course of several rheumatologic conditions, and monoclonal antibodies (mAbs) targeting cytokines and interleukins represent a considerable portion of this family of drugs. In systemic lupus erythematosus (SLE) dysregulation of different cytokine and interleukin-related pathways have been linked to disease development and perpetration, offering palatable therapeutic targets addressable via such mAbs. AREAS COVERED In this review, we provide an overview of the different biological therapies under development targeting cytokines and interleukins, with a focus on mAbs, while providing the rationale behind their choice as therapeutic targets and analyzing the scientific evidence linking them to SLE pathogenesis. EXPERT OPINION An unprecedented number of clinical trials on biological drugs targeting different immunological pathways are ongoing in SLE. Their success might allow us to tackle present challenges of SLE management, including the overuse of glucocorticoids in daily clinical practice, as well as SLE heterogenicity in treatment response among different individuals, hopefully paving the way toward precision medicine.
Collapse
Affiliation(s)
- Claudio Cruciani
- Rheumatology Unit, Department of Medicine, University of Padua, Padova, Italy
| | - Mariele Gatto
- Rheumatology Unit, Department of Clinical and Biological Sciences, University of Turin and Turin Mauriziano Hospital, Turin, Italy
| | - Luca Iaccarino
- Rheumatology Unit, Department of Medicine, University of Padua, Padova, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine, University of Padua, Padova, Italy
| | - Margherita Zen
- Rheumatology Unit, Department of Medicine, University of Padua, Padova, Italy
| |
Collapse
|
22
|
Liang D, Huang S, Ding R. Effects of belimumab on the lipid profile in systemic lupus erythematosus patients: an observational study. Clin Rheumatol 2024; 43:2513-2520. [PMID: 38877375 DOI: 10.1007/s10067-024-07029-9] [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: 02/01/2024] [Revised: 05/19/2024] [Accepted: 06/09/2024] [Indexed: 06/16/2024]
Abstract
This study is asked to investigate the effects of belimumab on the lipid profile in systemic lupus erythematosus (SLE) patients. Forty-one SLE patients who received at least 6 months of belimumab treatment were retrospectively analyzed. The control group consisted of 56 age- and sex-matched lupus patients not treated with belimumab. The changes in lipid profile after a 6-month treatment were compared between the two groups. Generalized estimating equation (GEE) analyses were performed to examine lipid levels longitudinally during the period and the effect of clinical response variables and medication on the lipid profile in the belimumab group. In the belimumab group, high-density lipoprotein (HDL) cholesterol levels increased significantly after the 6-month treatment (P = 0.02). After 1 month, HDL, apolipoprotein A-I (apoA-I) significantly increased by 13.8 and 11.4%, compared with baseline, respectively. After 3 months, HDL and apoA-I increased by 9.0 and 7.1%, respectively. After 6 months, HDL increased by 7.6% compared with baseline. Total cholesterol, triglycerides, low-density lipoprotein cholesterol, and apolipoprotein B did not change significantly over the course of treatment. GEE analyses indicated a significant association between HDL and disease activity indexes, such as IgG, anti-dsDNA, and complement C3. Subgroup analysis revealed significant changes in HDL only in patients who had achieved a ≥ 4-point reduction in SLEDAI-2 K after 6 months of belimumab treatment. Belimumab treatment may result in a long-term increase in HDL level in SLE patients by improving control of lupus activity. This might have beneficial effects on controlling cardiovascular risk in lupus patients. Key Points • Treatment with belimumab resulted in a significant and sustained increase in the HDL levels in SLE patients. • Significant changes in HDL were observed in lupus patients treated with belimumab who had a better clinical response.
Collapse
Affiliation(s)
- Di Liang
- Department of Rheumatology, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shimei Huang
- Department of Rheumatology, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Rui Ding
- Department of Rheumatology, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
23
|
Calabrese M, Preziosa P, Scalfari A, Colato E, Marastoni D, Absinta M, Battaglini M, De Stefano N, Di Filippo M, Hametner S, Howell OW, Inglese M, Lassmann H, Martin R, Nicholas R, Reynolds R, Rocca MA, Tamanti A, Vercellino M, Villar LM, Filippi M, Magliozzi R. Determinants and Biomarkers of Progression Independent of Relapses in Multiple Sclerosis. Ann Neurol 2024; 96:1-20. [PMID: 38568026 DOI: 10.1002/ana.26913] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/04/2024] [Accepted: 02/15/2024] [Indexed: 06/20/2024]
Abstract
Clinical, pathological, and imaging evidence in multiple sclerosis (MS) suggests that a smoldering inflammatory activity is present from the earliest stages of the disease and underlies the progression of disability, which proceeds relentlessly and independently of clinical and radiological relapses (PIRA). The complex system of pathological events driving "chronic" worsening is likely linked with the early accumulation of compartmentalized inflammation within the central nervous system as well as insufficient repair phenomena and mitochondrial failure. These mechanisms are partially lesion-independent and differ from those causing clinical relapses and the formation of new focal demyelinating lesions; they lead to neuroaxonal dysfunction and death, myelin loss, glia alterations, and finally, a neuronal network dysfunction outweighing central nervous system (CNS) compensatory mechanisms. This review aims to provide an overview of the state of the art of neuropathological, immunological, and imaging knowledge about the mechanisms underlying the smoldering disease activity, focusing on possible early biomarkers and their translation into clinical practice. ANN NEUROL 2024;96:1-20.
Collapse
Affiliation(s)
- Massimiliano Calabrese
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Paolo Preziosa
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Antonio Scalfari
- Centre of Neuroscience, Department of Medicine, Imperial College, London, UK
| | - Elisa Colato
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Damiano Marastoni
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Martina Absinta
- Translational Neuropathology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Battaglini
- Siena Imaging S.r.l., Siena, Italy
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Massimiliano Di Filippo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Simon Hametner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Owain W Howell
- Institute of Life Sciences, Swansea University Medical School, Swansea, UK
| | - Matilde Inglese
- Dipartimento di neuroscienze, riabilitazione, oftalmologia, genetica e scienze materno-infantili - DINOGMI, University of Genova, Genoa, Italy
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Roland Martin
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Therapeutic Design Unit, Center for Molecular Medicine, Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden
- Cellerys AG, Schlieren, Switzerland
| | - Richard Nicholas
- Department of Brain Sciences, Faculty of Medicine, Burlington Danes, Imperial College London, London, UK
| | - Richard Reynolds
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Agnese Tamanti
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Marco Vercellino
- Multiple Sclerosis Center & Neurologia I U, Department of Neuroscience, University Hospital AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Luisa Maria Villar
- Department of Immunology, Ramon y Cajal University Hospital. IRYCIS. REI, Madrid, Spain
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Roberta Magliozzi
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| |
Collapse
|
24
|
Reyes-Huerta RF, Mandujano-López V, Velásquez-Ortiz MG, Alcalá-Carmona B, Ostos-Prado MJ, Reyna-Juárez Y, Meza-Sánchez DE, Juárez-Vega G, Mejía-Domínguez NR, Torres-Ruiz J, Gómez-Martín D, Maravillas-Montero JL. Novel B-cell subsets as potential biomarkers in idiopathic inflammatory myopathies: insights into disease pathogenesis and disease activity. J Leukoc Biol 2024; 116:84-94. [PMID: 38554062 DOI: 10.1093/jleuko/qiae083] [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: 10/31/2023] [Revised: 03/01/2024] [Accepted: 03/19/2024] [Indexed: 04/01/2024] Open
Abstract
Idiopathic inflammatory myopathies are a heterogeneous group of rare autoimmune disorders characterized by progressive muscle weakness and the histopathologic findings of inflammatory infiltrates in muscle tissue. Although their pathogenesis remains indefinite, the association of autoantibodies with clinical manifestations and the evidence of high effectiveness of depleting therapies suggest that B cells could be implicated. Therefore, we explored the landscape of peripheral B cells in this disease by multiparametric flow cytometry, finding significant numerical decreases in memory and double-negative subsets, as well as an expansion of the naive compartment relative to healthy controls, that contribute to defining disease-associated B-cell subset signatures and correlating with different clinical features of patients. Additionally, we determined the potential value of these subsets as diagnostic biomarkers, thus positioning B cells as neglected key elements possibly participating in idiopathic inflammatory myopathy onset or development.
Collapse
Affiliation(s)
- Raúl F Reyes-Huerta
- B cell Immunology Laboratory, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México, Circuito de la Investigación Científica S/N, CU, Coyoacán, Mexico City 04510, Mexico
- Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Unidad de Posgrado, CU, Coyoacán, Mexico City 04510, Mexico
| | - Vladimir Mandujano-López
- B cell Immunology Laboratory, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México, Circuito de la Investigación Científica S/N, CU, Coyoacán, Mexico City 04510, Mexico
| | - Ma Guadalupe Velásquez-Ortiz
- B cell Immunology Laboratory, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México, Circuito de la Investigación Científica S/N, CU, Coyoacán, Mexico City 04510, Mexico
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Unidad de Posgrado, CU, Coyoacán, Mexico City 04510, Mexico
| | - Beatriz Alcalá-Carmona
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - María J Ostos-Prado
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Yatzil Reyna-Juárez
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - David E Meza-Sánchez
- B cell Immunology Laboratory, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México, Circuito de la Investigación Científica S/N, CU, Coyoacán, Mexico City 04510, Mexico
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán y Universidad Nacional Autónoma de México, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Guillermo Juárez-Vega
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán y Universidad Nacional Autónoma de México, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Nancy R Mejía-Domínguez
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán y Universidad Nacional Autónoma de México, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Jiram Torres-Ruiz
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Diana Gómez-Martín
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán y Universidad Nacional Autónoma de México, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - José L Maravillas-Montero
- B cell Immunology Laboratory, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México, Circuito de la Investigación Científica S/N, CU, Coyoacán, Mexico City 04510, Mexico
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán y Universidad Nacional Autónoma de México, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
| |
Collapse
|
25
|
Mitul MT, Kastenschmidt JM, Sureshchandra S, Wagoner ZW, Sorn AM, Mcllwain DR, Hernandez-Davies JE, Jain A, de Assis R, Trask D, Davies DH, Wagar LE. Tissue-specific sex differences in pediatric and adult immune cell composition and function. Front Immunol 2024; 15:1373537. [PMID: 38812520 PMCID: PMC11133680 DOI: 10.3389/fimmu.2024.1373537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/26/2024] [Indexed: 05/31/2024] Open
Abstract
Sex-based differences in immune cell composition and function can contribute to distinct adaptive immune responses. Prior work has quantified these differences in peripheral blood, but little is known about sex differences within human lymphoid tissues. Here, we characterized the composition and phenotypes of adaptive immune cells from male and female ex vivo tonsils and evaluated their responses to influenza antigens using an immune organoid approach. In a pediatric cohort, female tonsils had more memory B cells compared to male tonsils direct ex vivo and after stimulation with live-attenuated but not inactivated vaccine, produced higher influenza-specific antibody responses. Sex biases were also observed in adult tonsils but were different from those measured in children. Analysis of peripheral blood immune cells from in vivo vaccinated adults also showed higher frequencies of tissue homing CD4 T cells in female participants. Together, our data demonstrate that distinct memory B and T cell profiles are present in male vs. female lymphoid tissues and peripheral blood respectively and suggest that these differences may in part explain sex biases in response to vaccines and viruses.
Collapse
Affiliation(s)
- Mahina Tabassum Mitul
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
- Institute for Immunology, University of California, Irvine, Irvine, CA, United States
- Center for Virus Research, University of California, Irvine, Irvine, CA, United States
- Vaccine Research and Development Center, University of California, Irvine, Irvine, CA, United States
| | - Jenna M. Kastenschmidt
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
- Institute for Immunology, University of California, Irvine, Irvine, CA, United States
- Center for Virus Research, University of California, Irvine, Irvine, CA, United States
- Vaccine Research and Development Center, University of California, Irvine, Irvine, CA, United States
| | - Suhas Sureshchandra
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
- Institute for Immunology, University of California, Irvine, Irvine, CA, United States
- Center for Virus Research, University of California, Irvine, Irvine, CA, United States
- Vaccine Research and Development Center, University of California, Irvine, Irvine, CA, United States
| | - Zachary W. Wagoner
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
- Institute for Immunology, University of California, Irvine, Irvine, CA, United States
- Center for Virus Research, University of California, Irvine, Irvine, CA, United States
- Vaccine Research and Development Center, University of California, Irvine, Irvine, CA, United States
| | - Andrew M. Sorn
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
- Institute for Immunology, University of California, Irvine, Irvine, CA, United States
- Center for Virus Research, University of California, Irvine, Irvine, CA, United States
- Vaccine Research and Development Center, University of California, Irvine, Irvine, CA, United States
| | - David R. Mcllwain
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, NV, United States
| | - Jenny E. Hernandez-Davies
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
- Institute for Immunology, University of California, Irvine, Irvine, CA, United States
- Center for Virus Research, University of California, Irvine, Irvine, CA, United States
- Vaccine Research and Development Center, University of California, Irvine, Irvine, CA, United States
| | - Aarti Jain
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
- Institute for Immunology, University of California, Irvine, Irvine, CA, United States
- Vaccine Research and Development Center, University of California, Irvine, Irvine, CA, United States
| | - Rafael de Assis
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
- Institute for Immunology, University of California, Irvine, Irvine, CA, United States
- Vaccine Research and Development Center, University of California, Irvine, Irvine, CA, United States
| | - Douglas Trask
- Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, CA, United States
| | - D. Huw Davies
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
- Institute for Immunology, University of California, Irvine, Irvine, CA, United States
- Center for Virus Research, University of California, Irvine, Irvine, CA, United States
- Vaccine Research and Development Center, University of California, Irvine, Irvine, CA, United States
| | - Lisa E. Wagar
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
- Institute for Immunology, University of California, Irvine, Irvine, CA, United States
- Center for Virus Research, University of California, Irvine, Irvine, CA, United States
- Vaccine Research and Development Center, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
26
|
Saadh MJ, Ahmed HM, Alani ZK, Al Zuhairi RAH, Almarhoon ZM, Ahmad H, Ubaid M, Alwan NH. The Role of Gut-derived Short-Chain Fatty Acids in Multiple Sclerosis. Neuromolecular Med 2024; 26:14. [PMID: 38630350 DOI: 10.1007/s12017-024-08783-4] [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: 02/04/2024] [Accepted: 03/08/2024] [Indexed: 04/19/2024]
Abstract
Multiple sclerosis (MS) is a chronic condition affecting the central nervous system (CNS), where the interplay of genetic and environmental factors influences its pathophysiology, triggering immune responses and instigating inflammation. Contemporary research has been notably dedicated to investigating the contributions of gut microbiota and their metabolites in modulating inflammatory reactions within the CNS. Recent recognition of the gut microbiome and dietary patterns as environmental elements impacting MS development emphasizes the potential influence of small, ubiquitous molecules from microbiota, such as short-chain fatty acids (SCFAs). These molecules may serve as vital molecular signals or metabolic substances regulating host cellular metabolism in the intricate interplay between microbiota and the host. A current emphasis lies on optimizing the health-promoting attributes of colonic bacteria to mitigate urinary tract issues through dietary management. This review aims to spotlight recent investigations on the impact of SCFAs on immune cells pivotal in MS, the involvement of gut microbiota and SCFAs in MS development, and the considerable influence of probiotics on gastrointestinal disruptions in MS. Comprehending the gut-CNS connection holds promise for the development of innovative therapeutic approaches, particularly probiotic-based supplements, for managing MS.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Hani Moslem Ahmed
- Department of Dental Industry Techniques, Al-Noor University College, Nineveh, Iraq
| | - Zaid Khalid Alani
- College of Health and Medical Technical, Al-Bayan University, Baghdad, Iraq
| | | | - Zainab M Almarhoon
- Department of Chemistry, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Hijaz Ahmad
- Section of Mathematics, International Telematic University Uninettuno, Corso Vittorio Emanuele II, 39, 00186, Rome, Italy.
- Center for Applied Mathematics and Bioinformatics, Gulf University for Science and Technology, Mubarak Al-Abdullah, Kuwait.
- Department of Computer Science and Mathematics, Lebanese American University, Beirut, Lebanon.
| | - Mohammed Ubaid
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | | |
Collapse
|
27
|
Forabosco P, Pala M, Crobu F, Diana MA, Marongiu M, Cusano R, Angius A, Steri M, Orrù V, Schlessinger D, Fiorillo E, Devoto M, Cucca F. Transcriptome organization of white blood cells through gene co-expression network analysis in a large RNA-seq dataset. Front Immunol 2024; 15:1350111. [PMID: 38629067 PMCID: PMC11018966 DOI: 10.3389/fimmu.2024.1350111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/13/2024] [Indexed: 04/19/2024] Open
Abstract
Gene co-expression network analysis enables identification of biologically meaningful clusters of co-regulated genes (modules) in an unsupervised manner. We present here the largest study conducted thus far of co-expression networks in white blood cells (WBC) based on RNA-seq data from 624 individuals. We identify 41 modules, 13 of them related to specific immune-related functions and cell types (e.g. neutrophils, B and T cells, NK cells, and plasmacytoid dendritic cells); we highlight biologically relevant lncRNAs for each annotated module of co-expressed genes. We further characterize with unprecedented resolution the modules in T cell sub-types, through the availability of 95 immune phenotypes obtained by flow cytometry in the same individuals. This study provides novel insights into the transcriptional architecture of human leukocytes, showing how network analysis can advance our understanding of coding and non-coding gene interactions in immune system cells.
Collapse
Affiliation(s)
- Paola Forabosco
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Cagliari, Italy
| | - Mauro Pala
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Cagliari, Italy
| | - Francesca Crobu
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Cagliari, Italy
| | - Maria Antonietta Diana
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Cagliari, Italy
| | - Mara Marongiu
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Cagliari, Italy
| | - Roberto Cusano
- CRS4-Next Generation Sequencing (NGS) Core, Parco POLARIS, Cagliari, Italy
| | - Andrea Angius
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Cagliari, Italy
| | - Maristella Steri
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Cagliari, Italy
| | - Valeria Orrù
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Cagliari, Italy
| | - David Schlessinger
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health (NIH), Baltimore, MA, United States
| | - Edoardo Fiorillo
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Cagliari, Italy
| | - Marcella Devoto
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Cagliari, Italy
- Dipartimento di Medicina Traslazionale e di Precisione, Università Sapienza, Roma, Italy
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Cagliari, Italy
- Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy
| |
Collapse
|
28
|
Wang AA, Luessi F, Neziraj T, Pössnecker E, Zuo M, Engel S, Hanuscheck N, Florescu A, Bugbee E, Ma XI, Rana F, Lee D, Ward LA, Kuhle J, Himbert J, Schraad M, van Puijenbroek E, Klein C, Urich E, Ramaglia V, Pröbstel AK, Zipp F, Gommerman JL. B cell depletion with anti-CD20 promotes neuroprotection in a BAFF-dependent manner in mice and humans. Sci Transl Med 2024; 16:eadi0295. [PMID: 38446903 DOI: 10.1126/scitranslmed.adi0295] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 02/13/2024] [Indexed: 03/08/2024]
Abstract
Anti-CD20 therapy to deplete B cells is highly efficacious in preventing new white matter lesions in patients with relapsing-remitting multiple sclerosis (RRMS), but its protective capacity against gray matter injury and axonal damage is unclear. In a passive experimental autoimmune encephalomyelitis (EAE) model whereby TH17 cells promote brain leptomeningeal immune cell aggregates, we found that anti-CD20 treatment effectively spared myelin content and prevented myeloid cell activation, oxidative damage, and mitochondrial stress in the subpial gray matter. Anti-CD20 treatment increased B cell survival factor (BAFF) in the serum, cerebrospinal fluid, and leptomeninges of mice with EAE. Although anti-CD20 prevented gray matter demyelination, axonal loss, and neuronal atrophy, co-treatment with anti-BAFF abrogated these benefits. Consistent with the murine studies, we observed that elevated BAFF concentrations after anti-CD20 treatment in patients with RRMS were associated with better clinical outcomes. Moreover, BAFF promoted survival of human neurons in vitro. Together, our data demonstrate that BAFF exerts beneficial functions in MS and EAE in the context of anti-CD20 treatment.
Collapse
Affiliation(s)
- Angela A Wang
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Felix Luessi
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Tradite Neziraj
- Department of Neurology, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
| | - Elisabeth Pössnecker
- Department of Neurology, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
| | - Michelle Zuo
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Sinah Engel
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Nicholas Hanuscheck
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Alexandra Florescu
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Eryn Bugbee
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Xianjie I Ma
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Fatima Rana
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Dennis Lee
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Lesley A Ward
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Jens Kuhle
- Department of Neurology, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
| | - Johannes Himbert
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Muriel Schraad
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | | | - Christian Klein
- Roche Innovation Center Zurich, Roche Glycart AG, 8952 Schlieren, Switzerland
| | - Eduard Urich
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4052 Basel, Switzerland
| | - Valeria Ramaglia
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Anne-Katrin Pröbstel
- Department of Neurology, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | | |
Collapse
|
29
|
Song Z, Li W, Han Y, Xu Y, Ding H, Wang Y. Association of immune cell traits with Parkinson's disease: a Mendelian randomization study. Front Aging Neurosci 2024; 16:1340110. [PMID: 38455666 PMCID: PMC10917892 DOI: 10.3389/fnagi.2024.1340110] [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: 11/20/2023] [Accepted: 02/12/2024] [Indexed: 03/09/2024] Open
Abstract
Background Immunity and neuroinflammation play crucial roles in the pathogenesis of Parkinson's disease (PD). Nonetheless, prior investigations into the correlation between immune inflammation and PD have produced varying results. Identifying specific immune cell phenotypes that are truly associated with PD is challenging, and the causal relationship between immune cells and PD remains elusive. Methods This study conducted a comprehensive two-sample Mendelian randomization (MR) analysis, employing five distinct analytical approaches, to clarify the causal connection between immune cell characteristics and the risk of PD. Utilizing GWAS data, we investigated the causal relationship between 731 immune cell traits and PD. These immune cell phenotypes encompass absolute cell (AC) counts, median fluorescence intensity (MFI), and relative cell (RC) counts for B cells, cDCs, mature stage T cells, monocytes, myeloid cells, TBNK (T cells, B cells, and natural killer cells), and Tregs, as well as the logistic parameter (MP) for cDCs and TBNK. Results The inverse variance weighted (IVW) analysis indicated that Myeloid DCs (p = 0.004), HVEM expression on CD45RA- CD4+ T cells (p = 0.007), CD62L- CD86+ Myeloid DCs (p = 0.015), and HLA DR expression on monocytes (p = 0.019) were associated with a reduced risk of PD. CD14+ CD16+ monocytes (p = 0.005), HLA DR+ NK cells within CD3- lymphocytes (p = 0.023), and CD28 expression on activated & secreting Tregs (p = 0.032) were associated with an increased risk of PD. Conclusion This study establishes a causal link between immune cell phenotype and the pathogenesis of PD, identifying several specific immune cell characteristics associated with PD. This could inspire researchers to delve into the pathogenesis of PD at the cellular subtype level, and aid in the identification of potential pharmacological protein targets for PD.
Collapse
Affiliation(s)
- Zhiwei Song
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Wangyu Li
- Department of Pain Management, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Yupeng Han
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Yiya Xu
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Haiqi Ding
- Department of Orthopedic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yinzhou Wang
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
- Fujian Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou, Fujian, China
| |
Collapse
|
30
|
Yu Y, Lu C, Yu W, Lei Y, Sun S, Liu P, Bai F, Chen Y, Chen J. B Cells Dynamic in Aging and the Implications of Nutritional Regulation. Nutrients 2024; 16:487. [PMID: 38398810 PMCID: PMC10893126 DOI: 10.3390/nu16040487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Aging negatively affects B cell production, resulting in a decrease in B-1 and B-2 cells and impaired antibody responses. Age-related B cell subsets contribute to inflammation. Investigating age-related alterations in the B-cell pool and developing targeted therapies are crucial for combating autoimmune diseases in the elderly. Additionally, optimal nutrition, including carbohydrates, amino acids, vitamins, and especially lipids, play a vital role in supporting immune function and mitigating the age-related decline in B cell activity. Research on the influence of lipids on B cells shows promise for improving autoimmune diseases. Understanding the aging B-cell pool and considering nutritional interventions can inform strategies for promoting healthy aging and reducing the age-related disease burden.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Juan Chen
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100091, China; (Y.Y.)
| |
Collapse
|
31
|
Huang T, Pi C, Xu X, Feng Y, Zhang J, Gu H, Fang J. Effect of BAFF blockade on the B cell receptor repertoire and transcriptome in a mouse model of systemic lupus erythematosus. Front Immunol 2024; 14:1307392. [PMID: 38264661 PMCID: PMC10803406 DOI: 10.3389/fimmu.2023.1307392] [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: 10/04/2023] [Accepted: 12/18/2023] [Indexed: 01/25/2024] Open
Abstract
Introduction Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease. Anti-B-cell-activating factor (BAFF) therapy effectively depletes B cells and reduces SLE disease activity. This research aimed to evaluate the effect of BAFF blockade on B cell receptor (BCR) repertoire and gene expression. Methods Through next-generation sequencing, we analyzed gene expression and BCR repertoire in MRL/lpr mice that received long-term anti-BAFF therapy. Based on gene expression profiles, we predicted the relative proportion of immune cells using ImmuCellAI-mouse, validating our predictions via flow cytometry and FluoroSpot. Results The loss of BCR repertoire diversity and richness, along with increased clonality and differential frequency distribution of the immunoglobulin heavy chain variable (IGHV) segment gene usage, were observed in BAFF-blockade mice. Meanwhile, the distribution of complementarity-determining region 3 (CDR3) length and CDR3 amino acid usage remained unaffected. BAFF blockade resulted in extensive changes in gene expression, particularly that of genes related to B cells and immunoglobulins. Besides, the tumor necrosis factor (TNF)-α responses and interferon (IFN)-α/γ were downregulated, consistent with the decrease in IFN-γ and TNF-α serum levels following anti-BAFF therapy. In addition, BAFF blockade significantly reduced B cell subpopulations and plasmacytoid dendritic cells, and caused the depletion of antibody-secreting cells. Discussion Our comparative BCR repertoire and transcriptome analyses of MRL/lpr mice subjected to BAFF blockade provide innovative insights into the molecular pathophysiology of SLE.
Collapse
Affiliation(s)
- Tao Huang
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Chenyu Pi
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiaoqing Xu
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yan Feng
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jingming Zhang
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Hua Gu
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jianmin Fang
- School of Life Sciences and Technology, Tongji University, Shanghai, China
- Biomedical Research Center, Tongji University Suzhou Institute, Suzhou, Jiangsu, China
- Department of Neurology, Tongji Hospital, Tongji University, Shanghai, China
| |
Collapse
|
32
|
Neri C, Ciliberti A, Dessì DA, Airoldi C, Basello K, Costanzi A, Familiari A, Tersigni C, Cappelletti M, Speciani AF, Lanzone A. B-cell-activating factor (BAFF) and platelet-activating factor (PAF) in pregnancies complicated by maternal obesity and diabetes: a preliminary study. J Matern Fetal Neonatal Med 2023; 36:2272010. [PMID: 37872771 DOI: 10.1080/14767058.2023.2272010] [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: 07/03/2023] [Accepted: 10/12/2023] [Indexed: 10/25/2023]
Abstract
OBJECTIVE In pregnancies complicated by maternal obesity and diabetes, a disruption in inflammatory mediators occurs, resulting in endothelial microvascular dysfunction, oxidative stress, tissue damage, and maternal and feto-neonatal complications. To outline this proinflammatory status, an innovative approach is represented by the measurement of proinflammatory cytokines. Among these biomarkers, B-cell-activating factor (BAFF) and platelet-activating factor (PAF) play a key role in metabolic regulation, immune response to infections, tissue homeostasis, and "food-related inflammation." The aim of the present study is to investigate the blood expression of BAFF and PAF in a cohort of pregnant women affected by obesity and diabetes compared with a control group of healthy pregnant women. METHODS A prospective longitudinal cohort study has been conducted on pregnant women referred to Fondazione Policlinico Universitario Gemelli IRCCS in Rome. For each pregnant woman, a capillary sample was collected with a swab in three different consecutive evaluations carried out in the three trimesters of pregnancy. RESULTS A total of 77 pregnant women have been enrolled. No significant differences in BAFF and PAF levels were longitudinally observed between groups. Focusing on the exposed group, in the third trimester of pregnancy, both PAF and BAFF levels were lower than the basal time. Among the selected group of patients who developed Gestational Diabetes, only PAF values were longitudinally lower when compared to other groups. The multivariate analysis showed that BAFF levels were positively correlated with thyroid-stimulating hormone levels. No macrosomia, no shoulder dystocia, no major perineal lacerations at birth, and no intrauterine growth restriction were observed in the whole population. CONCLUSIONS This study supports the involvement of metabolic and proinflammatory biomarkers in the mechanisms related to pregnancy complications. Improving a good metabolic environment for obese and diabetic pregnant women could break the vicious cycle connecting inflammation, oxidative stress, and metabolic disorders.
Collapse
Affiliation(s)
- Caterina Neri
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Alessandra Ciliberti
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Davide Archelao Dessì
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Chiara Airoldi
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | | | | | - Alessandra Familiari
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Chiara Tersigni
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | | | | | - Antonio Lanzone
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| |
Collapse
|
33
|
Skarlis C, Papadopoulos V, Raftopoulou S, Mavragani CP, Evangelopoulos ME. B-cell activating factor gene variants in multiple sclerosis: Possible associations with disease susceptibility among females. Clin Immunol 2023; 257:109847. [PMID: 37995946 DOI: 10.1016/j.clim.2023.109847] [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: 08/03/2023] [Revised: 10/24/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023]
Abstract
Although B cells and B cell activating factor (BAFF) have been previously implicated in MS pathogenesis, data regarding the genetic influence of BAFF polymorphisms on MS susceptibility are limited. Here we aim to explore whether BAFF polymorphisms could contribute to MS susceptibility. 156 RRMS patients fulfilling the revised McDonald criteria for MS diagnosis and 220 HCs were enrolled. Clinical, laboratory, and imaging characteristics were recorded. BAFF rs9514827, rs1041569, and rs9514828 polymorphisms were assessed by RFLP-PCR in DNA samples extracted from whole peripheral blood. The BAFF rs1041569 TT genotype along with the CTT and TTC haplotypes were associated with significantly increased risk for MS development in female MS patients compared to healthy female counterparts. These findings were not confirmed in males. The rs1041569 BAFF variant together with the CTT and TTC BAFF haplotypes derived from the BAFF rs9514827, rs1041569, and rs9514828 polymorphisms may represent novel genetic contributors to the development of MS in females.
Collapse
Affiliation(s)
- Charalampos Skarlis
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens (NKUA), M. Asias 75, 11527, Athens, Greece
| | - Vassilis Papadopoulos
- First Department of Neurology, School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Sylvia Raftopoulou
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens (NKUA), M. Asias 75, 11527, Athens, Greece
| | - Clio P Mavragani
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens (NKUA), M. Asias 75, 11527, Athens, Greece; Joint Academic Rheumatology Program, NKUA, Greece.
| | - Maria-Eleftheria Evangelopoulos
- First Department of Neurology, School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
34
|
Zheng A, Hu N, Xu J, Yuan Y, Zhang S, Chen W, Bai Y, Sun H. Associations between TNFSF13B polymorphisms and primary Sjögren's syndrome susceptibility in primary Sjögren's syndrome patients: A meta-analysis. Immun Inflamm Dis 2023; 11:e1103. [PMID: 38156381 PMCID: PMC10698818 DOI: 10.1002/iid3.1103] [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: 06/22/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 12/30/2023] Open
Abstract
OBJECTIVE B-cell activating factor (BAFF) is a key regulator of primary Sjögren's syndrome (pSS), which is characterized by B-lymphocyte hyperactivity. BAFF, also known as tumor necrosis factor ligand superfamily member 13B, is encoded by TNFSF13B. This study aimed to explore the possible relationships between five single-nucleotide polymorphisms (SNPs) of TNFSF13B (rs9514827, rs1041569, rs9514828, rs1224141, and rs12583006) and pSS susceptibility. METHODS We searched the following databases for articles on TNFSF13B polymorphism and pSS published up to January 2023: PubMed, Cochrane, Elsevier, Web of Science, CNKI, CQVIP, and WanFang. The odds ratios (with 95% confidence intervals) of genotypes and SNP alleles of TNFSF13B were investigated in patients with pSS to determine their relationships with pSS. RESULTS This meta-analysis employing the fixed-effect model comprised three studies of pSS patients and randomly selected healthy controls (HCs), revealing statistically significant relationships between pSS susceptibility and two SNPs: rs1041569 and rs12583006. Because rs1041569 was not in Hardy-Weinberg equilibrium in the HC group, it was eliminated from the analysis. CONCLUSIONS Polymorphisms in the BAFF (TNFSF13B) gene were related to vulnerability to pSS among pSS patients and HCs alike. The SNP rs12583006 was significantly related to pSS susceptibility in pSS patients.
Collapse
Affiliation(s)
- Anhao Zheng
- Medical Integration and Practice CenterCheeloo College of Medicine, Shandong UniversityJinanChina
- Department of Rheumatology and ImmunologyShandong Provincial Hospital, Cheeloo College of Medicine, Shandong UniversityJinanChina
| | - Naiwen Hu
- Department of Rheumatology and ImmunologyShandong Provincial Hospital, Cheeloo College of Medicine, Shandong UniversityJinanChina
- Department of Rheumatology and ImmunologyShandong Provincial Hospital, Affiliated to Shandong First Medical UniversityJinanChina
| | - Jing Xu
- Graduate SchoolShandong First Medical UniversityJinanChina
| | - Ye Yuan
- Graduate SchoolShandong First Medical UniversityJinanChina
| | - Shumin Zhang
- Graduate SchoolShandong First Medical UniversityJinanChina
| | - Wenbin Chen
- Department of Rheumatology and ImmunologyShandong Provincial Hospital, Cheeloo College of Medicine, Shandong UniversityJinanChina
- Department of Rheumatology and ImmunologyShandong Provincial Hospital, Affiliated to Shandong First Medical UniversityJinanChina
| | - Yanyan Bai
- Department of Rheumatology and ImmunologyShandong Provincial Hospital, Cheeloo College of Medicine, Shandong UniversityJinanChina
- Department of Rheumatology and ImmunologyShandong Provincial Hospital, Affiliated to Shandong First Medical UniversityJinanChina
| | - Hongsheng Sun
- Department of Rheumatology and ImmunologyShandong Provincial Hospital, Cheeloo College of Medicine, Shandong UniversityJinanChina
- Department of Rheumatology and ImmunologyShandong Provincial Hospital, Affiliated to Shandong First Medical UniversityJinanChina
| |
Collapse
|
35
|
Yeo SC, Barratt J. The contribution of a proliferation-inducing ligand (APRIL) and other TNF superfamily members in pathogenesis and progression of IgA nephropathy. Clin Kidney J 2023; 16:ii9-ii18. [PMID: 38053976 PMCID: PMC10695512 DOI: 10.1093/ckj/sfad200] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Indexed: 12/07/2023] Open
Abstract
Advances in our understanding of the pathogenesis of immunoglobulin A nephropathy (IgAN) have led to the identification of novel therapeutic targets and potential disease-specific treatments. Specifically, a proliferation-inducing ligand (APRIL) has been implicated in the pathogenesis of IgAN, mediating B-cell dysregulation and overproduction of pathogenic galactose-deficient IgA1 (Gd-IgA1). Animal and clinical studies support the involvement of APRIL in the pathogenesis and progression of IgAN. An elevated level of APRIL is found in IgAN when compared with controls, which correlates with the level of Gd-IgA1 and associates with more severe disease presentation and worse outcomes. Conversely, anti-APRIL therapy reduces pathogenic Gd-IgA1 and IgA immune complex formation and ameliorates the severity of kidney inflammation and injury. Genome-wide association studies in IgAN have identified TNFSF13 and TNFRSF13B, a cytokine ligand-receptor gene pair encoding APRIL and its receptor, respectively, as risk susceptibility loci in IgAN, further supporting the causal role of the APRIL signalling pathway in IgAN. Several novel experimental agents targeting APRIL, including atacicept, telitacicept, zigakibart and sibeprenlimab, are currently under investigation as potential therapies in IgAN. Preliminary results suggest that these agents are well-tolerated, and reduce levels of Gd-IgA1, with corresponding improvement in proteinuria. Further studies are ongoing to confirm the safety and efficacy of anti-APRIL approaches as an effective therapeutic strategy in IgAN.
Collapse
Affiliation(s)
- See Cheng Yeo
- Department of Renal Medicine, Tan Tock Seng Hospital, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Jonathan Barratt
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- John Walls Renal Unit, University Hospitals of Leicester NHS Trust, Leicester, UK
| |
Collapse
|
36
|
Gu J, Yan GM, Kong XL, Zhang YY, Huang LH, Lu HM. Assessing the causal relationship between immune traits and systemic lupus erythematosus by bi-directional Mendelian randomization analysis. Mol Genet Genomics 2023; 298:1493-1503. [PMID: 37845373 DOI: 10.1007/s00438-023-02071-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 09/23/2023] [Indexed: 10/18/2023]
Abstract
Previous studies have observed relationships between immune cells and systemic lupus erythematosus (SLE), but their causal links remain undetermined. Based on the public available genome-wide association studies (GWAS) summary statistics, we conducted two-sample Mendelian randomization (MR) to evaluate the associations between 731 immune phenotypes and SLE pairs. Pairwise pleiotropy analysis was performed to identify pleiotropic genes for significant immunophenotype-SLE pairs. A comprehensive gene function analysis was undertaken to explore the mechanisms of immune cells in SLE. By using the instrumental variables extracted from GWAS data, we observed that increased levels of five immune phenotypes were causally associated with SLE risk (FDR < 0.05), that were CD20 on IgD+ CD38- naïve, BAFF-R on IgD+ CD38dim, CD39+ secreting Treg AC, CD14- CD16+ monocyte AC, and HLA DR on CD14+ monocyte. Pairwise gene-based analyses identified a total of 38 pleiotropic genes for 5 significant pairs identified and gene set enrichment analysis revealed the involvement of the identified pleiotropic genes in complex pathways (i.e., systemic lupus erythematosus, an integral component of luminal side of endoplasmic reticulum membrane, C-type lectin receptor signaling pathway and regulation of hormone secretion). This study demonstrates that the immune response influences the progression of SLE in a complex pattern. These findings greatly improve our understanding of the interaction between immune response and SLE risk and also aid in the design of therapeutic strategies from an immunological perspective.
Collapse
Affiliation(s)
- Jie Gu
- Department of Outpatient and Emergency, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Guan-Min Yan
- Department of Outpatient and Emergency, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Xiao-Lin Kong
- Department of Outpatient and Emergency, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Yin-Ying Zhang
- Department of Outpatient and Emergency, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Li-Hong Huang
- Department of Biostatistics, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.
| | - Hui-Min Lu
- Department of Outpatient and Emergency, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China.
| |
Collapse
|
37
|
Pandey R, Bakay M, Hakonarson H. SOCS-JAK-STAT inhibitors and SOCS mimetics as treatment options for autoimmune uveitis, psoriasis, lupus, and autoimmune encephalitis. Front Immunol 2023; 14:1271102. [PMID: 38022642 PMCID: PMC10643230 DOI: 10.3389/fimmu.2023.1271102] [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: 08/01/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023] Open
Abstract
Autoimmune diseases arise from atypical immune responses that attack self-tissue epitopes, and their development is intricately connected to the disruption of the JAK-STAT signaling pathway, where SOCS proteins play crucial roles. Conditions such as autoimmune uveitis, psoriasis, lupus, and autoimmune encephalitis exhibit immune system dysfunctions associated with JAK-STAT signaling dysregulation. Emerging therapeutic strategies utilize JAK-STAT inhibitors and SOCS mimetics to modulate immune responses and alleviate autoimmune manifestations. Although more research and clinical studies are required to assess their effectiveness, safety profiles, and potential for personalized therapeutic approaches in autoimmune conditions, JAK-STAT inhibitors and SOCS mimetics show promise as potential treatment options. This review explores the action, effectiveness, safety profiles, and future prospects of JAK inhibitors and SOCS mimetics as therapeutic agents for psoriasis, autoimmune uveitis, systemic lupus erythematosus, and autoimmune encephalitis. The findings underscore the importance of investigating these targeted therapies to advance treatment options for individuals suffering from autoimmune diseases.
Collapse
Affiliation(s)
- Rahul Pandey
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Marina Bakay
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, The University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
38
|
Sun BB, Chiou J, Traylor M, Benner C, Hsu YH, Richardson TG, Surendran P, Mahajan A, Robins C, Vasquez-Grinnell SG, Hou L, Kvikstad EM, Burren OS, Davitte J, Ferber KL, Gillies CE, Hedman ÅK, Hu S, Lin T, Mikkilineni R, Pendergrass RK, Pickering C, Prins B, Baird D, Chen CY, Ward LD, Deaton AM, Welsh S, Willis CM, Lehner N, Arnold M, Wörheide MA, Suhre K, Kastenmüller G, Sethi A, Cule M, Raj A, Burkitt-Gray L, Melamud E, Black MH, Fauman EB, Howson JMM, Kang HM, McCarthy MI, Nioi P, Petrovski S, Scott RA, Smith EN, Szalma S, Waterworth DM, Mitnaul LJ, Szustakowski JD, Gibson BW, Miller MR, Whelan CD. Plasma proteomic associations with genetics and health in the UK Biobank. Nature 2023; 622:329-338. [PMID: 37794186 PMCID: PMC10567551 DOI: 10.1038/s41586-023-06592-6] [Citation(s) in RCA: 361] [Impact Index Per Article: 180.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/31/2023] [Indexed: 10/06/2023]
Abstract
The Pharma Proteomics Project is a precompetitive biopharmaceutical consortium characterizing the plasma proteomic profiles of 54,219 UK Biobank participants. Here we provide a detailed summary of this initiative, including technical and biological validations, insights into proteomic disease signatures, and prediction modelling for various demographic and health indicators. We present comprehensive protein quantitative trait locus (pQTL) mapping of 2,923 proteins that identifies 14,287 primary genetic associations, of which 81% are previously undescribed, alongside ancestry-specific pQTL mapping in non-European individuals. The study provides an updated characterization of the genetic architecture of the plasma proteome, contextualized with projected pQTL discovery rates as sample sizes and proteomic assay coverages increase over time. We offer extensive insights into trans pQTLs across multiple biological domains, highlight genetic influences on ligand-receptor interactions and pathway perturbations across a diverse collection of cytokines and complement networks, and illustrate long-range epistatic effects of ABO blood group and FUT2 secretor status on proteins with gastrointestinal tissue-enriched expression. We demonstrate the utility of these data for drug discovery by extending the genetic proxied effects of protein targets, such as PCSK9, on additional endpoints, and disentangle specific genes and proteins perturbed at loci associated with COVID-19 susceptibility. This public-private partnership provides the scientific community with an open-access proteomics resource of considerable breadth and depth to help to elucidate the biological mechanisms underlying proteo-genomic discoveries and accelerate the development of biomarkers, predictive models and therapeutics1.
Collapse
Affiliation(s)
- Benjamin B Sun
- Translational Sciences, Research & Development, Biogen, Cambridge, MA, USA.
| | - Joshua Chiou
- Internal Medicine Research Unit, Worldwide Research, Development and Medical, Pfizer, Cambridge, MA, USA
| | - Matthew Traylor
- Human Genetics Centre of Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
| | | | | | - Tom G Richardson
- Human Genetics Centre of Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
- Genomic Sciences, GlaxoSmithKline, Stevenage, UK
| | | | | | - Chloe Robins
- Genomic Sciences, GlaxoSmithKline, Collegeville, PA, USA
| | | | - Liping Hou
- Population Analytics, Janssen Research & Development, Spring House, PA, USA
| | | | - Oliver S Burren
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | - Kyle L Ferber
- Biostatistics, Research and Development, Biogen, Cambridge, MA, USA
| | | | - Åsa K Hedman
- External Science and Innovation Target Sciences, Worldwide Research, Development and Medical, Pfizer, Stockholm, Sweden
| | - Sile Hu
- Human Genetics Centre of Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Tinchi Lin
- Analytics and Data Sciences, Biogen, Cambridge, MA, USA
| | - Rajesh Mikkilineni
- Data Science Institute, Takeda Development Center Americas, Cambridge, MA, USA
| | | | | | - Bram Prins
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Denis Baird
- Translational Sciences, Research & Development, Biogen, Cambridge, MA, USA
| | - Chia-Yen Chen
- Translational Sciences, Research & Development, Biogen, Cambridge, MA, USA
| | - Lucas D Ward
- Alnylam Human Genetics, Discovery & Translational Research, Alnylam Pharmaceuticals, Cambridge, MA, USA
| | - Aimee M Deaton
- Alnylam Human Genetics, Discovery & Translational Research, Alnylam Pharmaceuticals, Cambridge, MA, USA
| | | | - Carissa M Willis
- Alnylam Human Genetics, Discovery & Translational Research, Alnylam Pharmaceuticals, Cambridge, MA, USA
| | - Nick Lehner
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Matthias Arnold
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Maria A Wörheide
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Karsten Suhre
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | | | | | - Anil Raj
- Calico Life Sciences, San Francisco, CA, USA
| | | | | | - Mary Helen Black
- Population Analytics, Janssen Research & Development, Spring House, PA, USA
| | - Eric B Fauman
- Internal Medicine Research Unit, Worldwide Research, Development and Medical, Pfizer, Cambridge, MA, USA
| | - Joanna M M Howson
- Human Genetics Centre of Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
| | | | | | - Paul Nioi
- Alnylam Human Genetics, Discovery & Translational Research, Alnylam Pharmaceuticals, Cambridge, MA, USA
| | - Slavé Petrovski
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
- Department of Medicine, University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| | | | - Erin N Smith
- Takeda Development Center Americas, San Diego, CA, USA
| | - Sándor Szalma
- Takeda Development Center Americas, San Diego, CA, USA
| | | | | | | | | | - Melissa R Miller
- Internal Medicine Research Unit, Worldwide Research, Development and Medical, Pfizer, Cambridge, MA, USA
| | - Christopher D Whelan
- Translational Sciences, Research & Development, Biogen, Cambridge, MA, USA.
- Neuroscience Data Science, Janssen Research & Development, Cambridge, MA, USA.
| |
Collapse
|
39
|
Salfi G, Casiraghi F, Remuzzi G. Current understanding of the molecular mechanisms of circulating permeability factor in focal segmental glomerulosclerosis. Front Immunol 2023; 14:1247606. [PMID: 37795085 PMCID: PMC10546017 DOI: 10.3389/fimmu.2023.1247606] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/05/2023] [Indexed: 10/06/2023] Open
Abstract
The pathogenetic mechanisms underlying the onset and the post-transplant recurrence of primary focal segmental glomerulosclerosis (FSGS) are complex and remain yet to be fully elucidated. However, a growing body of evidence emphasizes the pivotal role of the immune system in both initiating and perpetuating the disease. Extensive investigations, encompassing both experimental models and patient studies, have implicated T cells, B cells, and complement as crucial actors in the pathogenesis of primary FSGS, with various molecules being proposed as potential "circulating factors" contributing to the disease and its recurrence post kidney-transplantation. In this review, we critically assessed the existing literature to identify essential pathways for a comprehensive characterization of the pathogenesis of FSGS. Recent discoveries have shed further light on the intricate interplay between these mechanisms. We present an overview of the current understanding of the engagement of distinct molecules and immune cells in FSGS pathogenesis while highlighting critical knowledge gaps that require attention. A thorough characterization of these intricate immune mechanisms holds the potential to identify noninvasive biomarkers that can accurately identify patients at high risk of post-transplant recurrence. Such knowledge can pave the way for the development of targeted and personalized therapeutic approaches in the management of FSGS.
Collapse
Affiliation(s)
| | - Federica Casiraghi
- Istituto di Ricerche Farmacologiche Mario Negri Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Bergamo, Italy
| | | |
Collapse
|
40
|
Bogers L, Kuiper KL, Smolders J, Rip J, van Luijn MM. Epstein-Barr virus and genetic risk variants as determinants of T-bet + B cell-driven autoimmune diseases. Immunol Lett 2023; 261:66-74. [PMID: 37451321 DOI: 10.1016/j.imlet.2023.07.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 06/07/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
B cells expressing the transcription factor T-bet are found to have a protective role in viral infections, but are also considered major players in the onset of different types of autoimmune diseases. Currently, the exact mechanisms driving such 'atypical' memory B cells to contribute to protective immunity or autoimmunity are unclear. In addition to general autoimmune-related factors including sex and age, the ways T-bet+ B cells instigate autoimmune diseases may be determined by the close interplay between genetic risk variants and Epstein-Barr virus (EBV). The impact of EBV on T-bet+ B cells likely relies on the type of risk variants associated with each autoimmune disease, which may affect their differentiation, migratory routes and effector function. In this hypothesis-driven review, we discuss the lines of evidence pointing to such genetic and/or EBV-mediated influence on T-bet+ B cells in a range of autoimmune diseases, including systemic lupus erythematosus (SLE) and multiple sclerosis (MS). We provide examples of how genetic risk variants can be linked to certain signaling pathways and are differentially affected by EBV to shape T-bet+ B-cells. Finally, we propose options to improve current treatment of B cell-related autoimmune diseases by more selective targeting of pathways that are critical for pathogenic T-bet+ B-cell formation.
Collapse
Affiliation(s)
- Laurens Bogers
- MS Center ErasMS, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 80, Rotterdam 3015 CN, The Netherlands
| | - Kirsten L Kuiper
- MS Center ErasMS, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 80, Rotterdam 3015 CN, The Netherlands
| | - Joost Smolders
- MS Center ErasMS, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 80, Rotterdam 3015 CN, The Netherlands; MS Center ErasMS, Department of Neurology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 CN, The Netherlands; Netherlands Institute for Neuroscience, Neuroimmunology research group, Amsterdam 1105 BA, The Netherlands
| | - Jasper Rip
- MS Center ErasMS, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 80, Rotterdam 3015 CN, The Netherlands
| | - Marvin M van Luijn
- MS Center ErasMS, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 80, Rotterdam 3015 CN, The Netherlands.
| |
Collapse
|
41
|
Rezazadeh M, Jokar MH, Mehrnaz Aghili S, Mirfeizi Z, Mahmoudi M, Morovatdar N, Hashemzadeh K. Association between levels of serum and urinary B cell-activating factor and systemic lupus erythematosus disease activity. Arch Rheumatol 2023; 38:429-440. [PMID: 38046245 PMCID: PMC10689013 DOI: 10.46497/archrheumatol.2023.9549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 09/15/2022] [Indexed: 12/05/2023] Open
Abstract
Objectives This study investigated the correlation between serum and urinary B cell-activating factor (BAFF) levels and systemic lupus erythematosus (SLE) disease activity. Patients and methods This case-control study was conducted with 87 participants between December 2020 and September 2021. Sixty-two SLE patients who fulfilled the eligibility criteria were enrolled. SLE patients were categorized into active (n=34) and inactive (n=28) groups based on their Systemic Lupus Erythematosus Disease Activity Index 2000 (SLEDAI-2K) scores. The control group consisted of 25 healthy subjects. Serum and urine samples were collected for the measurement of BAFF levels. Finally, the relationship between these variables and SLE disease activity was investigated. Results The mean age of active (SLEDAI-2K >4) and inactive (SLEDAI-2K ≤4) SLE patients and healthy individuals were 32.8±7.8, 32.5±6.8, and 31.7±7.8 years, respectively (p=0.62). The median serum BAFF (s-BAFF) and urinary BAFF (u-BAFF) in active lupus patients (10.4 [2.3] ng/mL and 8.2 [3.7] ng/mL, respectively) were significantly higher than in inactive lupus patients (6 (7.1) ng/mL and 1.7 (4.7) ng/mL, respectively; p<0.001) and the control group (3 (3.7) ng/mL and 1.6 (2.2) ng/mL, respectively; p<0.001). However, s-BAFF (p=0.07) and u-BAFF (p=0.43) did not significantly differ between the inactive group and the control group. A significant positive correlation was observed between s-BAFF (r=0.41 and p=0.001) and u-BAFF (r=0.78 and p<0.001) levels and the SLEDAI-2K score. Conclusion There is a significant positive correlation between serum and urinary BAFF levels and SLE disease activity. Furthermore, significantly higher levels of s-BAFF and u-BAFF have been observed in patients with active lupus compared to inactive and healthy subjects, indicating a possible role for BAFF in the pathogenesis of SLE disease activity.
Collapse
Affiliation(s)
- Maryam Rezazadeh
- Mashhad University of Medical Sciences, Rheumatic Diseases Research Center, Mashhad, Iran
| | - Mohammad Hasan Jokar
- Mashhad University of Medical Sciences, Rheumatic Diseases Research Center, Mashhad, Iran
| | - Seyedeh Mehrnaz Aghili
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Zahra Mirfeizi
- Mashhad University of Medical Sciences, Rheumatic Diseases Research Center, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Mashhad University of Medical Sciences, Bu-Ali Research Institute, Mashhad, Iran
| | - Negar Morovatdar
- Mashhad University of Medical Sciences, Clinical Research Unit, Mashhad, Iran
| | - Kamila Hashemzadeh
- Mashhad University of Medical Sciences, Rheumatic Diseases Research Center, Mashhad, Iran
| |
Collapse
|
42
|
Jia X, Tan L, Chen S, Tang R, Chen W. Monogenic lupus: Tracing the therapeutic implications from single gene mutations. Clin Immunol 2023; 254:109699. [PMID: 37481012 DOI: 10.1016/j.clim.2023.109699] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/21/2023] [Accepted: 07/18/2023] [Indexed: 07/24/2023]
Abstract
Monogenic lupus, a distinctive variant of systemic lupus erythematosus (SLE), is characterized by early onset, family-centric clustering, and heightened disease severity. So far, over thirty genetic variations have been identified as single-gene etiology of SLE and lupus-like phenotypes. The critical role of these gene mutations in disrupting various immune pathways is increasingly recognized. In particular, single gene mutation-driven dysfunction within the innate immunity, notably deficiencies in the complement system, impedes the degradation of free nucleic acid and immune complexes, thereby promoting activation of innate immune cells. The accumulation of these components in various tissues and organs creates a pro-inflammatory microenvironment, characterized by a surge in pro-inflammatory cytokines, chemokines, reactive oxygen species, and type I interferons. Concurrently, single gene mutation-associated defects in the adaptive immune system give rise to the emergence of autoreactive T cells, hyperactivated B cells and plasma cells. The ensuing spectrum of cytokines and autoimmune antibodies drives systemic disease manifestations, primarily including kidney, skin and central nervous system-related phenotypes. This review provides a thorough overview of the single gene mutations and potential consequent immune dysregulations in monogenic lupus, elucidating the pathogenic mechanisms of monogenic lupus. Furthermore, it discusses the recent advances made in the therapeutic interventions for monogenic lupus.
Collapse
Affiliation(s)
- Xiuzhi Jia
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Li Tan
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Sixiu Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Ruihan Tang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China.
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China.
| |
Collapse
|
43
|
Bechara R, Vagner S, Mariette X. Post-transcriptional checkpoints in autoimmunity. Nat Rev Rheumatol 2023; 19:486-502. [PMID: 37311941 DOI: 10.1038/s41584-023-00980-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2023] [Indexed: 06/15/2023]
Abstract
Post-transcriptional regulation is a fundamental process in gene expression that has a role in diverse cellular processes, including immune responses. A core concept underlying post-transcriptional regulation is that protein abundance is not solely determined by transcript abundance. Indeed, transcription and translation are not directly coupled, and intervening steps occur between these processes, including the regulation of mRNA stability, localization and alternative splicing, which can impact protein abundance. These steps are controlled by various post-transcription factors such as RNA-binding proteins and non-coding RNAs, including microRNAs, and aberrant post-transcriptional regulation has been implicated in various pathological conditions. Indeed, studies on the pathogenesis of autoimmune and inflammatory diseases have identified various post-transcription factors as important regulators of immune cell-mediated and target effector cell-mediated pathological conditions. This Review summarizes current knowledge regarding the roles of post-transcriptional checkpoints in autoimmunity, as evidenced by studies in both haematopoietic and non-haematopoietic cells, and discusses the relevance of these findings for developing new anti-inflammatory therapies.
Collapse
Affiliation(s)
- Rami Bechara
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), Le Kremlin Bicêtre, France.
| | - Stephan Vagner
- Institut Curie, CNRS UMR3348, INSERM U1278, PSL Research University, Université Paris-Saclay, Orsay, France
| | - Xavier Mariette
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), Le Kremlin Bicêtre, France
- Assistance Publique - Hôpitaux de Paris, Hôpital Bicêtre, Department of Rheumatology, Le Kremlin Bicêtre, France
| |
Collapse
|
44
|
Gloss AD, Steiner MC, Novembre J, Bergelson J. The design of mapping populations: Impacts of geographic scale on genetic architecture and mapping efficacy for defense and immunity. CURRENT OPINION IN PLANT BIOLOGY 2023; 74:102399. [PMID: 37307746 PMCID: PMC10441534 DOI: 10.1016/j.pbi.2023.102399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 04/29/2023] [Accepted: 05/15/2023] [Indexed: 06/14/2023]
Abstract
Genome-wide association studies (GWAS) have yielded tremendous insight into the genetic architecture of trait variation. However, the collections of loci they uncover are far from exhaustive. As many of the complicating factors that confound or limit the efficacy of GWAS are exaggerated over broad geographic scales, a shift toward more analyses using mapping panels sampled from narrow geographic localities ("local" populations) could provide novel, complementary insights. Here, we present an overview of the major complicating factors, review mounting evidence from genomic analyses that these factors are pervasive, and synthesize theoretical and empirical evidence for the power of GWAS in local populations.
Collapse
Affiliation(s)
- Andrew D Gloss
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA.
| | | | - John Novembre
- Department of Human Genetics, University of Chicago, Chicago, IL, USA; Department of Ecology & Evolution, University of Chicago, Chicago, IL, USA
| | - Joy Bergelson
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA.
| |
Collapse
|
45
|
Lodde V, Floris M, Zoroddu E, Zarbo IR, Idda ML. RNA-binding proteins in autoimmunity: From genetics to molecular biology. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 14:e1772. [PMID: 36658783 DOI: 10.1002/wrna.1772] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 11/18/2022] [Accepted: 12/05/2022] [Indexed: 01/21/2023]
Abstract
Autoimmune diseases (ADs) are chronic pathologies generated by the loss of immune tolerance to the body's own cells and tissues. There is growing recognition that RNA-binding proteins (RBPs) critically govern immunity in healthy and pathological conditions by modulating gene expression post-transcriptionally at all levels: nuclear mRNA splicing and modification, export to the cytoplasm, as well as cytoplasmic mRNA transport, storage, editing, stability, and translation. Despite enormous efforts to identify new therapies for ADs, definitive solutions are not yet available in many instances. Recognizing that many ADs have a strong genetic component, we have explored connections between the molecular biology and the genetics of RBPs in ADs. Here, we review the genetics and molecular biology of RBPs in four major ADs, multiple sclerosis (MS), type 1 diabetes mellitus (T1D), systemic lupus erythematosus (SLE), and rheumatoid arthritis (RA). We anticipate that gaining insights into the genetics and biology of ADs can facilitate the discovery of new therapies. This article is categorized under: RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Valeria Lodde
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Matteo Floris
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Enrico Zoroddu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Ignazio Roberto Zarbo
- Department of Medical, Surgical and Experimental Sciences, University of Sassari - Neurology Unit Azienza Ospedaliera Universitaria (AOU), Sassari, Italy
| | - Maria Laura Idda
- Institute for Genetic and Biomedical Research - National Research Council (IRGB-CNR), Sassari, Italy
| |
Collapse
|
46
|
Ortiz GG, Torres-Mendoza BMG, Ramírez-Jirano J, Marquez-Pedroza J, Hernández-Cruz JJ, Mireles-Ramirez MA, Torres-Sánchez ED. Genetic Basis of Inflammatory Demyelinating Diseases of the Central Nervous System: Multiple Sclerosis and Neuromyelitis Optica Spectrum. Genes (Basel) 2023; 14:1319. [PMID: 37510224 PMCID: PMC10379341 DOI: 10.3390/genes14071319] [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: 05/10/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Demyelinating diseases alter myelin or the coating surrounding most nerve fibers in the central and peripheral nervous systems. The grouping of human central nervous system demyelinating disorders today includes multiple sclerosis (MS) and neuromyelitis optica spectrum disorders (NMOSD) as distinct disease categories. Each disease is caused by a complex combination of genetic and environmental variables, many involving an autoimmune response. Even though these conditions are fundamentally similar, research into genetic factors, their unique clinical manifestations, and lesion pathology has helped with differential diagnosis and disease pathogenesis knowledge. This review aims to synthesize the genetic approaches that explain the differential susceptibility between these diseases, explore the overlapping clinical features, and pathological findings, discuss existing and emerging hypotheses on the etiology of demyelination, and assess recent pathogenicity studies and their implications for human demyelination. This review presents critical information from previous studies on the disease, which asks several questions to understand the gaps in research in this field.
Collapse
Affiliation(s)
- Genaro Gabriel Ortiz
- Department of Philosophical and Methodological Disciplines and Service of Molecular Biology in Medicine Hospital, Civil University Health Sciences Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara 44329, Jalisco, Mexico
| | - Blanca M G Torres-Mendoza
- Department of Philosophical and Methodological Disciplines and Service of Molecular Biology in Medicine Hospital, Civil University Health Sciences Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Neurosciences Division, Western Biomedical Research Center, Mexican Social Security Institute (Instituto Mexicano del Seguro Social, IMSS), Guadalajara 44340, Jalisco, Mexico
| | - Javier Ramírez-Jirano
- Neurosciences Division, Western Biomedical Research Center, Mexican Social Security Institute (Instituto Mexicano del Seguro Social, IMSS), Guadalajara 44340, Jalisco, Mexico
| | - Jazmin Marquez-Pedroza
- Neurosciences Division, Western Biomedical Research Center, Mexican Social Security Institute (Instituto Mexicano del Seguro Social, IMSS), Guadalajara 44340, Jalisco, Mexico
- Coordination of Academic Activities, Western Biomedical Research Center, Mexican Social Security Institute (Instituto Mexicano del Seguro Social, IMSS), Guadalajara 44340, Jalisco, Mexico
| | - José J Hernández-Cruz
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara 44329, Jalisco, Mexico
| | - Mario A Mireles-Ramirez
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara 44329, Jalisco, Mexico
| | - Erandis D Torres-Sánchez
- Department of Medical and Life Sciences, University Center of la Cienega, University of Guadalajara, Ocotlan 47820, Jalisco, Mexico
| |
Collapse
|
47
|
Ceccarelli F, Perricone C, Natalucci F, Picciariello L, Olivieri G, Cafaro G, Bartoloni E, Roberto G, Conti F. Organ damage in Systemic Lupus Erythematosus patients: A multifactorial phenomenon. Autoimmun Rev 2023:103374. [PMID: 37301273 DOI: 10.1016/j.autrev.2023.103374] [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: 05/09/2023] [Accepted: 06/04/2023] [Indexed: 06/12/2023]
Abstract
The prevention of chronic damage, especially in early disease phases, remains an unmet need in the management of Systemic Lupus Erythematous (SLE) patients, despite the application of a so-called treat-to-target strategy. The high proportion of SLE patients developing chronic damage suggests a multifactorial aetiology. Thus, besides disease activity, other factors may contribute to the development of damage. The revision of data published so far underlines that, next to disease activity, it is possible to identify other factors playing a relevant role in damage development and progression. In summary, the presence of antiphospholipid antibodies and drugs used to treat SLE patients, in particular glucocorticoids, is strongly associated with SLE-related damage. Furthermore, recent data suggests the possible role of genetic background in determining the development of specific organ damage, in particular renal and neurological. Nonetheless, demographic factors, such as age, sex and disease duration could exert a role along with the presence of comorbidities. The contribution of different factors in determining damage development suggests the need for new outcomes to assess a comprehensive disease control including not only the assessment of disease activity, but also the evaluation of chronic damage development.
Collapse
Affiliation(s)
- Fulvia Ceccarelli
- Lupus Clinic, Division of Rheumatology, Department of Internal Clinical Sciences, Anaesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Carlo Perricone
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| | - Francesco Natalucci
- Lupus Clinic, Division of Rheumatology, Department of Internal Clinical Sciences, Anaesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Licia Picciariello
- Lupus Clinic, Division of Rheumatology, Department of Internal Clinical Sciences, Anaesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Giulio Olivieri
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy; Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, Rome, Italy
| | - Giacomo Cafaro
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Elena Bartoloni
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Gerli Roberto
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Fabrizio Conti
- Lupus Clinic, Division of Rheumatology, Department of Internal Clinical Sciences, Anaesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
48
|
Yin L, Li W, Chen X, Wang R, Zhang T, Meng J, Li Z, Xu L, Yin R, Cheng B, Yang H. HOOK1 Inhibits the Progression of Renal Cell Carcinoma via TGF-β and TNFSF13B/VEGF-A Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206955. [PMID: 37085921 PMCID: PMC10265082 DOI: 10.1002/advs.202206955] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 03/14/2023] [Indexed: 05/03/2023]
Abstract
Accumulating evidence shows HOOK1 disordered in human malignancies. However, the clinicopathological and biological significance of HOOK1 in renal cell carcinoma (RCC) remains rarely studied. In this study, the authors demonstrate that HOOK1 is downregulated in RCC samples with predicted poorer clinical prognosis. Mechanistically, HOOK1 inhibits tumor growth and metastasis via canonical TGF-β/ALK5/p-Smad3 and non-canonical TGF-β/MEK/ERK/c-Myc pathway. At the same time, HOOK1 inhibits RCC angiogenesis and sunitinib resistance by promoting degradation of TNFSF13B through the ubiquitin-proteasome pathway. In addition, HOOK1 is transcriptionally regulated by nuclear factor E2F3 in VHL dependent manner. Notably, an agonist of HOOK1, meletin, is screened and it shows antitumor activity more effectively when combined with sunitinib or nivolumab than it is used alone. The findings reveal a pivotal role of HOOK1 in anti-cancer treatment, and identify a novel therapeutic strategy for renal cell carcinoma.
Collapse
Affiliation(s)
- Lei Yin
- Department of UrologyPutuo People's HospitalTongji UniversityShanghai200060P. R. China
- Department of UrologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| | - Wenjia Li
- Department of Cardiovascular MedicineRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| | - Xuxiao Chen
- Department of General SurgeryHepatobiliary SurgeryShanghai Institute of Digestive SurgeryRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| | - Ronghao Wang
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthwest Medical UniversityLuzhou646000P. R. China
| | - Tao Zhang
- Department of UrologyPutuo People's HospitalTongji UniversityShanghai200060P. R. China
| | - Jialin Meng
- Department of UrologyThe First Affiliated Hospital of Anhui Medical UniversityAnhui Province Key Laboratory of Genitourinary DiseasesAnhui Medical UniversityHefei230032P. R. China
| | - Zhao Li
- Department of AnesthesiologyXiangya Hospital Central South UniversityChangsha410008P. R. China
| | - Li Xu
- Department of AnesthesiologyThe First People's Hospital of ChangdeChangde415000P. R. China
| | - Rui Yin
- Center for Reproductive MedicineShandong UniversityJinan250012P. R. China
| | - Bo Cheng
- Department of UrologyThe Affiliated Hospital of Southwest Medical UniversityLuzhou646000P. R. China
| | - Huan Yang
- Department of UrologyTongji HospitalTongji Medical College of Huazhong University of Science and TechnologyWuhan430030P. R. China
| |
Collapse
|
49
|
Jacobs BM, Schalk L, Dunne A, Scalfari A, Nandoskar A, Gran B, Mein CA, Sellers C, Spilker C, Rog D, Visentin E, Bezzina EL, Uzochukwu E, Tallantyre E, Wozniak E, Sacre E, Hassan-Smith G, Ford HL, Harris J, Bradley J, Breedon J, Brooke J, Kreft KL, Tuite Dalton K, George K, Papachatzaki M, O'Malley M, Peter M, Mattoscio M, Rhule N, Evangelou N, Vinod N, Quinn O, Shamji R, Kaimal R, Boulton R, Tanveer R, Middleton R, Murray R, Bellfield R, Hoque S, Patel S, Raj S, Gumus S, Mitchell S, Sawcer S, Arun T, Pogreban T, Brown TL, Begum T, Antoine V, Rashid W, Noyce AJ, Silber E, Morris H, Giovannoni G, Dobson R. ADAMS project: a genetic Association study in individuals from Diverse Ancestral backgrounds with Multiple Sclerosis based in the UK. BMJ Open 2023; 13:e071656. [PMID: 37197821 PMCID: PMC10193065 DOI: 10.1136/bmjopen-2023-071656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/14/2023] [Indexed: 05/19/2023] Open
Abstract
PURPOSE Genetic studies of multiple sclerosis (MS) susceptibility and severity have focused on populations of European ancestry. Studying MS genetics in other ancestral groups is necessary to determine the generalisability of these findings. The genetic Association study in individuals from Diverse Ancestral backgrounds with Multiple Sclerosis (ADAMS) project aims to gather genetic and phenotypic data on a large cohort of ancestrally-diverse individuals with MS living in the UK. PARTICIPANTS Adults with self-reported MS from diverse ancestral backgrounds. Recruitment is via clinical sites, online (https://app.mantal.co.uk/adams) or the UK MS Register. We are collecting demographic and phenotypic data using a baseline questionnaire and subsequent healthcare record linkage. We are collecting DNA from participants using saliva kits (Oragene-600) and genotyping using the Illumina Global Screening Array V.3. FINDINGS TO DATE As of 3 January 2023, we have recruited 682 participants (n=446 online, n=55 via sites, n=181 via the UK MS Register). Of this initial cohort, 71.2% of participants are female, with a median age of 44.9 years at recruitment. Over 60% of the cohort are non-white British, with 23.5% identifying as Asian or Asian British, 16.2% as Black, African, Caribbean or Black British and 20.9% identifying as having mixed or other backgrounds. The median age at first symptom is 28 years, and median age at diagnosis is 32 years. 76.8% have relapsing-remitting MS, and 13.5% have secondary progressive MS. FUTURE PLANS Recruitment will continue over the next 10 years. Genotyping and genetic data quality control are ongoing. Within the next 3 years, we aim to perform initial genetic analyses of susceptibility and severity with a view to replicating the findings from European-ancestry studies. In the long term, genetic data will be combined with other datasets to further cross-ancestry genetic discoveries.
Collapse
Affiliation(s)
- Benjamin M Jacobs
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Luisa Schalk
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Angie Dunne
- Leeds Centre for Neurosciences, Leeds teaching Hospitals NHS Trust, Leeds, UK
| | - Antonio Scalfari
- Centre of Neuroscience, Department of Medicine, Imperial College London, London, UK
| | | | - Bruno Gran
- Department of Neurology, Nottingham University Hospitals NHS Trust, Mental Health and Clinical Neuroscience Academic Unit, University of Nottingham School of Medicine, Nottingham, UK
| | - Charles A Mein
- Barts and the London Genome Centre, Queen Mary University of London, London, UK
| | - Charlotte Sellers
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Cord Spilker
- Bradford Teaching Hospital Foundation Trust, Bradford, UK
| | - David Rog
- Manchester Centre for Clinical Neurosciences, Northern Care Alliance NHS Trust, Manchester, UK
| | - Elisa Visentin
- Research and Innovation, Queen's Hospital, BHRUT, London, UK
| | | | - Emeka Uzochukwu
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Emma Tallantyre
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
- Department of Clinical Neurology, University Hospital of Wales, Cardiff, UK
| | - Eva Wozniak
- Barts and the London Genome Centre, Queen Mary University of London, London, UK
| | - Eve Sacre
- Leeds Centre for Neurosciences, Leeds teaching Hospitals NHS Trust, Leeds, UK
| | | | - Helen L Ford
- Leeds Centre for Neurosciences, Leeds teaching Hospitals NHS Trust, Leeds, UK
| | - Jade Harris
- Northern Care Alliance NHS Trust, Manchester, UK
| | | | - Joshua Breedon
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | | | - Karim L Kreft
- Department of Neurology, University Hospital of Wales, Cardiff, UK
| | | | - Katila George
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | | | - Martin O'Malley
- Leeds Centre for Neurosciences, Leeds teaching Hospitals NHS Trust, Leeds, UK
| | - Michelle Peter
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Miriam Mattoscio
- Department of neuroscience, Queen's Hospital, BHRUT NHS Trust, Romford, UK
| | - Neisha Rhule
- Queen Elizabeth Hospital (Lewisham and Greenwich NHS Trust), London, UK
| | - Nikos Evangelou
- Department of Neurology, Nottingham University Hospitals NHS Trust; Mental Health and Clinical Neuroscience Academic Unit, University of Nottingham School of Medicine, Nottingham, UK
| | | | - Outi Quinn
- Bradford Teaching Hospital Foundation Trust, Bradford, UK
| | - Ramya Shamji
- Research and Innovation, Queen's Hospital, BHRUT, London, UK
| | - Rashmi Kaimal
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Rebecca Boulton
- Department of Neurology, Nottingham University Hospitals NHS Trust; Mental Health and Clinical Neuroscience Academic Unit, University of Nottingham School of Medicine, Nottingham, UK
| | - Riffat Tanveer
- Lancashire Teaching Hospital NHS Foundation Trust, Preston, UK
| | - Rod Middleton
- Population Data Science, Swansea University Medical School, Swansea, UK
| | - Roxanne Murray
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Ruth Bellfield
- Bradford Teaching Hospital Foundation Trust, Bradford, UK
| | - Sadid Hoque
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Shakeelah Patel
- Lancashire Teaching Hospital NHS Foundation Trust, Preston, UK
| | - Sonia Raj
- Lancashire Teaching Hospital NHS Foundation Trust, Preston, UK
| | - Stephanie Gumus
- Mid and South Essex NHS Foundation Trust, Southend-on-Sea, UK
| | | | - Stephen Sawcer
- University of Cambridge, Department of Clinical Neuroscience, Addenbrookes Hospital, Hills Road, Cambridge, UK
| | - Tarunya Arun
- University Hospitals of Coventry and Warwickshire, Coventry, UK
| | | | - Terri-Louise Brown
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Thamanna Begum
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | | | - Waqar Rashid
- St George's University Hospitals NHS Foundation Trust, London, UK
| | - Alastair J Noyce
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Eli Silber
- Kings College Hospital and Lewisham and Greenwich NHS Trusts, London, UK
| | - Huw Morris
- Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London, UK
| | - Gavin Giovannoni
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Ruth Dobson
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| |
Collapse
|
50
|
Abstract
Autoimmune diseases display a high degree of comorbidity within individuals and families, suggesting shared risk factors. Over the past 15 years, genome-wide association studies have established the polygenic basis of these common conditions and revealed widespread sharing of genetic effects, indicative of a shared immunopathology. Despite ongoing challenges in determining the precise genes and molecular consequences of these risk variants, functional experiments and integration with multimodal genomic data are providing valuable insights into key immune cells and pathways driving these diseases, with potential therapeutic implications. Moreover, genetic studies of ancient populations are shedding light on the contribution of pathogen-driven selection pressures to the increased prevalence of autoimmune disease. This Review summarizes the current understanding of autoimmune disease genetics, including shared effects, mechanisms, and evolutionary origins.
Collapse
Affiliation(s)
- Adil Harroud
- Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada
- Department of Human Genetics, McGill University, Montréal, Quebec, Canada
- The Neuro (Montreal Neurological Institute and Hospital), McGill University, Montréal, Quebec, Canada
| | - David A Hafler
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| |
Collapse
|