1
|
Zack SR, Alzoubi O, Satoeya N, Singh KP, Deen S, Nijim W, Lewis MJ, Pitzalis C, Sweiss N, Ivashkiv LB, Shahrara S. Another Notch in the Belt of Rheumatoid Arthritis. Arthritis Rheumatol 2024; 76:1475-1487. [PMID: 38961731 PMCID: PMC11421962 DOI: 10.1002/art.42937] [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: 04/22/2024] [Revised: 05/28/2024] [Accepted: 06/13/2024] [Indexed: 07/05/2024]
Abstract
Notch ligands and receptors, including JAG1/2, DLL1/4, and Notch1/3, are enriched on macrophages (MΦs), fibroblast-like synoviocytes (FLS), and/or endothelial cells in rheumatoid arthritis (RA) compared with normal synovial tissues (ST). Power Doppler ultrasound-guided ST studies reveal that the Notch family is highly involved in early active RA, especially during neovascularization. In contrast, the Notch family is not implicated during the erosive stage, evidenced by their lack of correlation with radiographic damage in RA ST. Toll-like receptors and tumor necrosis factor (TNF) are the common inducers of Notch expression in RA MΦs, FLS, and endothelial cells. Among Notch ligands, JAG1 and/or DLL4 are most inducible by inflammatory responses in RA MΦs or endothelial cells and transactivate their receptors on RA FLS. TNF plays a central role on Notch ligands, as anti-TNF good responders display JAG1/2 and DLL1/4 transcriptional downregulation in RA ST myeloid cells. In in vitro studies, TNF increases Notch3 expression in MΦs, which is further amplified by RA FLS addition. Specific disease-modifying antirheumatic drugs reduced JAG1 and Notch3 expression in MΦ and RA FLS cocultures. Organoids containing FLS and endothelial cells have increased expression of JAG1 and Notch3. Nonetheless, Methotrexate, interleukin-6 receptor (IL-6R) antibodies, and B cell blockers are mostly ineffective at decreasing Notch family expression. NF-κB, MAPK, and AKT pathways are involved in Notch signaling, whereas JAK/STATs are not. Although Notch blockade has been effective in RA preclinical studies, its small molecule inhibitors have failed in phase I and II studies, suggesting that alternative strategies may be required to intercept their function.
Collapse
Affiliation(s)
- Stephanie R. Zack
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago; IL, USA
| | - Osama Alzoubi
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago; IL, USA
| | - Neha Satoeya
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago; IL, USA
| | - Kunwar P. Singh
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago; IL, USA
| | - Sania Deen
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago; IL, USA
| | - Wes Nijim
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago; IL, USA
| | - Myles J. Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London, United Kingdom
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, London, United Kingdom
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London, United Kingdom
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, London, United Kingdom
- Department of Biomedical Sciences, Humanitas University, and Humanitas Research Hospital, Milan, Italy
| | - Nadera Sweiss
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago; IL, USA
| | - Lionel B. Ivashkiv
- Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York 10021, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, New York 10065, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York 10065, USA
| | - Shiva Shahrara
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago; IL, USA
| |
Collapse
|
2
|
Bell RD, Brendel M, Konnaris MA, Xiang J, Otero M, Fontana MA, Bai Z, Krenitsky DM, Meednu N, Rangel-Moreno J, Scheel-Toellner D, Carr H, Nayar S, McMurray J, DiCarlo E, Anolik JH, Donlin LT, Orange DE, Kenney HM, Schwarz EM, Filer A, Ivashkiv LB, Wang F. Automated multi-scale computational pathotyping (AMSCP) of inflamed synovial tissue. Nat Commun 2024; 15:7503. [PMID: 39209814 PMCID: PMC11362542 DOI: 10.1038/s41467-024-51012-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Rheumatoid arthritis (RA) is a complex immune-mediated inflammatory disorder in which patients suffer from inflammatory-erosive arthritis. Recent advances on histopathology heterogeneity of RA synovial tissue revealed three distinct phenotypes based on cellular composition (pauci-immune, diffuse and lymphoid), suggesting that distinct etiologies warrant specific targeted therapy which motivates a need for cost effective phenotyping tools in preclinical and clinical settings. To this end, we developed an automated multi-scale computational pathotyping (AMSCP) pipeline for both human and mouse synovial tissue with two distinct components that can be leveraged together or independently: (1) segmentation of different tissue types to characterize tissue-level changes, and (2) cell type classification within each tissue compartment that assesses change across disease states. Here, we demonstrate the efficacy, efficiency, and robustness of the AMSCP pipeline as well as the ability to discover novel phenotypes. Taken together, we find AMSCP to be a valuable cost-effective method for both pre-clinical and clinical research.
Collapse
Affiliation(s)
- Richard D Bell
- Arthritis and Tissue Degeneration Program and Research Institute, Hospital for Special Surgery, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
| | - Matthew Brendel
- Department of Population Health Sciences, Weill Cornell Medical College, New York, NY, USA
| | - Maxwell A Konnaris
- Huck Institute of the Life Sciences, Pennsylvania State University, State College, University Park, PA, USA
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, NY, USA
| | | | - Miguel Otero
- Weill Cornell Medical College, New York, NY, USA
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, NY, USA
| | - Mark A Fontana
- Arthritis and Tissue Degeneration Program and Research Institute, Hospital for Special Surgery, New York, NY, USA
- Department of Population Health Sciences, Weill Cornell Medical College, New York, NY, USA
| | - Zilong Bai
- Department of Population Health Sciences, Weill Cornell Medical College, New York, NY, USA
| | - Daria M Krenitsky
- Allergy, Immunology and Rheumatology Division, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Nida Meednu
- Allergy, Immunology and Rheumatology Division, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Javier Rangel-Moreno
- Allergy, Immunology and Rheumatology Division, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Dagmar Scheel-Toellner
- Rheumatology Research Group, Institute for Inflammation and Ageing, University of Birmingham, NIHR Birmingham Biomedical Research Center and Clinical Research Facility, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Hayley Carr
- Rheumatology Research Group, Institute for Inflammation and Ageing, University of Birmingham, NIHR Birmingham Biomedical Research Center and Clinical Research Facility, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Saba Nayar
- Rheumatology Research Group, Institute for Inflammation and Ageing, University of Birmingham, NIHR Birmingham Biomedical Research Center and Clinical Research Facility, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Jack McMurray
- Rheumatology Research Group, Institute for Inflammation and Ageing, University of Birmingham, NIHR Birmingham Biomedical Research Center and Clinical Research Facility, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Edward DiCarlo
- Department of Pathology and Laboratory Medicine, Hospital for Special Surgery, New York, NY, USA
| | - Jennifer H Anolik
- Allergy, Immunology and Rheumatology Division, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Laura T Donlin
- Arthritis and Tissue Degeneration Program and Research Institute, Hospital for Special Surgery, New York, NY, USA
| | - Dana E Orange
- Arthritis and Tissue Degeneration Program and Research Institute, Hospital for Special Surgery, New York, NY, USA
- The Rockefeller University, New York, NY, USA
| | - H Mark Kenney
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Andrew Filer
- Rheumatology Research Group, Institute for Inflammation and Ageing, University of Birmingham, NIHR Birmingham Biomedical Research Center and Clinical Research Facility, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Lionel B Ivashkiv
- Arthritis and Tissue Degeneration Program and Research Institute, Hospital for Special Surgery, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Fei Wang
- Department of Population Health Sciences, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
3
|
Peng M, Yao Z, Zhang J, Lin Y, Xu L, Zhang Q, Liao J, Cai X. Discovery and validation of anti-arthritic ingredients and mechanisms of Qingfu Juanbi Tang, a Chinese herbal formulation, on rheumatoid arthritis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 329:118140. [PMID: 38565409 DOI: 10.1016/j.jep.2024.118140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 03/27/2024] [Accepted: 03/30/2024] [Indexed: 04/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qingfu Juanbi Tang (QFJBT), a novel and improved Chinese herbal formulation, has surged in recent years for its potential in the therapy of rheumatoid arthritis (RA). Anti-arthritic effects and underlying molecular mechanisms of QFJBT have increasingly become a focal point in research. AIM OF THE STUDY This study utilized network pharmacology, molecular docking, and experimental validation to elucidate effective ingredients and anti-arthritic mechanisms of QFJBT. MATERIALS AND METHODS Targets associated with QFJBT and RA were identified from relevant databases and standardized using the Uniprot for gene nomenclature. A "QFJBT-ingredient-target network" and a "Venn diagram of QFJBT and RA targets" were created from the data. The overlap in the Venn diagram highlighted potential targets of QFJBT in the treatment of RA. These targets were subjected to PPI network, GO, and KEGG pathway analysis. The findings were subsequently confirmed through molecular docking and pharmacological experiments to propose the mechanism of action of QFJBT. RESULTS The study identified 236 active ingredients in QFJBT, with 120 predicted to be effective against RA. Molecular docking showed high binding affinity of key targets (JUN, PTGS2, and TNF-α) with bioactive compounds (rhein, sinomenine, calycosin, and paeoniflorin) of QFJBT. Pharmacodynamic evaluation demonstrated the effects of QFJBT at the dose of 4.56 g/kg in ameliorating symptoms of AIA rats and in reducing levels of JUN, PTGS2, and TNF-α in synovial tissues. In vitro studies further exhibited that rhein, paeoniflorin, sinomenine, calycosin, and QFJBT-containing serum significantly inhibited abnormal proliferation of RA fibroblast-like synoviocytes. Interestingly, rhein and paeoniflorin specifically decreased p-JUN/JUN expression and TNF-α release, respectively, while sinomenine and calycosin selectively increased PTGS2 expression. Consistently, QFJBT-containing serum demonstrated similar effects as those active ingredients identified in QFJBT did. CONCLUSIONS QFJBT, QFJBT-containing serum, and its active ingredients (rhein, paeoniflorin, sinomenine, and calycosin) suppress inflammatory responses in RA. Anti-arthritic effects of QFJBT and its active ingredients are likely linked to their modulatory impact on identified hub targets.
Collapse
Affiliation(s)
- Muzi Peng
- Department of Rheumatology of First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, 410007, China; Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Zhongliu Yao
- Department of Rheumatology of First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, 410007, China
| | - Junlan Zhang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Ye Lin
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Li Xu
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Qin Zhang
- Department of Rheumatology of First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, 410007, China.
| | - Jing Liao
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Xiong Cai
- Department of Rheumatology of First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, 410007, China; Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| |
Collapse
|
4
|
Ruscitti P, Allanore Y, Baldini C, Barilaro G, Bartoloni Bocci E, Bearzi P, Bellis E, Berardicurti O, Biaggi A, Bombardieri M, Cantarini L, Cantatore FP, Caporali R, Caso F, Cervera R, Ciccia F, Cipriani P, Chatzis L, Colafrancesco S, Conti F, Corberi E, Costa L, Currado D, Cutolo M, D'Angelo S, Del Galdo F, Di Cola I, Di Donato S, Distler O, D'Onofrio B, Doria A, Fautrel B, Fasano S, Feist E, Fisher BA, Gabini M, Gandolfo S, Gatto M, Genovali I, Gerli R, Grembiale RD, Guggino G, Hoffmann-Vold AM, Iagnocco A, Iaquinta FS, Liakouli V, Manoussakis MN, Marino A, Mauro D, Montecucco C, Mosca M, Naty S, Navarini L, Occhialini D, Orefice V, Perosa F, Perricone C, Pilato A, Pitzalis C, Pontarini E, Prete M, Priori R, Rivellese F, Sarzi-Puttini P, Scarpa R, Sebastiani G, Selmi C, Shoenfeld Y, Triolo G, Trunfio F, Yan Q, Tzioufas AG, Giacomelli R. Tailoring the treatment of inflammatory rheumatic diseases by a better stratification and characterization of the clinical patient heterogeneity. Findings from a systematic literature review and experts' consensus. Autoimmun Rev 2024; 23:103581. [PMID: 39069240 DOI: 10.1016/j.autrev.2024.103581] [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: 05/12/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Inflammatory rheumatic diseases are different pathologic conditions associated with a deregulated immune response, codified along a spectrum of disorders, with autoinflammatory and autoimmune diseases as two-end phenotypes of this continuum. Despite pathogenic differences, inflammatory rheumatic diseases are commonly managed with a limited number of immunosuppressive drugs, sometimes with partial evidence or transferring physicians' knowledge in different patients. In addition, several randomized clinical trials, enrolling these patients, did not meet the primary pre-established outcomes and these findings could be linked to the underlying molecular diversities along the spectrum of inflammatory rheumatic disorders. In fact, the resulting patient heterogeneity may be driven by differences in underlying molecular pathology also resulting in variable responses to immunosuppressive drugs. Thus, the identification of different clinical subsets may possibly overcome the major obstacles that limit the development more effective therapeutic strategies for these patients with inflammatory rheumatic diseases. This clinical heterogeneity could require a diverse therapeutic management to improve patient outcomes and increase the frequency of clinical remission. Therefore, the importance of better patient stratification and characterization is increasingly pointed out according to the precision medicine principles, also suggesting a new approach for disease treatment. In fact, based on a better proposed patient profiling, clinicians could more appropriately balance the therapeutic management. On these bases, we synthetized and discussed the available literature about the patient profiling in regard to therapy in the context of inflammatory rheumatic diseases, mainly focusing on randomized clinical trials. We provided an overview of the importance of a better stratification and characterization of the clinical heterogeneity of patients with inflammatory rheumatic diseases identifying this point as crucial in improving the management of these patients.
Collapse
Affiliation(s)
- Piero Ruscitti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Yannick Allanore
- Rheumatology Department, Cochin Hospital, APHP, INSERM U1016, Université Paris Cité, Paris, France
| | - Chiara Baldini
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giuseppe Barilaro
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases, Vasculitis and Autoinflammatory Diseases of the Catalan and Spanish Health Systems, Member of ERN-ReCONNET/RITA, Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain
| | - Elena Bartoloni Bocci
- Section of Rheumatology, Department of Medicine and Surgery, University of Perugia, Italy
| | - Pietro Bearzi
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Elisa Bellis
- Academic Rheumatology Centre, Dipartimento di Scienze Cliniche e Biologiche Università di Torino - AO Mauriziano di Torino, Turin, Italy
| | - Onorina Berardicurti
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Alice Biaggi
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK
| | - Luca Cantarini
- Department of Medical Sciences, Surgery and Neurosciences, Research Center of Systemic Autoinflammatory Diseases and Behçet's Disease Clinic, University of Siena, Siena, Italy; Azienda Ospedaliero-Universitaria Senese [European Reference Network (ERN) for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) Center] Siena, Italy
| | - Francesco Paolo Cantatore
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Roberto Caporali
- Department of Clinical Sciences and Community Health, University of Milan, Paediatric Rheumatology Unit, and Clinical Rheumatology Unit, ASST Pini-CTO, Milan, Italy
| | - Francesco Caso
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Ricard Cervera
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases, Vasculitis and Autoinflammatory Diseases of the Catalan and Spanish Health Systems, Member of ERN-ReCONNET/RITA, Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain
| | - Francesco Ciccia
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Paola Cipriani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Loukas Chatzis
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Serena Colafrancesco
- Department of Internal Medicine and Medical Specialties, Rheumatology Unit, Sapienza University of Rome, Viale del Policlinico 155, 00185 Rome, Italy
| | - Fabrizio Conti
- Department of Internal Medicine and Medical Specialties, Rheumatology Unit, Sapienza University of Rome, Viale del Policlinico 155, 00185 Rome, Italy
| | - Erika Corberi
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Luisa Costa
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Damiano Currado
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Maurizio Cutolo
- Laboratory of Experimental Rheumatology and Academic Division of Rheumatology, Department of Internal Medicine and Specialties, University of Genova Italy, IRCCS Polyclinic Hospital, Genova, Italy
| | - Salvatore D'Angelo
- Rheumatology Depatment of Lucania, San Carlo Hospital of Potenza and Madonna delle Grazie Hospital of Matera, Potenza, Italy
| | - Francesco Del Galdo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Ilenia Di Cola
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Stefano Di Donato
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Bernardo D'Onofrio
- Department of Internal Medicine and Therapeutics, Università di Pavia, Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Bruno Fautrel
- Sorbonne Université - Assistance Publique Hôpitaux de Paris, INSERM UMRS 1136, Hôpital de La Pitié Salpêtrière, Paris, France
| | - Serena Fasano
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Eugen Feist
- Department of Rheumatology, Helios Fachklinik, Sophie-von-Boetticher-Straße 1, 39245, Vogelsang-Gommern, Germany; Charité - Universitätsmedizin Berlin, Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Berlin, Germany
| | - Benjamin A Fisher
- Institute of Inflammation and Ageing, University Hospitals Birmingham, Birmingham, UK; Department of Rheumatology, National Institute for Health Research (NIHR), Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Marco Gabini
- Rheumatology Unit, Santo Spirito Hospital, Pescara, Italy
| | - Saviana Gandolfo
- Unit of Rheumatology, San Giovanni Bosco Hospital, Naples, Italy
| | - Mariele Gatto
- Academic Rheumatology Centre, Dipartimento di Scienze Cliniche e Biologiche Università di Torino - AO Mauriziano di Torino, Turin, Italy
| | - Irene Genovali
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Roberto Gerli
- Section of Rheumatology, Department of Medicine and Surgery, University of Perugia, Italy
| | - Rosa Daniela Grembiale
- Rheumatology Research Unit, Dipartimento di Scienze della Salute, Università degli studi "Magna Graecia" di Catanzaro, Catanzaro, Italy
| | - Giuliana Guggino
- Rheumatology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Italy
| | - Anna Maria Hoffmann-Vold
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Department of Rheumatology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Annamaria Iagnocco
- Academic Rheumatology Centre, Dipartimento di Scienze Cliniche e Biologiche Università di Torino - AO Mauriziano di Torino, Turin, Italy
| | - Francesco Salvatore Iaquinta
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK
| | - Vasiliki Liakouli
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Menelaos N Manoussakis
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Annalisa Marino
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Daniele Mauro
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Carlomaurizio Montecucco
- Department of Internal Medicine and Therapeutics, Università di Pavia, Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marta Mosca
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Saverio Naty
- Department of Health Sciences, "Magna Græcia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Luca Navarini
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Daniele Occhialini
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine (DIM), University of Bari Medical School, Italy
| | - Valeria Orefice
- Rheumatology Unit, San Camillo-Forlanini Hospital, Rome, Italy
| | - Federico Perosa
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine (DIM), University of Bari Medical School, Italy
| | - Carlo Perricone
- Section of Rheumatology, Department of Medicine and Surgery, University of Perugia, Italy
| | - Andrea Pilato
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK; Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Elena Pontarini
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK
| | - Marcella Prete
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine (DIM), University of Bari Medical School, Italy
| | - Roberta Priori
- Department of Internal Medicine and Medical Specialties, Rheumatology Unit, Sapienza University of Rome, Viale del Policlinico 155, 00185 Rome, Italy
| | - Felice Rivellese
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK
| | - Piercarlo Sarzi-Puttini
- Rheumatology Department, ASST Fatebenefratelli Luigi Sacco University Hospital, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Raffaele Scarpa
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | | | - Carlo Selmi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Yehuda Shoenfeld
- Zabludovwicz autoimmunity center, Sheba medical center, Tel Hashomer Israel, Reichman University, Herzeliya, Israel
| | - Giovanni Triolo
- Rheumatology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Italy
| | - Francesca Trunfio
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Qingran Yan
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Athanasios G Tzioufas
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Roberto Giacomelli
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| |
Collapse
|
5
|
Bykerk VP. Clinical implications of synovial tissue phenotypes in rheumatoid arthritis. Front Med (Lausanne) 2024; 10:1093348. [PMID: 38841268 PMCID: PMC11150787 DOI: 10.3389/fmed.2023.1093348] [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: 11/08/2022] [Accepted: 10/12/2023] [Indexed: 06/07/2024] Open
Abstract
Autoimmune forms of inflammatory arthritis, such as Rheumatoid Arthritis (RA), are clinically heterogeneous in presentation and disease course. Treatment-related outcomes vary despite patient exposure to similar treatment strategies. It is likely that variation seen in synovial pathogenesis influences outcomes and is heterogeneous outcomes influenced by patient factors, including environmental exposures, microbiota, behaviors, timely access to therapy, and synovial cell variation. Patients' unique complex factors manifest as specific synovial phenotypes characterized by clusters of synovial cell types and states. Precision medicine aims to use such clinical and biological data to identify the right treatment for the right patient at the right time, enabling patients to achieve sustained remission. Identifying synovial targets susceptible to a given treatment, enabling the choice of effective therapy for a given patient, will realize the goals of precision medicine. Over the last 7 years, improved acquisition and processing of synovial tissue obtained by ultra-sound guided biopsy has enabled researchers to define synovial pathotypes using histologic features and predominant cell types associated with clinical manifestations. Technical advances have enabled single-cell simultaneous sequencing of proteins and gene expression that, through increasingly sophisticated bioinformatics methods, have taken transcriptional and proteomic data to identify diverse and novel cell types and states that cluster in the RA synovium to further define patient subgroups. Synovial pathotypes and endotypes are now integrated into clinical studies and trials to explain clinical heterogeneity in disease course and treatment response. Rapidly evolving clinical-translational research has linked an expanded understanding of RA synovial pathogenesis with clinically meaningful subgroups and treatment outcomes and the clinical heterogeneity in RA.
Collapse
|
6
|
Zack SR, Meyer A, Zanotti B, Volin MV, Deen S, Satoeya N, Sweiss N, Lewis MJ, Pitzalis C, Kitajewski JK, Shahrara S. Notch ligands are biomarkers of anti-TNF response in RA patients. Angiogenesis 2024; 27:273-283. [PMID: 37796367 PMCID: PMC10995106 DOI: 10.1007/s10456-023-09897-2] [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: 07/21/2023] [Accepted: 09/05/2023] [Indexed: 10/06/2023]
Abstract
Notch and its ligands play a critical role in rheumatoid arthritis (RA) pathogenesis. Hence, studies were conducted to delineate the functional significance of the Notch pathway in RA synovial tissue (ST) cells and the influence of RA therapies on their expression. Morphological studies reveal that JAG1, DLL4, and Notch1 are highly enriched in RA ST lining and sublining CD68+CD14+ MΦs. JAG1 and DLL4 transcription is jointly upregulated in RA MΦs reprogrammed by TLR4/5 ligation and TNF, whereas Syntenin-1 exposure expands JAG1, DLL4, and Notch1 expression levels in these cells. Single-cell RNA-seq data exhibit that JAG1 and Notch3 are overexpressed on all fibroblast-like synoviocyte (FLS) subpopulations, in parallel, JAG2, DLL1, and Notch1 expression levels are modest on RA FLS and are predominately potentiated by TLR4 ligation. Intriguingly, JAG1, DLL1/4, and Notch1/3 are presented on RA endothelial cells, and their expression is mutually reconfigured by TLR4/5 ligation in the endothelium. Synovial JAG1/JAG2/DLL1 or Notch1/3 transcriptomes were unchanged in patients who received disease-modifying anti-rheumatic drugs (DMARDs) or IL-6R Ab therapy regardless of disease activity score. Uniquely, RA MΦs and endothelial cells rewired by IL-6 displayed DLL4 transcriptional upregulation, and IL-6R antibody treatment disrupted RA ST DLL4 transcription in good responders compared to non-responders or moderate responders. Nevertheless, the JAG1/JAG2/DLL1/DLL4 transcriptome was diminished in anti-TNF good responders with myeloid pathotype and was unaltered in the fibroid pathotype except for DLL4. Taken together, our findings suggest that RA myeloid Notch ligands can serve as markers for anti-TNF responsiveness and trans-activate Notch receptors expressed on RA FLS and/or endothelial cells.
Collapse
Affiliation(s)
- Stephanie R Zack
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Anja Meyer
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Brian Zanotti
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, USA
| | - Michael V Volin
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, USA
| | - Sania Deen
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Neha Satoeya
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Nadera Sweiss
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Myles J Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, London, UK
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, London, UK
- Department of Biomedical Sciences, Humanitas University, and Humanitas Research Hospital, Milan, Italy
| | - Jan K Kitajewski
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL, 60612, USA
- University of Illinois Cancer Center, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Shiva Shahrara
- Jesse Brown VA Medical Center, Chicago, IL, USA.
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
7
|
Gentileschi S, Gaggiano C, Damiani A, Coccia C, Bernardini P, Cazzato M, D'Alessandro F, Vallifuoco G, Terribili R, Bardelli M, Baldi C, Cantarini L, Mosca M, Frediani B, Guiducci S. Impact of age and cardiovascular risk factors on the incidence of adverse events in patients with rheumatoid arthritis treated with Janus Kinase inhibitors: data from a real-life multicentric cohort. Clin Exp Med 2024; 24:62. [PMID: 38554250 PMCID: PMC10981583 DOI: 10.1007/s10238-024-01325-z] [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: 01/08/2024] [Accepted: 03/06/2024] [Indexed: 04/01/2024]
Abstract
Inhibiting Janus Kinases (JAK) is a crucial therapeutic strategy in rheumatoid arthritis (RA). However, the use of JAK inhibitors has recently raised serious safety concerns. The study aims to evaluate the safety profile of JAKi in patients with RA and identify potential risk factors (RFs) for adverse events (AEs). Data of RA patients treated with JAKi in three Italian centers from January 2017 to December 2022 were retrospectively analyzed. 182 subjects (F:117, 64.3%) underwent 193 treatment courses. 78.6% had at least one RF, including age ≥ 65 years, obesity, smoking habit, hypertension, dyslipidemia, hyperuricemia, diabetes, previous VTE or cancer, and severe mobility impairment. We identified 70 AEs (28/100 patients/year), among which 15 were serious (6/100 patients/year). A high disease activity was associated with AEs occurrence (p = 0.03 for CDAI at T0 and T6; p = 0.04 for SDAI at T0 and T6; p = 0.01 and p = 0.04 for DAS28ESR at T6 and T12, respectively). No significant differences in AEs occurrence were observed after stratification by JAKi molecules (p = 0.44), age groups (p = 0.08) nor presence of RFs (p > 0.05 for all of them). Neither the presence of any RFs, nor the cumulative number of RFs shown by the patient, nor age ≥ 65 did predict AEs occurrence. Although limited by the small sample size and the limited number of cardiovascular events, our data do not support the correlation between cardiovascular RFs-including age-and a higher incidence of AEs during JAKi therapy. The role of uncontrolled disease activity in AEs occurrence should by emphasized.
Collapse
Affiliation(s)
- Stefano Gentileschi
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, Azienda Ospedaliero-Univeristaria Senese, University of Siena, Siena, Italy.
| | - Carla Gaggiano
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, Azienda Ospedaliero-Univeristaria Senese, University of Siena, Siena, Italy
| | - Arianna Damiani
- Division of Rheumatology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Carmela Coccia
- Division of Rheumatology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Pamela Bernardini
- Division of Rheumatology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Massimiliano Cazzato
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesco D'Alessandro
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giulia Vallifuoco
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, Azienda Ospedaliero-Univeristaria Senese, University of Siena, Siena, Italy
| | - Riccardo Terribili
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, Azienda Ospedaliero-Univeristaria Senese, University of Siena, Siena, Italy
| | - Marco Bardelli
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, Azienda Ospedaliero-Univeristaria Senese, University of Siena, Siena, Italy
| | - Caterina Baldi
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, Azienda Ospedaliero-Univeristaria Senese, University of Siena, Siena, Italy
| | - Luca Cantarini
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, Azienda Ospedaliero-Univeristaria Senese, University of Siena, Siena, Italy
| | - Marta Mosca
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Bruno Frediani
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, Azienda Ospedaliero-Univeristaria Senese, University of Siena, Siena, Italy
| | - Serena Guiducci
- Division of Rheumatology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
8
|
Liu S, Liu M, Xiu J, Zhang T, Zhang B, Cun D, Yang C, Li K, Zhang J, Zhao X. Celastrol-loaded bovine serum albumin nanoparticles target inflamed neutrophils for improved rheumatoid arthritis therapy. Acta Biomater 2024; 174:345-357. [PMID: 38013018 DOI: 10.1016/j.actbio.2023.11.028] [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: 07/30/2023] [Revised: 10/25/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023]
Abstract
Inflammatory neutrophils (INEs), motivated by cytokines, continue to migrate into the inflamed joints, driving the development of RA. Hence, inducing apoptosis of INEs to reduce recruitment at inflamed joints is an effective strategy for the treatment of RA. However, simply apoptotic INEs may trigger the release of neutrophil extracellular traps (NETs) and accelerate the inflammatory process. To overcome these drawbacks, an RGD-modified bovine serum albumin (BSA) nanoparticles (CBR NPs) was fabricated to selectively target INEs in situ for intracellular delivery of CLT. Studies have demonstrated that CBR NPs can selectively target circulating INEs and induce INEs apoptosis. Meanwhile, CBR NPs inhibited the activation of NETs via NF-κB pathway and the release of Cit-H3 thereby blocking the release process of NETs. In collagen-induced arthritis (CIA) mouse model, CBR NPs suppressed the inflammatory response, and reduced the toxic effects of CLT. In summary, this study shed light on an innovative approach to treat RA by inducing apoptosis of circulating INEs and inhibiting NETs. STATEMENT OF SIGNIFICANCE: RGD-modified bovine serum albumin (BSA) nanoparticles for delivering celastrol, abbreviated as CBR NPs, were constructed to inhibit the infiltration of circulating inflammatory neutrophils (INEs) into inflamed joints while inhibiting the release of NETs to alleviate tissue damage. CBR NPs were prepared for the first time to induce apoptosis of INEs; CBR NPs could inhibit the release of NETs while inducing apoptosis of INEs in vivo and vitro cellular experiments; CBR NPs had favorable anti-inflammatory effects and low toxicity side-effects in collagen-induced arthritis (CIA) mouse models. The application of nanotechnology to induce apoptosis of INEs while inhibiting the release of NETs was a promising approach for the treatment of RA.
Collapse
Affiliation(s)
- Siyi Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110000, PR China
| | - Min Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110000, PR China
| | - Jingya Xiu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110000, PR China
| | - Tian Zhang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110000, PR China
| | - Bowen Zhang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110000, PR China
| | - Dongyun Cun
- Department of Hepatopancreatobiliary Surgery, The Second Afliated Hospital of Kunming Medical University, Kunming 650101, PR China
| | - Chunrong Yang
- Department of Pharmacy, Shantou University Medical College, Shantou 515000, PR China
| | - Kexin Li
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110000, PR China
| | - Jiulong Zhang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110000, PR China.
| | - Xiuli Zhao
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110000, PR China.
| |
Collapse
|
9
|
Garaffoni C, Tamussin M, Calciolari I, Lanza G, Bortoluzzi A, Scirè CA, Govoni M, Silvagni E. High-grade synovitis associates with clinical markers and response to therapy in chronic inflammatory arthritis: post hoc analysis of a synovial biomarkers prospective cohort study. Front Immunol 2024; 14:1298583. [PMID: 38274811 PMCID: PMC10808827 DOI: 10.3389/fimmu.2023.1298583] [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: 09/25/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Background Inflammatory arthritis (IAs), such as rheumatoid arthritis (RA) and psoriatic arthritis (PsA), are characterized by the presence of chronic synovitis. The Krenn's synovitis score (KSS), a simple tool detectable by haematoxylin/eosin staining of synovial biopsy samples, allows the discrimination between high-grade and low-grade synovitis. The aim of this study was to identify the clinical associations of KSS and to evaluate the relationship between high-grade synovitis and treatment response in IA patients. Methods Clinical, laboratory and ultrasound data were retrieved from RA and PsA patients recruited in the prospective MATRIX cohort study. Inclusion criteria were age≥18 years, RA or PsA diagnosis, and presence of active disease with eligibility to start/modify therapy. Patients underwent ultrasound-guided synovial biopsy of one of the most involved joints before starting/modifying treatment according to treat-to-target strategy. The samples were analysed by an expert pathologist for KSS calculation. Univariable and multivariable logistic regression analyses were performed to evaluate the relationship between KSS and baseline variables. The association between KSS and treatment response at 24 weeks of follow-up was investigated in univariable logistic regression analysis. Results 53 patients, 34 RA and 19 PsA, completed 24 weeks of follow-up after synovial biopsy. Patients were either treatment naïve (N=6, 11%), csDMARDs-experienced (N=46, 87%) or b/tsDMARDs-experienced (N=20, 38%). Median KSS was 6.00 (Q1-Q3 4.00-7.00) in RA and 4.00 (3.00-6.00) in PsA (p=0.040), and inflammatory infiltrates score was significantly higher in RA than in PsA patients (median 3.00 vs 2.00, p=0.021). In multivariable analysis, synovial effusion in the biopsied joint (OR 9.26, 95%CI 2.12-53.91) and erythrocyte sedimentation rate (ESR) (OR 1.04, 95%CI 1.01-1.08) associated with high KSS. High-grade synovitis significantly associated with a higher probability of achieving DAS28 remission, ACR20/50 response, and Boolean2.0 remission, independently from diagnosis. Conclusion Several markers of pro-inflammatory pathways associated with the presence of high-grade synovitis, and patients with higher KSS shared a higher probability of treatment targets achievement in the follow up. The integration of a simple and feasible tool like KSS in the clinical and prognostic stratification of patients with IA might help in intercepting patients with a disease more prone to respond to available treatment paradigms.
Collapse
Affiliation(s)
- Carlo Garaffoni
- Rheumatology Unit, Department of Medical Sciences, University of Ferrara and Azienda Ospedaliero-Universitaria S. Anna, Ferrara, Italy
| | - Marianna Tamussin
- Rheumatology Unit, Department of Medical Sciences, University of Ferrara and Azienda Ospedaliero-Universitaria S. Anna, Ferrara, Italy
| | - Ilaria Calciolari
- Rheumatology Unit, Department of Medical Sciences, University of Ferrara and Azienda Ospedaliero-Universitaria S. Anna, Ferrara, Italy
| | - Giovanni Lanza
- Anatomic Pathology, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Alessandra Bortoluzzi
- Rheumatology Unit, Department of Medical Sciences, University of Ferrara and Azienda Ospedaliero-Universitaria S. Anna, Ferrara, Italy
| | - Carlo Alberto Scirè
- Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) San Gerardo dei Tintori Foundation, Monza, Italy
- School of Medicine, University of Milano Bicocca, Milan, Italy
| | - Marcello Govoni
- Rheumatology Unit, Department of Medical Sciences, University of Ferrara and Azienda Ospedaliero-Universitaria S. Anna, Ferrara, Italy
| | - Ettore Silvagni
- Rheumatology Unit, Department of Medical Sciences, University of Ferrara and Azienda Ospedaliero-Universitaria S. Anna, Ferrara, Italy
| |
Collapse
|
10
|
Meyer A, Zack SR, Nijim W, Burgos A, Patel V, Zanotti B, Volin MV, Amin MA, Lewis MJ, Pitzalis C, Arami S, Karam JA, Sweiss NJ, Shahrara S. Metabolic reprogramming by Syntenin-1 directs RA FLS and endothelial cell-mediated inflammation and angiogenesis. Cell Mol Immunol 2024; 21:33-46. [PMID: 38105293 PMCID: PMC10757714 DOI: 10.1038/s41423-023-01108-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 11/01/2023] [Indexed: 12/19/2023] Open
Abstract
A novel rheumatoid arthritis (RA) synovial fluid protein, Syntenin-1, and its receptor, Syndecan-1 (SDC-1), are colocalized on RA synovial tissue endothelial cells and fibroblast-like synoviocytes (FLS). Syntenin-1 exacerbates the inflammatory landscape of endothelial cells and RA FLS by upregulating transcription of IRF1/5/7/9, IL-1β, IL-6, and CCL2 through SDC-1 ligation and HIF1α, or mTOR activation. Mechanistically, Syntenin-1 orchestrates RA FLS and endothelial cell invasion via SDC-1 and/or mTOR signaling. In Syntenin-1 reprogrammed endothelial cells, the dynamic expression of metabolic intermediates coincides with escalated glycolysis along with unchanged oxidative factors, AMPK, PGC-1α, citrate, and inactive oxidative phosphorylation. Conversely, RA FLS rewired by Syntenin-1 displayed a modest glycolytic-ATP accompanied by a robust mitochondrial-ATP capacity. The enriched mitochondrial-ATP detected in Syntenin-1 reprogrammed RA FLS was coupled with mitochondrial fusion and fission recapitulated by escalated Mitofusin-2 and DRP1 expression. We found that VEGFR1/2 and Notch1 networks are responsible for the crosstalk between Syntenin-1 rewired endothelial cells and RA FLS, which are also represented in RA explants. Similar to RA explants, morphological and transcriptome studies authenticated the importance of VEGFR1/2, Notch1, RAPTOR, and HIF1α pathways in Syntenin-1 arthritic mice and their obstruction in SDC-1 deficient animals. Consistently, dysregulation of SDC-1, mTOR, and HIF1α negated Syntenin-1 inflammatory phenotype in RA explants, while inhibition of HIF1α impaired synovial angiogenic imprint amplified by Syntenin-1. In conclusion, since the current therapies are ineffective on Syntenin-1 and SDC-1 expression in RA synovial tissue and blood, targeting this pathway and its interconnected metabolic intermediates may provide a novel therapeutic strategy.
Collapse
Affiliation(s)
- Anja Meyer
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Stephanie R Zack
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Wes Nijim
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Adel Burgos
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Vishwa Patel
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Brian Zanotti
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, USA
| | - Michael V Volin
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, USA
| | - M Asif Amin
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, MI, USA
| | - Myles J Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, London, UK
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, London, UK
- Department of Biomedical Sciences, Humanitas University, and Humanitas Research Hospital, Milan, Italy
| | - Shiva Arami
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Joseph A Karam
- Department of Orthopedic Surgery, the University of Illinois at Chicago, Chicago, IL, USA
| | - Nadera J Sweiss
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Shiva Shahrara
- Jesse Brown VA Medical Center, Chicago, IL, USA.
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
11
|
Ng S, Masarone S, Watson D, Barnes MR. The benefits and pitfalls of machine learning for biomarker discovery. Cell Tissue Res 2023; 394:17-31. [PMID: 37498390 PMCID: PMC10558383 DOI: 10.1007/s00441-023-03816-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/12/2023] [Indexed: 07/28/2023]
Abstract
Prospects for the discovery of robust and reproducible biomarkers have improved considerably with the development of sensitive omics platforms that can enable measurement of biological molecules at an unprecedented scale. With technical barriers to success lowering, the challenge is now moving into the analytical domain. Genome-wide discovery presents a problem of scale and multiple testing as standard statistical methods struggle to distinguish signal from noise in increasingly complex biological systems. Machine learning and AI methods are good at finding answers in large datasets, but they have a tendency to overfit solutions. It may be possible to find a local answer or mechanism in a specific patient sample or small group of samples, but this may not generalise to wider patient populations due to the high likelihood of false discovery. The rise of explainable AI offers to improve the opportunity for true discovery by providing explanations for predictions that can be explored mechanistically before proceeding to costly and time-consuming validation studies. This review aims to introduce some of the basic concepts of machine learning and AI for biomarker discovery with a focus on post hoc explanation of predictions. To illustrate this, we consider how explainable AI has already been used successfully, and we explore a case study that applies AI to biomarker discovery in rheumatoid arthritis, demonstrating the accessibility of tools for AI and machine learning. We use this to illustrate and discuss some of the potential challenges and solutions that may enable AI to critically interrogate disease and response mechanisms.
Collapse
Affiliation(s)
- Sandra Ng
- Centre for Translational Bioinformatics, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Sara Masarone
- Centre for Translational Bioinformatics, William Harvey Research Institute, Queen Mary University of London, London, UK
- Alan Turing Institute, London, UK
| | - David Watson
- Department of Informatics, King's College London, London, UK
| | - Michael R Barnes
- Centre for Translational Bioinformatics, William Harvey Research Institute, Queen Mary University of London, London, UK.
- Alan Turing Institute, London, UK.
| |
Collapse
|
12
|
Xie M, Zhu C, Ye Y. Ferroptosis-Related Molecular Clusters and Diagnostic Model in Rheumatoid Arthritis. Int J Mol Sci 2023; 24:ijms24087342. [PMID: 37108505 PMCID: PMC10138921 DOI: 10.3390/ijms24087342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/09/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by synovitis, joint damage and deformity. A newly described type of cell death, ferroptosis, has an important role in the pathogenesis of RA. However, the heterogeneity of ferroptosis and its association with the immune microenvironment in RA remain unknown. Synovial tissue samples from 154 RA patients and 32 healthy controls (HCs) were obtained from the Gene Expression Omnibus database. Twelve of twenty-six ferroptosis-related genes (FRGs) were differentially expressed between RA patients and HCs. Furthermore, the patterns of correlation among the FRGs were significantly different between the RA and HC groups. RA patients were classified into two distinct ferroptosis-related clusters, of which cluster 1 had a higher abundance of activated immune cells and a corresponding lower ferroptosis score. Enrichment analysis suggested that tumor necrosis factor-α signaling via nuclear factor-κB was upregulated in cluster 1. RA patients in cluster 1 responded better to anti-tumor necrosis factor (anti-TNF) therapy, which was verified by the GSE 198520 dataset. A diagnostic model to identify RA subtypes and immunity was constructed and verified, in which the area under the curve values in the training (70%) and validation (30%) cohorts were 0.849 and 0.810, respectively. This study demonstrated that there were two ferroptosis clusters in RA synovium that exhibited distinct immune profiles and ferroptosis sensitivity. Additionally, a gene scoring system was constructed to classify individual RA patients.
Collapse
Affiliation(s)
- Maosheng Xie
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Chao Zhu
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Yujin Ye
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
13
|
Ciliento MS, Venturelli V, Schettini N, Bertola R, Garaffoni C, Lanza G, Gafà R, Borghi A, Corazza M, Zabotti A, Missiroli S, Boncompagni C, Patergnani S, Perrone M, Giorgi C, Pinton P, Govoni M, Scirè CA, Bortoluzzi A, Silvagni E. Evaluation of the Synovial Effects of Biological and Targeted Synthetic DMARDs in Patients with Psoriatic Arthritis: A Systematic Literature Review and Meta-Analysis. Int J Mol Sci 2023; 24:5006. [PMID: 36902437 PMCID: PMC10002880 DOI: 10.3390/ijms24055006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/08/2023] Open
Abstract
The aims of this systematic literature review (SLR) were to identify the effects of approved biological and targeted synthetic disease modifying antirheumatic drugs (b/tsDMARDs) on synovial membrane of psoriatic arthritis (PsA) patients, and to determine the existence of histological/molecular biomarkers of response to therapy. A search was conducted on MEDLINE, Embase, Scopus, and Cochrane Library (PROSPERO:CRD42022304986) to retrieve data on longitudinal change of biomarkers in paired synovial biopsies and in vitro studies. A meta-analysis was conducted by adopting the standardized mean difference (SMD) as a measure of the effect. Twenty-two studies were included (19 longitudinal, 3 in vitro). In longitudinal studies, TNF inhibitors were the most used drugs, while, for in vitro studies, JAK inhibitors or adalimumab/secukinumab were assessed. The main technique used was immunohistochemistry (longitudinal studies). The meta-analysis showed a significant reduction in both CD3+ lymphocytes (SMD -0.85 [95% CI -1.23; -0.47]) and CD68+ macrophages (sublining, sl) (SMD -0.74 [-1.16; -0.32]) in synovial biopsies from patients treated for 4-12 weeks with bDMARDs. Reduction in CD3+ mostly correlated with clinical response. Despite heterogeneity among the biomarkers evaluated, the reduction in CD3+/CD68+sl cells during the first 3 months of treatment with TNF inhibitors represents the most consistent variation reported in the literature.
Collapse
Affiliation(s)
- Maria Sofia Ciliento
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara, Azienda Ospedaliero-Universitaria S. Anna, 44124 Cona, Italy
- Department of Precision Medicine, University della Campania L. Vanvitelli, 80138 Naples, Italy
| | - Veronica Venturelli
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara, Azienda Ospedaliero-Universitaria S. Anna, 44124 Cona, Italy
| | - Natale Schettini
- Section of Dermatology, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Riccardo Bertola
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara, Azienda Ospedaliero-Universitaria S. Anna, 44124 Cona, Italy
| | - Carlo Garaffoni
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara, Azienda Ospedaliero-Universitaria S. Anna, 44124 Cona, Italy
| | - Giovanni Lanza
- Anatomic Pathology, Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Roberta Gafà
- Anatomic Pathology, Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Alessandro Borghi
- Section of Dermatology, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Monica Corazza
- Section of Dermatology, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Alen Zabotti
- Department of Medical and Biological Sciences, Rheumatology Institute, University Hospital Santa Maria della Misericordia, 33100 Udine, Italy
| | - Sonia Missiroli
- Laboratory for Technologies of Advanced Therapies, Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Caterina Boncompagni
- Laboratory for Technologies of Advanced Therapies, Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Simone Patergnani
- Laboratory for Technologies of Advanced Therapies, Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Mariasole Perrone
- Laboratory for Technologies of Advanced Therapies, Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies, Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Paolo Pinton
- Laboratory for Technologies of Advanced Therapies, Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Marcello Govoni
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara, Azienda Ospedaliero-Universitaria S. Anna, 44124 Cona, Italy
| | - Carlo Alberto Scirè
- School of Medicine, University of Milano Bicocca, 20126 Milan, Italy
- Epidemiology Unit, Italian Society for Rheumatology, 20121 Milan, Italy
| | - Alessandra Bortoluzzi
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara, Azienda Ospedaliero-Universitaria S. Anna, 44124 Cona, Italy
| | - Ettore Silvagni
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara, Azienda Ospedaliero-Universitaria S. Anna, 44124 Cona, Italy
| |
Collapse
|
14
|
El Shikh MEM, El Sayed R, Aly NAR, Prediletto E, Hands R, Fossati-Jimack L, Bombardieri M, Lewis MJ, Pitzalis C. Follicular dendritic cell differentiation is associated with distinct synovial pathotype signatures in rheumatoid arthritis. Front Med (Lausanne) 2022; 9:1013660. [PMID: 36465908 PMCID: PMC9709129 DOI: 10.3389/fmed.2022.1013660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022] Open
Abstract
Follicular dendritic cells (FDCs) fundamentally contribute to the formation of synovial ectopic lymphoid-like structures in rheumatoid arthritis (RA) which is associated with poor clinical prognosis. Despite this critical role, regulation of FDC development in the RA synovium and its correlation with synovial pathotype differentiation remained largely unknown. Here, we demonstrate that CNA.42+ FDCs distinctively express the pericyte/fibroblast-associated markers PDGFR-β, NG2, and Thy-1 in the synovial perivascular space but not in established follicles. In addition, synovial RNA-Seq analysis revealed that expression of the perivascular FDC markers was strongly correlated with PDGF-BB and fibroid synovitis, whereas TNF-α/LT-β was significantly associated with lymphoid synovitis and expression of CR1, CR2, and FcγRIIB characteristic of mature FDCs in lymphoid follicles. Moreover, PDGF-BB induced CNA.42+ FDC differentiation and CXCL13 secretion from NG2+ synovial pericytes, and together with TNF-α/LT-β conversely regulated early and late FDC differentiation genes in unsorted RA synovial fibroblasts (RASF) and this was confirmed in flow sorted stromal cell subsets. Furthermore, RASF TNF-αR expression was upregulated by TNF-α/LT-β and PDGF-BB; and TNF-α/LT-β-activated RASF retained ICs and induced B cell activation in in vitro germinal center reactions typical of FDCs. Additionally, FDCs trapped peptidyl citrulline, and strongly correlated with IL-6 expression, and plasma cell, B cell, and T cell infiltration of the RA synovium. Moreover, synovial FDCs were significantly associated with RA disease activity and radiographic features of tissue damage. To the best of our knowledge, this is the first report describing the reciprocal interaction between PDGF-BB and TNF-α/LT-β in synovial FDC development and evolution of RA histological pathotypes. Selective targeting of this interplay could inhibit FDC differentiation and potentially ameliorate RA in clinically severe and drug-resistant patients.
Collapse
|