1
|
Zhou M, Lu Y, Tang Y, Zhang T, Xiao D, Zhang M, Zhang S, Li J, Cai X, Lin Y. A DNA-based nanorobot for targeting, hitchhiking, and regulating neutrophils to enhance sepsis therapy. Biomaterials 2025; 318:123183. [PMID: 39951831 DOI: 10.1016/j.biomaterials.2025.123183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/29/2024] [Accepted: 02/09/2025] [Indexed: 02/16/2025]
Abstract
Targeted regulation of neutrophils is an effective approach for treating neutrophil-driven inflammatory diseases, but challenges remain in minimizing off-target effects and extending drug half-life. A DNA-based nanorobot was developed to target neutrophils by using an N-acetyl Pro-Gly-Pro (Ac-PGP) peptide to specifically bind to the C-X-C motif of chemokine receptor 2 (CXCR2) on neutrophil membranes. This robot (a tetrahedral framework nucleic acid modified with Ac-PGP, APT) identified and hitchhiked neutrophils to accumulate at inflammatory sites and prolong its half-lives, whilst also was internalized to influence the neutrophil cell cycle and maturation process to regulate oxidative stress, inflammation, migration, and recruitment in both in vivo and in vitro inflammation experiments. Consequently, the tissue damage caused by sepsis was greatly reduced. This novel neutrophil-based nanorobot highlights the high precision of targeting and regulating neutrophils, and presents a potential strategy for treating multiple neutrophil-driven diseases.
Collapse
Affiliation(s)
- Mi Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yifei Lu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yuanlin Tang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Tianxu Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Dexuan Xiao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Mei Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Shunhao Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jun Li
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China; Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China; National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
2
|
Wilding S, Wu HHL, Brown N, Chinnadurai R. Anti-nuclear cytoplasmic antibody-associated vasculitis and kidney cancer: A mini review. World J Nephrol 2025; 14:105166. [DOI: 10.5527/wjn.v14.i2.105166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/26/2025] [Accepted: 03/08/2025] [Indexed: 04/09/2025] Open
Abstract
This mini review explores the links between anti-neutrophil cytoplasmic antibody-associated vasculitis (AAV) and kidney cancer. Several studies suggest an increased incidence of cancer for patients with AAV. Different cancer types have shown different standardized incidence ratios (SIRs) in association with AAV. The SIRs of kidney cancer were found to be between 1.7 and 3.3 as per three retrospective data analyses. This association is likely multifactorial, with increased de novo cancer risks associated with inflammatory diseases; carcinogenic therapies such as cyclophosphamide; and reduced immune surveillance of neoplastic cells in immunocompromised individuals. Some studies have proposed that cancers, including kidney cancer, could be a potential trigger for AAV. Due to variability in SIRs and a lack of multicenter studies looking specifically into the incidence of kidney cancer at AAV diagnosis and on follow-up post initiation of AAV treatment, there remains a lack of evidence to support formal screening for kidney cancer in the AAV patient cohort. Greater awareness on the increased risk of cancer in AAV patients, prompt urological assessment of “red flag” symptoms of kidney cancer, and smoking cessation advice to reduce cancer risk should be standard of care for patients with AAV.
Collapse
Affiliation(s)
- Samuel Wilding
- Donal O’Donoghue Renal Research Center and Department of Renal Medicine, Northern Care Alliance National Health Service Foundation Trust, Salford M6 8HD, United Kingdom
| | - Henry H L Wu
- Renal Research, Kolling Institute of Medical Research, Royal North Shore Hospital and The University of Sydney, Sydney 2065, Australia
| | - Nina Brown
- Donal O’Donoghue Renal Research Center and Department of Renal Medicine, Northern Care Alliance National Health Service Foundation Trust, Salford M6 8HD, United Kingdom
| | - Rajkumar Chinnadurai
- Donal O’Donoghue Renal Research Center and Department of Renal Medicine, Northern Care Alliance National Health Service Foundation Trust, Salford M6 8HD, United Kingdom
| |
Collapse
|
3
|
Monsalve DM, Acosta-Ampudia Y, Acosta NG, Celis-Andrade M, Şahin A, Yilmaz AM, Shoenfeld Y, Ramírez-Santana C. NETosis: A key player in autoimmunity, COVID-19, and long COVID. J Transl Autoimmun 2025; 10:100280. [PMID: 40071133 PMCID: PMC11894324 DOI: 10.1016/j.jtauto.2025.100280] [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: 02/04/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
NETosis, the process through which neutrophils release neutrophil extracellular traps (NETs), has emerged as a crucial mechanism in host defense and the pathogenesis of autoimmune responses. During the SARS-CoV-2 pandemic, this process received significant attention due to the central role of neutrophil recruitment and activation in infection control. However, elevated neutrophil levels and dysregulated NET formation have been linked to coagulopathy and endothelial damage, correlating with disease severity and poor prognosis in COVID-19. Moreover, it is known that SARS-CoV-2 can induce persistent low-grade systemic inflammation, known as long COVID, although the underlying causes remain unclear. It has been increasingly acknowledged that excessive NETosis and NET generation contribute to further pathophysiological abnormalities following SARS-CoV-2 infection. This review provides an updated overview of the role of NETosis in autoimmune diseases, but also the relationship between COVID-19 and long COVID with autoimmunity (e.g., latent and overt autoimmunity, molecular mimicry, epitope spreading) and NETosis (e.g., immune responses, NET markers). Finally, we discuss potential therapeutic strategies targeting dysregulated NETosis to mitigate the severe complications of COVID-19 and long COVID.
Collapse
Affiliation(s)
- Diana M. Monsalve
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Nicolás Guerrero Acosta
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Mariana Celis-Andrade
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Ali Şahin
- Selcuk University, Faculty of Medicine, Konya, Turkiye
| | - Ahsen Morva Yilmaz
- TUBITAK Marmara Research Center (TUBITAK-MAM), Life Sciences, Medical Biotechnology Unit, Kocaeli, Turkiye
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Reichman University, Herzelia, Israel
| | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
4
|
Rousselle A, Lodka D, Sonnemann J, Kling L, Kettritz R, Schreiber A. Endothelial but not systemic ferroptosis inhibition protects from antineutrophil cytoplasmic antibody-induced crescentic glomerulonephritis. Kidney Int 2025; 107:1037-1050. [PMID: 40122342 DOI: 10.1016/j.kint.2025.02.023] [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: 06/03/2024] [Revised: 12/20/2024] [Accepted: 02/25/2025] [Indexed: 03/25/2025]
Abstract
Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAV) are systemic autoimmune diseases featuring small blood vessel inflammation and organ damage, including necrotizing crescentic glomerulonephritis (NCGN). Persistent vascular inflammation leads to endothelial and kidney cell necrosis. Ferroptosis is a regulated cell death pathway executed by reactive oxygen species and iron-dependent lipid peroxidation culminating in cell membrane rupture. Here we show that ANCA-activated neutrophils induced endothelial cell (EC) death in vitro that was prevented by ferroptosis inhibition with Ferrostatin-1, Liproxstatin-1 and small inhibiting RNA against the enzyme AcylCoA Synthetase Long Chain Family Member 4 (ACSL4). In contrast, neither necroptosis nor apoptosis inhibition affected EC death. Moreover, both ferroptosis inhibitors alleviated lipid peroxide accumulation in EC. Increased lipid peroxidation was detected in kidney sections of AAV mice by immunohistochemistry. We generated MPO-/- ACSL4flox Tie2-Cre+ mice lacking ACSL4 specifically in EC (ACSL4ΔEC) to study the significance of endothelial ferroptosis in vivo. ACSL4ΔEC chimeric mice, but not control mice (ACSL4WT), were protected from NCGN in an MPO-AAV bone-marrow transplantation model. These data establish that EC ferroptosis contributes to ANCA-induced glomerulonephritis. However, systemic pharmacological ferroptosis inhibition with Ferrostatin-1 or Liproxstatin-1 did not protect from NCGN in a murine AAV model. Ferrostatin-1 treatment both directly activated T-cell proliferation and indirectly myeloid-mediated T-cell proliferation and polarization in vitro. Conceivably, both effects may cancel the beneficial effect of endothelial ferroptosis inhibition. Mechanistically, we describe the importance of EC ferroptosis for the development of AAV. However, the lack of protection with systemic pharmacological ferroptosis inhibition should discourage clinicians from evaluating this treatment strategy in clinical AAV studies.
Collapse
Affiliation(s)
- Anthony Rousselle
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine (MDC) and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dörte Lodka
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine (MDC) and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Janis Sonnemann
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine (MDC) and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Department of Nephrology and Medical Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lovis Kling
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine (MDC) and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Department of Nephrology and Medical Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ralph Kettritz
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine (MDC) and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Department of Nephrology and Medical Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Adrian Schreiber
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine (MDC) and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Department of Nephrology and Medical Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
5
|
Geetha D, Pagnoux C, Sattui SE, Merkel PA, Weiner M, Draibe J, Faguer S, Bray S, Gurlin RE, Balcells-Oliver M, Bruchfeld A, Jayne DR, ADVOCATE Study Group
. Efficacy and safety of avacopan in patients aged 65 years and older with ANCA-associated vasculitis: a post hoc analysis of data from the ADVOCATE trial. Rheumatology (Oxford) 2025; 64:3863-3871. [PMID: 40037556 DOI: 10.1093/rheumatology/keaf122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/24/2025] [Accepted: 02/07/2025] [Indexed: 03/06/2025] Open
Abstract
OBJECTIVES To evaluate the efficacy and safety of avacopan in patients aged ≥65 years with granulomatosis with polyangiitis (GPA) or microscopic polyangiitis (MPA) in the phase 3 ADVOCATE trial of avacopan vs a prednisone taper, plus either rituximab or cyclophosphamide. METHODS In this descriptive, post hoc analysis, patients receiving avacopan or a prednisone taper were stratified by age. Key efficacy outcomes included the rate of remission at week 26 and sustained remission at week 52. RESULTS Of 160 patients aged ≥65, 109 were aged 65-74 and 51 were ≥75. Remission at week 26 was achieved in 71.7% vs 69.4% of patients aged 65-74 and 73.1% vs 72.0% aged ≥75 in the avacopan vs prednisone taper groups, respectively. Sustained remission at week 52 was observed in 65.0% vs 55.1% of patients aged 65-74 and 65.4% vs 56.0% aged ≥75. Relapse rates in the avacopan vs prednisone taper groups were 12.3% vs 18.8% and 3.8% vs 20.8% in the 65-74 and ≥75 subgroups, respectively. Improvements in estimated glomerular filtration rate and health-related quality of life were observed in both treatment groups. Use of avacopan compared with a prednisone taper was associated with a 61% and 49% reduction in mean glucocorticoid dose in the 65-74 and ≥75 subgroups, respectively, and lower glucocorticoid toxicity. The proportions of patients with adverse events were similar between treatment groups within each age subgroup. CONCLUSION These data support the efficacy and safety of an avacopan-based regimen to treat patients with GPA or MPA aged ≥65.
Collapse
Affiliation(s)
- Duvuru Geetha
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christian Pagnoux
- Division of Rheumatology, Vasculitis Clinic, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Sebastian E Sattui
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peter A Merkel
- Division of Rheumatology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Epidemiology, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria Weiner
- Department of Nephrology and Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Juliana Draibe
- Department of Nephrology, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Stanislas Faguer
- Department of Nephrology and Organ Transplantation, Toulouse University Hospital, Toulouse, France
| | | | | | | | - Annette Bruchfeld
- Department of Nephrology and Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Department of Renal Medicine, Karolinska University Hospital, CLINTEC Karolinska Institutet, Stockholm, Sweden
| | - David R Jayne
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | |
Collapse
Collaborators
C Au Peh, A Chakera, B Cooper, J Kurtkoti, D Langguth, V Levidiotis, G Luxton, P Mount, D Mudge, E Noble, R Phoon, D Ranganathan, A Ritchie, J Ryan, M Suranyi, A Rosenkranz, K Lhotta, A Kronbichler, N Demoulin, C Bovy, R Hellemans, J Hougardy, B Sprangers, K Wissing, C Pagnoux, S Barbour, S Brachemi, S Cournoyer, L Girard, L Laurin, P Liang, D Philibert, M Walsh, V Tesar, R Becvar, P Horak, I Rychlik, W Szpirt, H Dieperink, J Gregersen, P Ivarsen, E Krarup, C Lyngsoe, C Rigothier, J Augusto, A Belot, D Chauveau, D Cornec, N Jourde-Chiche, M Ficheux, A Karras, A Klein, F Maurier, R Mesbah, O Moranne, A Neel, T Quemeneur, D Saadoun, B Terrier, P Zaoui, M Schaier, U Benck, R Bergner, M Busch, J Floege, F Grundmann, H Haller, M Haubitz, B Hellmich, J Henes, B Hohenstein, C Hugo, C Iking-Konert, F Arndt, T Kubacki, I Kotter, P Lamprecht, T Lindner, J Halbritter, H Mehling, U Schönermarck, N Venhoff, V Vielhauer, O Witzke, I Szombati, G Szucs, G Garibotto, F Alberici, E Brunetta, L Dagna, S De Vita, G Emmi, A Gabrielli, L Manenti, F Pieruzzi, D Roccatello, C Salvarani, M Harigai, H Dobashi, T Atsumi, S Fujimoto, N Hagino, A Ihata, S Kaname, Y Kaneko, A Katagiri, M Katayama, Y Kirino, K Kitagawa, A Komatsuda, H Kono, T Kurasawa, R Matsumura, T Mimura, A Morinobu, Y Murakawa, T Naniwa, T Nanki, N Ogawa, H Oshima, K Sada, E Sugiyama, T Takeuchi, H Taki, N Tamura, T Tsukamoto, K Yamagata, M Yamamura, P van Daele, A Rutgers, Y Teng, R Walker, I Chua, M Collins, K Rabindranath, J de Zoysa, M Svensson, B Grevbo, S Kalstad, M Little, M Clarkson, E Molloy, I Agraz Pamplona, J Anton, V Barrio Lucia, S Ciggaran, M Cinta Cid, M Diaz Encarnacion, X Fulladosa Oliveras, M Jose Soler, H Marco Rusinol, M Praga, L Quintana Porras, A Segarra, A Bruchfeld, M Segelmark, I Soveri, E Thomaidi, K Westman, T Neumann, M Burnier, T Daikeler, J Dudler, T Hauser, H Seeger, B Vogt, D Jayne, J Burton, R Al Jayyousi, T Amin, J Andrews, L Baines, P Brogan, B Dasgupta, T Doulton, O Flossmann, S Griffin, J Harper, L Harper, D Kidder, R Klocke, P Lanyon, R Luqmani, J McLaren, D Makanjuola, L McCann, A Nandagudi, S Selvan, E O'Riordan, M Patel, R Patel, C Pusey, R Rajakariar, J Robson, M Robson, A Salama, L Smyth, J Sznajd, J Taylor, P Merkel, A Sreih, E Belilos, A Bomback, J Carlin, Y Chang Chen Lin, V Derebail, S Dragoi, A Dua, L Forbess, D Geetha, P Gipson, R Gohh, G T Greenwood, S Hugenberg, R Jimenez, M Kaskas, T Kermani, A Kivitz, C Koening, C Langford, G Marder, A Mohamed, P Monach, N Neyra, G Niemer, J Niles, R Obi, C Owens, D Parks, A Podoll, B Rovin, R Sam, W Shergy, A Silva, U Specks, R Spiera, J Springer, C Striebich, A Swarup, S Thakar, A Tiliakos, Y Tsai, D Waguespack, M Chester Wasko,
Collapse
|
6
|
Wang H, Engesser J, Khatri R, Schaub DP, Paust HJ, Sultana Z, Jauch-Speer SL, Peters A, Kaffke A, Bonn S, Huber TB, Mittrücker HW, Krebs CF, Panzer U, Asada N. Type I interferon drives T cell cytotoxicity by upregulation of interferon regulatory factor 7 in autoimmune kidney diseases in mice. Nat Commun 2025; 16:4686. [PMID: 40393992 DOI: 10.1038/s41467-025-59819-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 05/06/2025] [Indexed: 05/22/2025] Open
Abstract
In anti-neutrophil cytoplasmic antibody-associated vasculitis (AAV) and systemic lupus erythematosus (SLE), glomerulonephritis is a severe kidney complication driven by immune cells, including T cells. However, the mechanisms underlying T cell activation in these contexts remain elusive. Here we report that in patients with AAV and SLE, type I interferon (IFN-I) induces T cell differentiation into interferon-stimulated genes-expressing T (ISG-T) cells, which are characterized by an elevated IFN-I signature, an immature phenotype, and cytotoxicity in inflamed tissue. Mechanistically, IFN-I stimulates the expression of interferon regulatory factor 7 (IRF7) in T cells, which in turn induces granzyme B production. In mice, blocking IFN-I signaling reduces IRF7 and granzyme B expression in T cells, thus ameliorating glomerulonephritis. In parallel, spatial transcriptomic analyses of kidney biopsies from patients with AAV or SLE reveal an elevated ISG signature and the presence of ISG-T cells in close proximity to plasmacytoid dendritic cells, the primary producers of IFN-I. Our results from both patients and animal models thus suggest that IFN-I production in inflamed tissue may drive ISG-T cell differentiation to expand the pool of cytotoxic T cells in autoimmune diseases.
Collapse
Affiliation(s)
- Huiying Wang
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonas Engesser
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Robin Khatri
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Darius P Schaub
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Joachim Paust
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Zeba Sultana
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Anett Peters
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Kaffke
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Bonn
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Haburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Haburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Willi Mittrücker
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute for Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian F Krebs
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Haburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulf Panzer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Haburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Nariaki Asada
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
7
|
Lermi N, Yağız B, Ekin A, Coşkun BN, Dalkılıç E, Pehlivan Y. Patients Diagnosed with Granulomatosis with Polyangiitis: The Journey to Receive Rheumatologist Care. J Clin Med 2025; 14:3523. [PMID: 40429518 DOI: 10.3390/jcm14103523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Revised: 05/11/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Granulomatosis with polyangiitis (GPA) is a necrotising vasculitis characterised by granulomatous inflammation involving small vessels. In addition to specific findings for the affected organ, constitutional symptoms and joint and muscle pain can be observed. The prodromal phase, where symptoms last for months before clinical diagnosis, may suggest infection or malignancy. This may cause a delay in GPA diagnosis. The period from the first symptoms to diagnosis may last from one month to three years. In this study, we aimed to demonstrate that, as the time between the onset of symptoms and diagnosis increases, the disease involvement may become more severe, and the possibility of recurrence may increase, indicating the importance of early diagnosis. Methods: For this cross-sectional retrospective study, data from 40 patients with GPA were evaluated. Demographic, clinical, treatment, and follow-up characteristics of the patients were obtained from their medical records. Results: The mean time between the presentation of the first complaints and GPA diagnosis was 7.40 ± 11.84 (0, 60, 3; mean ± standard deviation [minimum, maximum, and median]) months. The time between the first complaints and diagnosis was longer for patients with a history of recurrence (11.44 ± 16.73 [0, 60, 4.5] vs. 4.71 ± 6.04 [0, 24, 2.5], p value = 0.260). Conclusions: GPA is an inflammatory disease with various clinical presentations. In the management of patients with GPA, awareness of its presentation is important for rheumatologists as well as other clinicians during the initial evaluation, demonstrating the importance of interclinical collaboration.
Collapse
Affiliation(s)
- Nihal Lermi
- Department of Rheumatology, Harakani Public Hospital, Kars 36200, Turkey
| | - Burcu Yağız
- Division of Rheumatology, Faculty of Medicine, Uludag University, Bursa 16059, Turkey
| | - Ali Ekin
- Division of Rheumatology, Van Yüzüncü Yıl University Faculty of Medicine, Van 65090, Turkey
| | - Belkıs Nihan Coşkun
- Division of Rheumatology, Faculty of Medicine, Uludag University, Bursa 16059, Turkey
| | - Ediz Dalkılıç
- Division of Rheumatology, Faculty of Medicine, Uludag University, Bursa 16059, Turkey
| | - Yavuz Pehlivan
- Division of Rheumatology, Faculty of Medicine, Uludag University, Bursa 16059, Turkey
| |
Collapse
|
8
|
Quintero-Giraldo LP, Barahona-Correa J, Corredor-Orlandelli D, García-Alfonso C, Herrera-Leaño N, Fernández-Ávila DG. Nervous system involvement in ANCA-associated vasculitis: Single center experience from Latin America. Semin Arthritis Rheum 2025; 73:152751. [PMID: 40409118 DOI: 10.1016/j.semarthrit.2025.152751] [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: 12/08/2024] [Revised: 04/01/2025] [Accepted: 04/24/2025] [Indexed: 05/25/2025]
Abstract
INTRODUCTION Anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitides (AAV) present heterogeneous neurological symptoms that are often misdiagnosed, contributing to delays in identification and prompt treatment. Few studies in Latin America have described the frequency of neurological involvement in AVV; none have explicitly described the characteristics of nervous system involvement. DESIGN/METHODS This case-control study examined patient records for AVV treated at a university hospital in Colombia between 2005 and 2023. Patients with and without neurological manifestations were compared and a survival analysis was performed. RESULTS Forty-eight cases and seventy-nine controls were included. The median age was 58 years, 57.5 % were female. The diagnosis was made in 67.7 % of cases during the hospital stay, and in-hospital mortality was 14 %. Nervous system involvement was more frequent in undifferentiated AAV (100 %), followed by eosinophilic granulomatosis with polyangiitis (75 %), microscopic polyangiitis (33.3 %), and granulomatosis with polyangiitis (25.9 %). The most common neurological manifestations were peripheral neuropathy (50 %), patient-reported symptoms of sensory dysfunction (43.7 %), and cranial neuropathy (39.6 %); headache was frequent among patients with neurological involvement. Patients with neurological manifestations presented a lower median creatinine at admission and a lower proportion of patients with a five-factor score > 2. No differences in one-year all-cause mortality were observed. CONCLUSIONS This study presents an exhaustive clinical characterization of the neurological profile of patients with AAV from a single center in Latin America. Patients with nervous system involvement showed less severe renal involvement and a lower proportion of 5-year risk of mortality scores; one-year all-cause mortality was similar between groups.
Collapse
Affiliation(s)
| | - Julian Barahona-Correa
- Department of Internal Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia; Division of Rheumatology, Hospital Universitario San Ignacio, Bogotá, Colombia
| | | | | | - Nancy Herrera-Leaño
- Department of Internal Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia; Division of Cardiology, Hospital Universitario San Ignacio, Bogotá, Colombia
| | - Daniel G Fernández-Ávila
- Department of Internal Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia; Division of Rheumatology, Hospital Universitario San Ignacio, Bogotá, Colombia
| |
Collapse
|
9
|
Sato M, Nagai K, Sato T, Yoshimoto R, Shibano Y, Shibahara M, Satokawa H, Anzai M, Uchida T, Tsutiya A, Takakuwa Y, Omoteyama K, Arito M, Suematsu N, Ooka S, Kawahata K, Kato T, Kurokawa MS. Aberrant oxidative modifications of neutrophil myeloperoxidase in anti-neutrophil cytoplasmic antibody-associated vasculitis. J Proteomics 2025; 315:105412. [PMID: 39993524 DOI: 10.1016/j.jprot.2025.105412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
Anti-neutrophil cytoplasmic antibodies directed to myeloperoxidase (MPO-ANCA) are key molecules in the pathogenesis of ANCA-associated vasculitis (AAV), however, the mechanisms of autoantibody production have not been elucidated. We hypothesized that an aberrant PTM occurs in the MPO of MPO-ANCA-positive AAV (MPO-AAV), which induces immune responses to self MPO. To test this, we purified MPO proteins from neutrophils of 8 patients with MPO-AAV and 8 healthy subjects, digested them with trypsin, and comprehensively quantified PTMs of the MPO peptides using the sequential window acquisition of all theoretical fragment ion spectra (SWATH) method of LC-MS. Among the 1034 detected MPO peptides, 38 peptides were increased in the patients with MPO-AAV relative to the healthy subjects, whereas 10 peptides were decreased in the patients (p < 0.05). Interestingly, oxidative modifications were found in 11 of the 38 increased peptides (1.14- to 3.29-fold), but not in the decreased peptides. These included oxidation of Met577, Phe686, Met688 and Met719, dioxidation of Met409, Phe605, Trp679 and Met719, and kynurenylation of Trp255. Conversely, glycosylation was detected in 4 of the 10 decreased peptides (-1.32- to -2.32-fold), but not in the increased peptides. They were O-type glycans at Ser357 and Ser731, and N-type glycans at Asn355 and Asn729. In animal experiments, immunization of mice with in vitro oxidized or unoxidized mouse MPO (mMPO) showed that not only anti-oxidized mMPO antibodies but also anti-unoxidized mMPO antibodies were preferentially produced in the oxidized mMPO-immunized mice relative to the unoxidized mMPO-immunized mice (anti-oxidized mMPO antibodies, 6/8 vs 1/9, p < 0.05; anti-unoxidized mMPO antibodies, 4/8 vs 0/9, p < 0.05). Our results suggest that the increased oxidative modifications of MPO in MPO-AAV may break immune tolerance and trigger the MPO-ANCA production. SIGNIFICANCE: AAV is a systemic and refractory disease that causes life-threatening multi-organ involvement such as necrotizing glomerulonephritis and lung hemorrhage. MPO-ANCA is an autoantibody that plays a key role in the pathogenesis of AAV. Therefore, elucidation of the mechanism of MPO-ANCA production is crucial to overcoming this disease. In this study, we applied a SWATH-MS analysis to the detection of aberrant PTMs, and found increased oxidative modifications of neutrophil MPO in patients with MPO-AAV for the first time. Immunization of in vitro oxidized MPO induced autoantibodies to the intact unoxidized MPO, suggesting that the increased oxidative modifications of MPO may break the immune tolerance in MPO-AAV. This study suggests a novel trigger mechanism for MPO-ANCA production.
Collapse
Affiliation(s)
- Masaaki Sato
- Clinical Proteomics and Molecular Medicine, St. Marianna University Graduate School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa 216-8511, Japan.
| | - Kouhei Nagai
- Graduate School of Biology-Oriented Science and Technology, Kindai University, 930 Nishimitani, Kinokawa, Wakayama 649-6493, Japan.
| | - Toshiyuki Sato
- Clinical Proteomics and Molecular Medicine, St. Marianna University Graduate School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa 216-8511, Japan.
| | - Ryo Yoshimoto
- Graduate School of Biology-Oriented Science and Technology, Kindai University, 930 Nishimitani, Kinokawa, Wakayama 649-6493, Japan
| | - Yuto Shibano
- Faculty of Biology-Oriented Science and Technology, Kindai University, 930 Nishimitani, Kinokawa, Wakayama 649-6493, Japan
| | - Minori Shibahara
- Faculty of Biology-Oriented Science and Technology, Kindai University, 930 Nishimitani, Kinokawa, Wakayama 649-6493, Japan
| | - Haruka Satokawa
- Faculty of Biology-Oriented Science and Technology, Kindai University, 930 Nishimitani, Kinokawa, Wakayama 649-6493, Japan
| | - Masayuki Anzai
- Institute of Advanced Technology, Kindai University, 14-1, Minamiakasaka, Kainan, Wakayama 642-0017, Japan.
| | - Teisuke Uchida
- Disease Biomarker Analysis and Molecular Regulation, St. Marianna University Graduate School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa 216-8511, Japan
| | - Atsuhiro Tsutiya
- Clinical Proteomics and Molecular Medicine, St. Marianna University Graduate School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa 216-8511, Japan.
| | - Yukiko Takakuwa
- Department of Rheumatology and Allergology, St. Marianna University School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa 216-8511, Japan.
| | - Kazuki Omoteyama
- Clinical Proteomics and Molecular Medicine, St. Marianna University Graduate School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa 216-8511, Japan.
| | - Mitsumi Arito
- Clinical Proteomics and Molecular Medicine, St. Marianna University Graduate School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa 216-8511, Japan.
| | - Naoya Suematsu
- Clinical Proteomics and Molecular Medicine, St. Marianna University Graduate School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa 216-8511, Japan.
| | - Seido Ooka
- Department of Rheumatology and Allergology, St. Marianna University School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa 216-8511, Japan.
| | - Kimito Kawahata
- Department of Rheumatology and Allergology, St. Marianna University School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa 216-8511, Japan.
| | - Tomohiro Kato
- Clinical Proteomics and Molecular Medicine, St. Marianna University Graduate School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa 216-8511, Japan.
| | - Manae S Kurokawa
- Disease Biomarker Analysis and Molecular Regulation, St. Marianna University Graduate School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa 216-8511, Japan.
| |
Collapse
|
10
|
Drouzas K, Kalogeropoulos P, Liapis G, Lionaki S. Current Understanding of the Pathogenesis of ANCA-Associated Vasculitis and Novel Treatment Options Targeting Complement Activation. Life (Basel) 2025; 15:756. [PMID: 40430184 DOI: 10.3390/life15050756] [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: 02/19/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/29/2025] Open
Abstract
Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (ANCA-vasculitis) is an autoimmune disease characterized by inflammation and necrosis of small or medium vessels. In the past, the role of the complement in the pathogenesis of ANCA-vasculitis has been underestimated, due to the paucity of the complement at sites of injured glomeruli. Following evidence from animal models of the major role of the complement in pathogenesis of ANCA-vasculitis, the complement has again attracted interest. Immunohistology analysis of pauci-immune glomerulonephritis-ANCA glomerulonephritis (ANCA-GN)-reveals the presence of complement products and membrane attack complex, suggesting their involvement in the disease process. Researchers emphasize the complement classical or lectin pathway as a contributor to the development of ANCA-vasculitis. The era of targeted therapies to suspend the complement activation as a therapy for ANCA-vasculitis has arrived, and thus, the comprehension of its role is very important. This review summarizes recent insights on the important role of complement activation in the development of ANCA-vasculitis as well as the emerging therapeutic possibilities that target complement components for the treatment of this condition.
Collapse
Affiliation(s)
- Konstantinos Drouzas
- Department of Nephrology, 2nd Propaedeutic Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12461 Athens, Greece
| | - Petros Kalogeropoulos
- Department of Nephrology, 2nd Propaedeutic Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12461 Athens, Greece
| | - George Liapis
- 1st Department of Pathology Medical School, National and Kapodistrian University of Athens and Laikon Hospital, 11527 Athens, Greece
| | - Sophia Lionaki
- Department of Nephrology, 2nd Propaedeutic Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12461 Athens, Greece
| |
Collapse
|
11
|
Bello F, Fagni F, Bagni G, Hill CL, Mohammad AJ, Moiseev S, Olivotto I, Seyahi E, Emmi G. Arterial and venous thrombosis in systemic and monogenic vasculitis. Nat Rev Rheumatol 2025:10.1038/s41584-025-01252-7. [PMID: 40329108 DOI: 10.1038/s41584-025-01252-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2025] [Indexed: 05/08/2025]
Abstract
Systemic vasculitis, common forms of which include anti-neutrophil cytoplasmic antibody-associated small-vessel vasculitis, large-vessel vasculitis and Behçet syndrome, are frequently complicated by arterial or venous thrombotic events (AVTEs). Newly identified entities such as DADA2 (deficiency of adenosine deaminase 2) and VEXAS (vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic) syndrome, which are driven by genetic mutations, also exhibit vasculitic features and are associated with a high risk of AVTEs. AVTEs in systemic vasculitis, including monogenic forms of vasculitis, are due to the complex interaction of inflammation and coagulation. New insights into the pathogenetic mechanisms implicate endothelial dysfunction, immune complex deposition and the interplay of pro-inflammatory cytokines with prothrombotic factors, which collectively promote thrombus formation. AVTEs impose a substantial disease burden, complicate diagnosis and negatively affect prognosis by increasing the risk of morbidity and mortality. Early diagnosis and treatment are crucial to prevent lasting damage. Management strategies should target both thrombosis and underlying inflammation. Antithrombotic therapies, including low-dose aspirin, or oral anticoagulants should be used on the basis of individual thrombotic risk assessment. Immunosuppressive therapy is the cornerstone of treatment for arterial and venous thrombosis, particularly in Behçet syndrome, in which vascular inflammation has a crucial role in thrombotic complications.
Collapse
Affiliation(s)
- Federica Bello
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Filippo Fagni
- Department of Internal Medicine 3, Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Giacomo Bagni
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Catherine L Hill
- Rheumatology Unit, The Queen Elizabeth Hospital, Woodville, South Australia, Australia
- Department of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Aladdin J Mohammad
- Department of Clinical Sciences, Rheumatology, Lund University, Lund, Sweden
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Sergey Moiseev
- Tareev Clinic of Internal Disease, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Iacopo Olivotto
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Meyer Children's Hospital IRCCS, Florence, Italy
| | - Emire Seyahi
- Division of Rheumatology, Department of Internal Medicine and Behçet's Disease Research Centre, Istanbul University-Cerrahpasa, School of Medicine, Istanbul, Turkey
| | - Giacomo Emmi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy.
- Clinical Medicine and Rheumatology Unit, Cattinara University Hospital, Trieste, Italy.
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Melbourne, Victoria, Australia.
| |
Collapse
|
12
|
Hamid A, Vandergheynst F, Ilzkovitz M. Exploring ANCA Pathogenicity in Ulcerative Colitis: A Case Report Highlighting the Risk of Progression to ANCA-Associated Vasculitis. Eur J Case Rep Intern Med 2025; 12:005061. [PMID: 40352707 PMCID: PMC12061215 DOI: 10.12890/2025_005061] [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: 11/23/2024] [Accepted: 01/06/2025] [Indexed: 05/14/2025] Open
Abstract
Background The relationship between anti-neutrophil cytoplasmic antibodies (ANCA) and inflammatory bowel disease has attracted significant attention due to shared immunopathological mechanisms and clinical associations. Case description This case report describes a 20-year-old African woman with a history of ulcerative colitis who developed diffuse alveolar haemorrhage in the setting of elevated proteinase 3 (PR3)-ANCA. She was treated for ANCA-associated vasculitis with corticosteroids, plasma exchange, and rituximab, leading to a complete resolution. Conclusion The case highlights the association between ulcerative colitis and ANCA-associated vasculitis, particularly with PR3-ANCA, and the importance of recognizing this overlap. Although ANCA are frequently present in inflammatory bowel disease, especially ulcerative colitis, their pathogenic role remains unclear. The incidence of ANCA-associated vasculitis is higher in inflammatory bowel disease patients, with ulcerative colitis often preceding ANCA-associated vasculitis. Despite the common presence of ANCA in inflammatory bowel disease, not all patients progress to ANCA-associated vasculitis. This case underscores the need for careful monitoring in ulcerative colitis patients with elevated PR3-ANCA and the role of ANCA in guiding diagnosis and treatment. LEARNING POINTS High proteinase 3 (PR3)-anti-neutrophil cytoplasmic antibodies (ANCA) titre impacts the severity and management of ulcerative colitis patients.ANCA-associated vasculitis is more frequent in ulcerative colitis patients and occurs several years after ANCA positivity.Close monitoring of ulcerative colitis patients with PR3-ANCA is essential for early detection of progression to ANCA-associated vasculitis.
Collapse
Affiliation(s)
- Abir Hamid
- Internal Medicine Department, Hôpital Erasme, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Frederic Vandergheynst
- Internal Medicine Department, Hôpital Erasme, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Maxime Ilzkovitz
- Internal Medicine Department, Hôpital Erasme, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
13
|
Toor KK, Chen A, Cabral DA, Mammen C, Bosman ES, Shen Y, Bone JN, Noone D, Al‐Abadi E, Benseler S, Berard R, Bohm M, Charuvanij S, Cook K, Dancey P, Deepak S, Duffy C, Eberhard B, Elder M, Foell D, Gerstbacher D, Heshin‐Bekenstein M, Huber A, James KE, Kim S, Klein‐Gitelman M, Martin N, McErlane F, Moorthy LN, Myrup C, Riley P, Shenoi S, Sivaraman V, Tanner T, Tarvin S, Wagner‐Weiner L, Yeung RSM, Brown KL, Morishita KA, the PedVas Investigators Network. Evaluating Renal Disease in Pediatric-Onset Antineutrophil Cytoplasmic Antibody-Associated Vasculitis: Disease Course, Outcomes, and Predictors of Outcome. Arthritis Rheumatol 2025; 77:606-614. [PMID: 39624015 PMCID: PMC12039468 DOI: 10.1002/art.43071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 10/15/2024] [Accepted: 10/23/2024] [Indexed: 01/16/2025]
Abstract
OBJECTIVE We aimed to study the disease course, outcomes, and predictors of outcome in pediatric-onset antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) affecting the kidneys. METHODS Patients eligible for this study had a diagnosis of granulomatosis with polyangiitis (GPA), microscopic polyangiitis, or ANCA-positive pauci-immune glomerulonephritis, were 18 years or younger at diagnosis, had renal disease defined by biopsy or dialysis dependence, and had clinical data at diagnosis and at either 12 or 24 months. Ambispective data from A Registry for Children with Vasculitis/Pediatric Vasculitis Initiative Registry was used. The primary outcome was inactive renal disease (pediatric vasculitis activity score = 0 or 1) at 12 months. Secondary outcomes included rates of improved renal function and damage within 24 months. Renal function, defined by estimated glomerular filtration rate, was categorized into Kidney Disease Improving Global Outcomes (KDIGO) stages at diagnosis and tested as a predictor of outcome using a proportional-odds logistic regression model. RESULTS A total of 145 patients were included: 68% were female, and 78% had GPA. At 12 months, 83% of patients achieved inactive renal disease; however, 42% had evidence of permanent renal damage. Compared with patients with normal renal function at diagnosis, patients with moderate to severely reduced renal function, or kidney failure at diagnosis, had an odds ratio of 8.62 (P = 0.002; 95% confidence interval [CI] 2.31-32.1) and 26.3 (P < 0.001; 95% CI 6.32-109), respectively, for being in a non-normal KDIGO category at 12 months. CONCLUSION The majority of patients with pediatric AAV achieve inactive renal disease by 12 months; however, almost half have evidence of damage. Renal function at diagnosis is a strong predictor of renal function at 12 months.
Collapse
Affiliation(s)
- Kirandeep K. Toor
- University of British Columbia and BC Children's Hospital and BC Children's Hospital Research InstituteVancouverBritish ColumbiaCanada
| | - Audrea Chen
- The Hospital for Sick Children, University of TorontoTorontoOntarioCanada
| | - David A. Cabral
- University of British Columbia and BC Children's Hospital and BC Children's Hospital Research InstituteVancouverBritish ColumbiaCanada
| | - Cherry Mammen
- University of British Columbia and BC Children's Hospital and BC Children's Hospital Research InstituteVancouverBritish ColumbiaCanada
| | - Else S. Bosman
- University of British ColumbiaVancouverBritish ColumbiaCanada
| | - Ye Shen
- BC Children's Hospital Research InstituteVancouverBritish ColumbiaCanada
| | - Jeffrey N. Bone
- BC Children's Hospital Research InstituteVancouverBritish ColumbiaCanada
| | - Damien Noone
- The Hospital for Sick Children, University of TorontoTorontoOntarioCanada
| | - Eslam Al‐Abadi
- Birmingham Children's Hospital NHS Foundation TrustBirminghamUnited Kingdom
| | | | | | - Marek Bohm
- Leeds Children's HospitalLeedsUnited Kingdom
| | | | | | - Paul Dancey
- Janeway Children's Health and Rehabilitation CentreSt. John'sNewfoundland and LabradorCanada
| | | | - Ciaran Duffy
- Children's Hospital of Eastern OntarioOttawaOntarioCanada
| | | | | | - Dirk Foell
- University Children's Hospital MunsterMunsterGermany
| | | | | | - Adam Huber
- IWK Health CentreHalifaxNova ScotiaCanada
| | - Karen E. James
- Primary Children's Medical Center, University of UtahSalt Lake City
| | - Susan Kim
- Benioff Children's Hospital, University of CaliforniaSan Francisco
| | | | - Neil Martin
- Royal Hospital for ChildrenGlasgowUnited Kingdom
| | | | | | | | - Phil Riley
- Manchester Children's HospitalManchesterUnited Kingdom
| | - Susan Shenoi
- Seattle Children's Hospital and Research Center, University of WashingtonSeattle
| | | | | | | | | | - Rae S. M. Yeung
- The Hospital for Sick Children, University of TorontoTorontoOntarioCanada
| | - Kelly L. Brown
- University of British Columbia and BC Children's Hospital and BC Children's Hospital Research InstituteVancouverBritish ColumbiaCanada
| | - Kimberly A. Morishita
- University of British Columbia and BC Children's Hospital and BC Children's Hospital Research InstituteVancouverBritish ColumbiaCanada
| | | |
Collapse
|
14
|
Yoon T, Yoon J, Ko E, Park YB, Lee SW. Clinical perspective on serum periostin in antineutrophil-cytoplasmic antibody-associated vasculitis. Korean J Intern Med 2025; 40:512-523. [PMID: 40360226 PMCID: PMC12081101 DOI: 10.3904/kjim.2024.254] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 10/15/2024] [Accepted: 10/28/2024] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND/AIMS This study evaluated the clinical utility of serum periostin measured at diagnosis in reflecting activity at diagnosis and predicting all-cause mortality during follow-up in patients with antineutrophil cytoplasmic antibody-associated vasculitis (AAV). METHODS This study included 76 patients with AAV whose serum periostin was measured from sera collected and stored at diagnosis. The correlation of either serum periostin or the Birmingham Vasculitis Activity Score (BVAS) with other variables was evaluated. Cumulative survival rates were compared using Kaplan-Meier survival analysis. The variables at diagnosis were compared between deceased and surviving patients. Hazard ratios were obtained by Cox proportional hazard analysis. RESULTS The median age of the 76 patients was 64.0 years and 60.5% were female. The median BVAS and serum periostin were 5.0 and 10.9 ng/mL, respectively. Five of the 76 patients (6.6%) died. Serum periostin was independently correlated with cross-sectional BVAS, the Vasculitis Damage Index (VDI), white blood cell count, and serum albumin. Patients with serum periostin ≥ 15.9 ng/mL at diagnosis had a significantly lower cumulative survival rate than those without. In addition to high VDI, dyslipidaemia frequency, and C-reactive protein, deceased patients showed higher serum periostin than surviving patients. In multivariable Cox analysis, however, only dyslipidaemia rather than serum periostin was identified as an independent predictor of all-cause mortality. CONCLUSION This study is the first to demonstrate that serum periostin at diagnosis could independently reflect cross-sectional BVAS and further partially contribute to all-cause mortality prediction in patients with AAV.
Collapse
Affiliation(s)
- Taejun Yoon
- Department of Medical Science, BK21 Plus Project, Yonsei University College of Medicine, Seoul,
Korea
| | - Jiyeol Yoon
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul,
Korea
| | - Eunhee Ko
- Department of Medical Science, BK21 Plus Project, Yonsei University College of Medicine, Seoul,
Korea
| | - Yong-Beom Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul,
Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul,
Korea
| | - Sang-Won Lee
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul,
Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul,
Korea
| |
Collapse
|
15
|
Yeung EK, Kitching AR, Gingold M, Polkinghorne KR, Ryan J. Burden of Infection-Related Hospitalization and the Impact of Multidisciplinary Care in ANCA-Associated Vasculitis: A Retrospective Cohort. Int J Rheum Dis 2025; 28:e70139. [PMID: 40285430 DOI: 10.1111/1756-185x.70139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 01/10/2025] [Accepted: 02/10/2025] [Indexed: 04/29/2025]
Affiliation(s)
- Emily K Yeung
- Department of Nephrology, Monash Health, Clayton, Victoria, Australia
| | - A Richard Kitching
- Department of Nephrology, Monash Health, Clayton, Victoria, Australia
- Department of Medicine, Centre for Inflammatory Diseases, Monash University, Clayton, Victoria, Australia
- Department of Paediatric Nephrology, Monash Health, Clayton, Victoria, Australia
| | - Michael Gingold
- Department of Rheumatology, Royal Hobart Hospital, Hobart, Tasmania, Australia
- Department of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Kevan R Polkinghorne
- Department of Nephrology, Monash Health, Clayton, Victoria, Australia
- Department of Epidemiology & Preventative Medicine, Monash University, Clayton, Victoria, Australia
- Faculty of Medicine, Nursing & Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Jessica Ryan
- Department of Nephrology, Monash Health, Clayton, Victoria, Australia
- Faculty of Medicine, Nursing & Health Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
16
|
Patil H, Bharadwaj RK, Dutta N, Subramanian R, Prasad S, Mamadapur M. CAR-T cell therapy in rheumatic diseases: a review article. Clin Rheumatol 2025:10.1007/s10067-025-07451-7. [PMID: 40285991 DOI: 10.1007/s10067-025-07451-7] [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: 01/06/2025] [Revised: 03/26/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
CAR-T cell therapy, a pioneering immune-modulating treatment that was initially designed for hematologic malignancies, is now being considered a potential treatment for autoimmune and rheumatic diseases. This method involves genetically engineering T cells to express chimeric antigen receptors (CARs), allowing them to target specific antigens associated with pathogenic immune cells. The review covers the possibility of CAR-T therapy in the treatment of autoimmune diseases like systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), systemic sclerosis (SSc). The therapy's ability to maintain remission by targeting autoreactive B cells in the course of disease has been an important aspect of studies involving SLE. In refractory RA, CAR-T cells also demonstrate a potential therapeutic modality in selectively killing immune cells driving the disease process. For SSc, CAR-T therapy may represent a novel therapeutic approach because it targets the dysregulated activity of B cells as well as the fibrotic processes that drive the disease pathology. Emerging evidence suggests potential applications in conditions such as Sjögren's syndrome and dermatomyositis. While CAR-T therapy promises accuracy, persistence, and the potential for long-term remission, many problems remain, including the risk of cytokine release syndrome, immune toxicity, and treatment affordability. The development of CAR-Tregs and advanced gene-editing techniques may increase the specificity and safety of therapy. In addition, clinical trials and long-term studies should be conducted to establish the efficacy, safety, and economic feasibility of this innovative approach. This review underscores the transformative potential of CAR-T therapy in the management of rheumatic diseases, particularly in refractory cases. Offering targeted immunomodulation with a minimum of systemic immune suppression, CAR-T therapy could redefine therapeutic paradigms and offer hope for improved outcomes in autoimmune diseases.
Collapse
Affiliation(s)
| | | | | | - Ramaswamy Subramanian
- Department of Clinical Immunology and Rheumatology, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, India
| | - Shiva Prasad
- Department of Clinical Immunology and Rheumatology, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, India
| | - Mahabaleshwar Mamadapur
- Department of Clinical Immunology and Rheumatology, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, India.
| |
Collapse
|
17
|
Triaille C, Terrier B, Hadchouel A, Haddad E, Vaglio A, Frémond ML. The emerging concept of ANCA-associated vasculitis related to inborn errors of immunity. Autoimmun Rev 2025; 24:103824. [PMID: 40294837 DOI: 10.1016/j.autrev.2025.103824] [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: 02/14/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
ANCA-associated vasculitis (AAV) is a group of rare small vessels vasculitis that preferentially affect the kidneys, lungs and upper airways. Although the detailed pathophysiology remains unclear, genetic background has been shown to play a role in sporadic forms of AAV. The discovery of these susceptibility genes (and associated biological pathways) involved in AAV have shaped the current understanding of AAV pathophysiology. In addition to common genetic polymorphisms, specific rare inborn errors of immunity (IEI) have been described with a high frequency of ANCA (antineutrophil cytoplasmic antibodies) positivity and vasculitis features in young individuals (in addition to other manifestations). A systematic literature search revealed that patients with pathogenic variants in COPA, STING1, DNASE1L3, and PIK3CD are at increased risk of developing ANCA and AAV features, including alveolar hemorrhage, interstitial lung disease, pauciimmune glomerulonephritis, and upper airways involvement (septum perforation, saddle-nose deformity, chronic nasal/sinuses ulceration). Some of these IEI may also present with a mixed phenotype and/or auto-antibodies profile associating features of AAV and other autoimmune diseases (in particular systemic lupus erythematosus). Notably, a proportion of reports and series lack serological (ANCA specificity and titers) and/or histopathological data, making challenging to assess the likelihood for ANCA pathogenicity in some patients with IEI (as opposed to unspecific signs of biologic autoimmunity). This point is nonetheless essential to make appropriate therapeutic decisions. In addition, since most of the genes mentioned above are involved in the type 1 interferon signaling, the role of this pathway in AAV etiopathogenesis deserves further investigation. In this review, we will describe these IEI, their overlap with sporadic AAV, and their evocative features. Next, we will discuss how these monogenic conditions might inform our general understanding of AAV pathophysiology. We also propose some directions for future research in order to better define the link between ANCA and IEI. Finally, we will consider how making the diagnosis of an IEI in a patient with AAV features might impact individual management.
Collapse
Affiliation(s)
- Clément Triaille
- Pôle de Pathologies Rhumatismales Systémiques et Inflammatoires (RUMA), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium; Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada.
| | - Benjamin Terrier
- Department of Internal Medicine, Hospital Cochin, AP-HP, Paris, France; Université Paris Cité, Paris, France
| | - Alice Hadchouel
- AP-HP, Hôpital Universitaire Necker-Enfants Malades, Service de Pneumologie Pédiatrique, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France; INSERM U1151, Institut Necker Enfants Malades, Université Paris Cité, Paris, France
| | - Elie Haddad
- Department of Pediatrics, Department of Microbiology, Immunology and Infectious Diseases, CHU Sainte-Justine Azrieli Research Center, Université de Montréal, Montreal, Quebec, Canada
| | - Augusto Vaglio
- Nephrology and Dialysis Unit, Meyer Children's Hospital IRCCS, Firenze, Italy; Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Firenze, Firenze, Italy
| | - Marie-Louise Frémond
- Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, INSERM UMR1163, Paris, France; Paediatric Immunology-Hematology and Rheumatology Unit, Necker Hospital, APHP Centre, Université Paris-Cité, Paris, France
| |
Collapse
|
18
|
Cortinovis M, Perico N, Remuzzi G. Innovative therapeutics for renoprotection: Where we are. Pharmacol Rev 2025; 77:100060. [PMID: 40382796 DOI: 10.1016/j.pharmr.2025.100060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 05/20/2025] Open
Abstract
Chronic kidney disease (CKD) has become highly prevalent worldwide, with major implications for public health, including increased risk of progression to kidney failure, cardiovascular events, and mortality. Up to a decade ago, renin-angiotensin system inhibitors, that is angiotensin-converting enzyme inhibitors and angiotensin II type 1 receptor blockers, were the only available pharmacological interventions to slow kidney function loss and limit the associated cardiovascular morbidity and mortality in this context. More recently, landmark trials have demonstrated the ability of novel therapeutics to significantly ameliorate kidney and cardiovascular outcomes in patients with CKD, when added on top of optimized renin-angiotensin system blockade. These include sodium-glucose cotransporter-2 inhibitors in patients with diabetic and nondiabetic kidney disease, as well as the nonsteroidal mineralcorticoid receptor antagonist finerenone and the glucagon-like peptide-1 receptor agonist semaglutide in patients with diabetic kidney disease. We herein review the evolving scenario and the latest evidence for the treatment of CKD, mainly focusing on proteinuric CKD. We started with a presentation of established and more recently approved classes of kidney protective drugs, followed by a discussion of therapeutic interventions under clinical investigation to slow CKD progression. Finally, we underscore the added value of personalized and multidrug interventions, which are becoming increasingly more feasible with the availability of a growing number of kidney protective agents, and are likely to stand as the most powerful tools to safely slow, or even prevent, the progression of proteinuric CKD. SIGNIFICANCE STATEMENT: Chronic kidney disease (CKD) is highly prevalent globally, and is associated with substantial morbidity and mortality. This review provides a comprehensive overview of the currently approved and emerging therapeutic options for the treatment of proteinuric CKD. As novel kidney protective agents have recently become available, the outcomes of patients with CKD could hopefully improve over the few decades ahead.
Collapse
Affiliation(s)
- Monica Cortinovis
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Norberto Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy.
| |
Collapse
|
19
|
Nakajima A, Hokari M, Yanagimura F, Saji E, Shimizu H, Toyoshima Y, Yanagawa K, Arakawa M, Yokoseki A, Wakasugi T, Okamoto K, Watanabe K, Minato K, Otsu Y, Nozawa Y, Kobayashi D, Sanpei K, Kikuchi H, Hirohata S, Awamori K, Nawata A, Yamada M, Takahashi H, Nishizawa M, Igarashi H, Sato N, Kakita A, Onodera O, Kawachi I. Long-Term Clinical Landscapes of Spinal Hypertrophic Pachymeningitis With Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis. Neurology 2025; 104:e213420. [PMID: 40106756 PMCID: PMC11919275 DOI: 10.1212/wnl.0000000000213420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/07/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Spinal hypertrophic pachymeningitis (HP) is an extremely rare disorder characterized by the thickening of the spinal dura mater, which harbors distinct repertoires of immune cells due to the unique partitioning of the arachnoid blood-CSF barrier. The objectives were to identify the pathogenesis and therapeutic strategies for spinal HP. METHODS This retrospective cohort study analyzed the clinical and pathologic profiles of patients with idiopathic/immune-mediated HP including spinal HP. RESULTS Among 61 patients with idiopathic/immune-mediated HP, all 6 Japanese patients with spinal HP, with a median observation period of 88.8 months, were myeloperoxidase (MPO)-anti-neutrophil cytoplasmic antibody (ANCA)-seropositive. The MPO-ANCA+ spinal HP cohort had the following characteristics: (1) a predominance of older women; (2) all patients were classified as having microscopic polyangiitis based on the 2022 American College of Rheumatology/European League Against Rheumatism criteria; (3) 83% of patients developed subacute/chronic myelopathy due to extramedullary spinal cord compression; (4) 50% of patients had lesion extension to the epidural compartment and vertebral column; (5) 50% of patients presented with chronic sinusitis, otitis media, or mastoiditis; (6) 33% of patients had involvement of the lower airways or kidneys; (7) a higher disease activity of the nervous system was noted based on the Birmingham Vasculitis Activity Score (BVAS), in contrast to MPO-ANCA+ cranial HP; (8) granulomatous inflammation with myofibroblasts, immune cells including granulocytes, and B-cell follicle-like structures were observed in the thickened dura mater; (9) immunotherapies (with or without surgical decompression) were effective in reducing the modified Rankin Scale score and reduced BVAS during the first active insults; (10) combined immunotherapies with glucocorticoids and cyclophosphamide/rituximab helped in reducing relapses in the long term; and (11) surgical decompression, including laminectomy and duraplasty, was necessary for compressive myelopathy. These data suggest that MPO-ANCA+ spinal HP shares common features with MPO-ANCA+ cranial HP (1, 2, 6, 8, 9, and 10), but also has unique clinical features (3, 4, 5, 7, and 11). DISCUSSION Our findings highlight the significant pathogenic role of ANCA in spinal HP. MPO-ANCA+ spinal HP, as an organ-threatening disease, should be positioned as having unique characteristics, whether limited to the CNS or as part of a generalized form in ANCA-associated vasculitis.
Collapse
Affiliation(s)
- Akihiro Nakajima
- Department of Neurology, Brain Research Institute, Niigata University, Japan
| | - Mariko Hokari
- Department of Neurology, Brain Research Institute, Niigata University, Japan
| | - Fumihiro Yanagimura
- Department of Neurology, Brain Research Institute, Niigata University, Japan
- Department of Neurology, NHO Niigata National Hospital, Kashiwazaki, Japan
| | - Etsuji Saji
- Department of Neurology, Brain Research Institute, Niigata University, Japan
- Department of Neurology, Niigata City General Hospital, Japan
| | - Hiroshi Shimizu
- Department of Pathology, Brain Research Institute, Niigata University, Japan
| | - Yasuko Toyoshima
- Department of Pathology, Brain Research Institute, Niigata University, Japan
- Department of Neurology, Brain Disease Center, Agano Hospital, Agano, Japan
| | - Kaori Yanagawa
- Department of Neurology, Brain Research Institute, Niigata University, Japan
| | - Musashi Arakawa
- Department of Neurology, Brain Research Institute, Niigata University, Japan
- Musashi Clinic, Niigata, Japan
| | - Akiko Yokoseki
- Department of Neurology, Brain Research Institute, Niigata University, Japan
- Department of Neurology, Niigata Medical Center, Japan
| | - Takahiro Wakasugi
- Department of Neurology, Brain Research Institute, Niigata University, Japan
- Department of Neurology, NHO Nishiniigata Chuo Hospital, Niigata, Japan
| | - Kouichirou Okamoto
- Department of Neurosurgery, Brain Research Institute, Niigata University, Japan
| | - Kei Watanabe
- Department of Orthopaedic Surgery, Niigata University Medical and Dental Hospital, Japan
- Niigata Spine Surgery Center, Kameda Daiichi Hospital, Niigata, Japan
| | - Keitaro Minato
- Department of Orthopaedic Surgery, Niigata University Medical and Dental Hospital, Japan
| | - Yutaka Otsu
- Department of Neurology, Brain Research Institute, Niigata University, Japan
| | - Yukiko Nozawa
- Division of Clinical Nephrology and Rheumatology, Graduate School of Medical and Dental Sciences, Niigata University, Japan
| | - Daisuke Kobayashi
- Division of Clinical Nephrology and Rheumatology, Graduate School of Medical and Dental Sciences, Niigata University, Japan
| | | | - Hirotoshi Kikuchi
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Shunsei Hirohata
- Department of Rheumatology, Nobuhara Hospital, Tatsuno, Japan
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Sagamihara, Japan
| | | | - Aya Nawata
- Department of Pathology and Oncology, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Mitsunori Yamada
- Department of Brain Disease Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hitoshi Takahashi
- Department of Pathology, Brain Research Institute, Niigata University, Japan
- Department of Pathology and Laboratory Medicine, Niigata Neurosurgical Hospital, Japan
| | - Masatoyo Nishizawa
- Department of Neurology, Brain Research Institute, Niigata University, Japan
- Niigata University of Health and Welfare, Japan
| | - Hironaka Igarashi
- Center for Integrated Human Brain Science, Brain Research Institute, Niigata University, Japan
| | - Noboru Sato
- Division of Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Japan; and
- Medical Education Center, Graduate School of Medical and Dental Sciences, Niigata University, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Japan
| | - Izumi Kawachi
- Department of Neurology, Brain Research Institute, Niigata University, Japan
- Medical Education Center, Graduate School of Medical and Dental Sciences, Niigata University, Japan
| |
Collapse
|
20
|
Casal Moura M, Tandon YK, Hartman TE, Ryu JH, Baqir M. Interstitial lung disease in patients with antineutrophil cytoplasmic antibody-associated vasculitis: chest CT patterns and correlation with survival. Semin Arthritis Rheum 2025; 73:152726. [PMID: 40273744 DOI: 10.1016/j.semarthrit.2025.152726] [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: 02/03/2025] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Interstitial lung disease (ILD) is common in antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV), with usual interstitial pneumonia (UIP) being the most frequent pattern. The impact of different ILD patterns on clinical outcomes remains unclear. METHODS Retrospective cohort study included patients with AAV (MPA and GPA) and ILD confirmed by chest CT scans between 1997 and 2021. ILD patterns were classified according to 2018 Fleischner Society criteria. RESULTS Of 1862 patients in the Mayo AAV Cohort, 143 (7.7 %) had ILD on chest CT. The median age at the time of ILD diagnosis (occurring before AAV diagnosis in 26.6 %) was 69 years (IQR 61-75); 60 % were male, and 75 % were MPO-positive. On chest CT, "typical UIP" pattern was identified in 44 patients (30.8 %), whereas 13 (9.1 %) manifested "probable UIP" pattern, 37 (25.9 %) "indeterminate for UIP" pattern, and 49 (34.3 %) "non-UIP" pattern. Among MPO-ANCA patients, typical UIP pattern was most common (37.4 %), while non-UIP pattern was most common (58.3 %) among PR3-ANCA patients. Patients with typical UIP pattern, when compared to those with non-UIP pattern, were more commonly male (70.5 %), MPO-ANCA (90.0 %), diagnosed before the onset of AAV (40.9 %), and had reduced diffusion capacity on pulmonary function tests. The presence of typical UIP was related with higher survival at 12 months and 10 years in MPO-ANCA patients when compared to other ILD patterns (IRR 8.201 and IRR 2.179). CONCLUSIONS The typical UIP pattern in AAV-ILD is associated with better survival, particularly in MPO-ANCA patients, suggesting distinct mechanisms for ILD development in MPO vs. PR3-AAV.
Collapse
Affiliation(s)
- Marta Casal Moura
- Division of Pulmonary and Critical Care, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.
| | - Yasmeen K Tandon
- Department of Radiology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.
| | - Thomas E Hartman
- Department of Radiology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.
| | - Jay H Ryu
- Division of Pulmonary and Critical Care, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.
| | - Misbah Baqir
- Division of Pulmonary and Critical Care, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.
| |
Collapse
|
21
|
Tang Y, Cao Q, Liu J, Zhuang Q. Immune Landscape Variation in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis Circulation Before and After Plasmapheresis by Single-Cell Transcriptome. Mediators Inflamm 2025; 2025:5531382. [PMID: 40256686 PMCID: PMC12006691 DOI: 10.1155/mi/5531382] [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: 07/29/2024] [Accepted: 03/08/2025] [Indexed: 04/22/2025] Open
Abstract
Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is a group of autoimmune diseases characterized by inflammation and destruction of small blood vessels. AAV could be fatal if left untreated. Prompt diagnosis and treatment are crucial to protect AAV-related organs and tissue. Plasmapheresis, a therapeutic intervention aimed at removing harmful substances from the blood, devotes benefits to AAV treatment. However, the specific immune mechanism underlying its effectiveness remains unclear. In our research, we used single-cell RNA sequencing (scRNA-seq) to study the variation of peripheral blood mononuclear cells (PBMCs) before and after plasmapheresis in AAV patients. From this work, we explored a novel method for monocyte classification. In addition, flow cytometry was used to detect the relationship between the monocyte clusters and AAV activity under the new monocyte clustering method. Our scRNA-seq results revealed significant changes in monocyte clusters following treatment, which could be classified into three clusters (CD14+ monocytes, FCGR1A+ monocytes, and FCGR3A+ monocytes). In addition, our flow cytometry results showed that FCGR3A+ (CD16+) monocytes were positively correlated with AAV activity, whereas FCGR1A+ (CD16-CD64+) monocytes were negatively correlated with AAV activity. This may be related to the different biological effects of CD16 and CD64 on monocytes after interacting with the Fc region of ANCAs. In conclusion, our research sheds light on the immune landscape of AAV before and after plasmapheresis, identifying specific monocyte clusters linked to disease activity. These findings offer insights for novel monitoring methods and therapeutic targets in AAV.
Collapse
Affiliation(s)
- Youzhou Tang
- Department of Nephropathy and Rheumatology, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Critical Kidney Disease Research Center, Central South University, Changsha, Hunan, China
| | - Qingtai Cao
- Transplantation Center, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jishi Liu
- Department of Nephropathy and Rheumatology, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Quan Zhuang
- Transplantation Center, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Center of National Health Ministry on Transplantation Medicine, Changsha, Hunan, China
| |
Collapse
|
22
|
Nishioka R, Omura S, Nakagomi D, Abe Y, Kadoya M, Takizawa N, Nomura A, Kukida Y, Kondo N, Yamano Y, Yanagida T, Endo K, Hirata S, Matsui K, Takeuchi T, Ichinose K, Kato M, Yanai R, Matsuo Y, Shimojima Y, Okazaki R, Takata T, Ito T, Moriyama M, Takatani A, Miyawaki Y, Ito-Ihara T, Kawaguchi T, Yajima N, Kida T, Kawahito Y, Mizushima I. Glucocorticoid tapering pace in microscopic polyangiitis and granulomatosis with polyangiitis in Japanese real-world practice: A propensity score-matched retrospective cohort study from the J-CANVAS registry. Mod Rheumatol 2025; 35:496-504. [PMID: 39716472 DOI: 10.1093/mr/roae112] [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: 08/19/2024] [Revised: 11/21/2024] [Accepted: 12/23/2024] [Indexed: 12/25/2024]
Abstract
OBJECTIVE To assess the prevalence and outcomes among regimens of glucocorticoid tapering for microscopic polyangiitis (MPA) and granulomatosis with polyangiitis (GPA) in real-world practice. METHODS We retrospectively examined the Japan Collaborative Registry of Antineutrophil Cytoplasmic Antibodies-associated Vasculitis (J-CANVAS) registry, and evaluated the prevalence of glucocorticoid tapering regimens in the PEXIVAS trial. In patients with newly diagnosed MPA and GPA, we compared outcomes among standard and reduced pace regimens. Relapse-free survival rates were compared after propensity score matching. RESULTS Of 364 eligible patients, 113 (31.0%) received standard tapering and 251 slower tapering. After matching, 87 pairs no significant difference was observed in relapse-free survival (P = .506). Regarding the reduced regimen, there were so few patients (14/364, 3.8%) that statistical analysis was not performed. CONCLUSIONS The glucocorticoid tapering for MPA and GPA in Japanese real-world practice was found to be generally slower than the standard regimen revealing a huge evidence-practice gap. Additionally, slower tapering did not improve relapse-free survival and might cause unnecessary glucocorticoid exposure.
Collapse
Affiliation(s)
- Ryo Nishioka
- Department of Nephrology and Rheumatology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
- Department of General Medicine, Kanazawa University Hospital, Kanazawa, Japan
| | - Satoshi Omura
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daiki Nakagomi
- Department of Rheumatology, University of Yamanashi Hospital, Yamanashi, Japan
| | - Yoshiyuki Abe
- Department of Internal Medicine and Rheumatology, Juntendo University, Tokyo, Japan
| | - Masatoshi Kadoya
- Center for Rheumatic Disease, Japanese Red Cross Society Kyoto Daiichi Hospital, Kyoto, Japan
| | - Naoho Takizawa
- Department of Rheumatology, Chubu Rosai Hospital, Nagoya, Japan
| | - Atsushi Nomura
- Immuno-Rheumatology Center, St. Luke's International Hospital, Tokyo, Japan
| | - Yuji Kukida
- Department of Rheumatology, Japanese Red Cross Society Kyoto Daini Hospital, Kyoto, Japan
| | - Naoya Kondo
- Department of Nephrology, Kyoto Katsura Hospital, Kyoto, Japan
| | - Yasuhiko Yamano
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, Seto, Japan
| | - Takuya Yanagida
- Department of Hematology and Rheumatology, Kagoshima University Hospital, Kagoshima, Japan
| | - Koji Endo
- Department of General internal medicine, Tottori Prefectural Central Hospital, Tottori-shi, Japan
| | - Shintaro Hirata
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Kiyoshi Matsui
- Department of Diabetes, Endocrinology and Clinical Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Tohru Takeuchi
- Department of Internal Medicine (IV), Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Kunihiro Ichinose
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Rheumatology, Shimane University, Faculty of Medicine, Izumo, Japan
| | - Masaru Kato
- Department of Rheumatology, Endocrinology and Nephrology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Ryo Yanai
- Division of Rheumatology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yusuke Matsuo
- Department of Rheumatology, Tokyo Kyosai Hospital, Kyoto, Japan
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yasuhiro Shimojima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Ryota Okazaki
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Tottori-shi, Japan
| | - Tomoaki Takata
- Division of Gastroenterology and Nephrology, Tottori University, Tottori-shi, Japan
| | - Takafumi Ito
- Division of Nephrology, Shimane University Hospital, Izumo, Japan
| | - Mayuko Moriyama
- Department of Rheumatology, Shimane University, Faculty of Medicine, Izumo, Japan
| | - Ayuko Takatani
- Rheumatic Disease Center, Sasebo Chuo Hospital, Sasebo, Japan
| | - Yoshia Miyawaki
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshiko Ito-Ihara
- The Clinical and Translational Research Center, University Hospital, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takashi Kawaguchi
- Department of Practical Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Nobuyuki Yajima
- Division of Rheumatology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
- Department of Healthcare Epidemiology, School of Public Health in the Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Center for Innovative Research for Communities and Clinical Excellence, Fukushima Medical University, Fukushima, Japan
| | - Takashi Kida
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yutaka Kawahito
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ichiro Mizushima
- Department of Nephrology and Rheumatology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
23
|
Noda H, Isogai S, Naniwa T. Granulomatosis with polyangiitis presenting with isolated external genital and urethral manifestations: a case-based review. Rheumatol Int 2025; 45:89. [PMID: 40183813 DOI: 10.1007/s00296-025-05837-z] [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: 01/07/2025] [Accepted: 03/18/2025] [Indexed: 04/05/2025]
Abstract
Granulomatosis with polyangiitis (GPA) is a systemic necrotizing vasculitis primarily affecting the respiratory tract and kidneys, with external genital and urethral lesions (EGUL) being exceedingly rare. We present a case of a middle-aged man with relapsing proteinase 3-antineutrophil antibody-positive GPA who developed isolated granulomatous, ulcerative balanitis, and urethritis. His condition abruptly worsened after a prolonged indolent course, requiring treatment with glucocorticoids and rituximab, leading to successful remission. To better characterize EGUL in GPA, a systematic literature search was performed in PubMed, Scopus, and the NPO Japanese Society of Medical Abstracts databases using keywords related to GPA and EGUL. Cases meeting the American College of Rheumatology or Japanese Ministry of Health, Labor, and Welfare criteria for GPA and the 2012 Chapel Hill Consensus Conference definitions were included for analysis. Our review identified that EGUL often presents as an initial symptom and can be classified by the presence or absence of preceding urethritis. Cases with preceding urethritis had a higher risk of severe complications with extensive penile or urethral involvement. In contrast, penile lesions without preceding urethritis typically presented as characteristic mucosal lesions localized around the glans. In females, GPA-associated urethritis frequently led to periurethral mass formation, vaginal involvement, and significant sequelae. Given the potential for delayed diagnosis and serious complications, clinicians should be vigilant for EGUL in patients with suspected or diagnosed GPA. Future prospective studies with larger cohorts are needed to elucidate the prevalence, clinical spectrum, and optimal management of these rare but significant manifestations of GPA.
Collapse
Affiliation(s)
- Haruka Noda
- Division of Rheumatology, Department of Internal Medicine, Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Hospital, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Shuntaro Isogai
- Division of Rheumatology, Department of Internal Medicine, Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Hospital, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Taio Naniwa
- Division of Rheumatology, Department of Internal Medicine, Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Hospital, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan.
| |
Collapse
|
24
|
Sahinoğlu I, Uslu S, Güc U, Can F, Ucar M, Tan A, Soysal Gündüz O. Case Report: Mitral Valve Chordae Tendineae Rupture and Splenic Infarction in Granulomatosis With Polyangiitis. Int J Rheum Dis 2025; 28:e70214. [PMID: 40207590 DOI: 10.1111/1756-185x.70214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/16/2025] [Accepted: 03/26/2025] [Indexed: 04/11/2025]
Affiliation(s)
- Irem Sahinoğlu
- Department of Internal Medicine, Division of Rheumatology, Celal Bayar University School of Medicine, Manisa, Turkey
| | - Sadettin Uslu
- Department of Internal Medicine, Division of Rheumatology, Celal Bayar University School of Medicine, Manisa, Turkey
| | - Ulkü Güc
- Department of Internal Medicine, Celal Bayar University School of Medicine, Manisa, Turkey
| | - Fatma Can
- Department of Radiology, Celal Bayar University School of Medicine, Manisa, Turkey
| | - Mustafa Ucar
- Department of Cardiology, Celal Bayar University School of Medicine, Manisa, Turkey
| | - Ayca Tan
- Department of Pathology, Celal Bayar University School of Medicine, Manisa, Turkey
| | - Ozgül Soysal Gündüz
- Department of Internal Medicine, Division of Rheumatology, Celal Bayar University School of Medicine, Manisa, Turkey
| |
Collapse
|
25
|
Kwon J, Choi YW, Kim H, Yoo SJ. Thoracic Manifestations of ANCA-associated Vasculitis: Review of the 2022 American College of Rheumatology-European Alliance of Associations of Rheumatology Classification Criteria. Radiographics 2025; 45:e240089. [PMID: 40146625 DOI: 10.1148/rg.240089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Antineutrophil cytoplasmic autoantibody (ANCA)-associated vasculitis (AAV) is a rare disease that manifests as necrotizing vasculitis involving small vessels without immune complex deposition. Granulomatosis with polyangiitis (GPA), eosinophilic granulomatosis with polyangiitis (EGPA), and microscopic polyangiitis (MPA) are included in this disease entity. Diagnosis and differentiation of AAV is challenging because of the diverse and overlapping clinical manifestations and lack of pathognomonic findings. Therefore, AAV classification criteria have been developed to increase the likelihood of diagnosis using multidisciplinary approaches, including clinical, radiologic, laboratory, and pathologic findings. The new American College of Rheumatology and European Alliance of Associations for Rheumatology classification criteria were released in 2022 to classify AAVs using weighted criteria and threshold scores. They are expected to make the classification of GPA, EGPA, and MPA more accurate in the setting of suspected small-vessel vasculitis. The criteria present key thoracic imaging discriminators of GPA as "pulmonary nodules, masses, or cavitation" and MPA as "interstitial fibrosis," whereas, radiologic criteria of EGPA are not present. ANCA positivity and eosinophil count are included as key laboratory discriminators. It is essential for radiologists to familiarize themselves with imaging findings of each AAV and know the key imaging discriminators to aid in the differential diagnosis of AAVs. By reviewing the radiologic findings of thoracic manifestations of each AAV and applying the new criteria in a series of cases, the authors aim to provide a practical and stepwise approach to AAV for radiologists. ©RSNA, 2025 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Jonghyeon Kwon
- From the Departments of Radiology (J.K., Y.W.C., S.J.Y.) and Pathology (H.K.), Hanyang University Medical Center, Hanyang University College of Medicine, 222 Wangsimni-ro, Seongdone-gu, Seoul 04763, Republic of Korea
| | - Yo Won Choi
- From the Departments of Radiology (J.K., Y.W.C., S.J.Y.) and Pathology (H.K.), Hanyang University Medical Center, Hanyang University College of Medicine, 222 Wangsimni-ro, Seongdone-gu, Seoul 04763, Republic of Korea
| | - Hyunsung Kim
- From the Departments of Radiology (J.K., Y.W.C., S.J.Y.) and Pathology (H.K.), Hanyang University Medical Center, Hanyang University College of Medicine, 222 Wangsimni-ro, Seongdone-gu, Seoul 04763, Republic of Korea
| | - Seung-Jin Yoo
- From the Departments of Radiology (J.K., Y.W.C., S.J.Y.) and Pathology (H.K.), Hanyang University Medical Center, Hanyang University College of Medicine, 222 Wangsimni-ro, Seongdone-gu, Seoul 04763, Republic of Korea
| |
Collapse
|
26
|
Wang L, Novoa-Laurentiev J, Cook C, Srivatsan S, Hua Y, Yang J, Miloslavsky E, Choi HK, Zhou L, Wallace ZS. Identification of an ANCA-associated vasculitis cohort using deep learning and electronic health records. Int J Med Inform 2025; 196:105797. [PMID: 39864108 DOI: 10.1016/j.ijmedinf.2025.105797] [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: 06/13/2024] [Revised: 12/16/2024] [Accepted: 01/12/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND ANCA-associated vasculitis (AAV) is a rare but serious disease. Traditional case-identification methods using claims data can be time-intensive and may miss important subgroups. We hypothesized that a deep learning model analyzing electronic health records (EHR) can more accurately identify AAV cases. METHODS We examined the Mass General Brigham (MGB) repository of clinical documentation from 12/1/1979 to 5/11/2021, using expert-curated keywords and ICD codes to identify a large cohort of potential AAV cases. Three labeled datasets (I, II, III) were created, each containing note sections. We trained and evaluated a range of machine learning and deep learning algorithms for note-level classification, using metrics like positive predictive value (PPV), sensitivity, F-score, area under the receiver operating characteristic curve (AUROC), and area under the precision and recall curve (AUPRC). The hierarchical attention network (HAN) was further evaluated for its ability to classify AAV cases at the patient-level, compared with rule-based algorithms in 2000 randomly chosen samples. RESULTS Datasets I, II, and III comprised 6000, 3008, and 7500 note sections, respectively. HAN achieved the highest AUROC in all three datasets, with scores of 0.983, 0.991, and 0.991. The deep learning approach also had among the highest PPVs across the three datasets (0.941, 0.954, and 0.800, respectively). In a test cohort of 2000 cases, the HAN model achieved a PPV of 0.262 and an estimated sensitivity of 0.975. Compared to the best rule-based algorithm, HAN identified six additional AAV cases, representing 13% of the total. CONCLUSION The deep learning model effectively classifies clinical note sections for AAV diagnosis. Its application to EHR notes can potentially uncover additional cases missed by traditional rule-based methods.
Collapse
Affiliation(s)
- Liqin Wang
- Division of General Internal Medicine and Primary Care, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School Boston MA USA.
| | - John Novoa-Laurentiev
- Division of General Internal Medicine and Primary Care, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School Boston MA USA.
| | - Claire Cook
- Rheumatology and Allergy Clinical Epidemiology Research Center and Division of Rheumatology, Allergy, and Immunology, and Mongan Institute, Department of Medicine, Massachusetts General Hospital Boston MA USA.
| | - Shruthi Srivatsan
- Rheumatology and Allergy Clinical Epidemiology Research Center and Division of Rheumatology, Allergy, and Immunology, and Mongan Institute, Department of Medicine, Massachusetts General Hospital Boston MA USA.
| | - Yining Hua
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, USA.
| | - Jie Yang
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School Boston MA USA.
| | - Eli Miloslavsky
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital and Harvard Medical School Boston MA USA.
| | - Hyon K Choi
- Rheumatology and Allergy Clinical Epidemiology Research Center and Division of Rheumatology, Allergy, and Immunology, and Mongan Institute, Department of Medicine, Massachusetts General Hospital Boston MA USA.
| | - Li Zhou
- Division of General Internal Medicine and Primary Care, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School Boston MA USA.
| | - Zachary S Wallace
- Rheumatology and Allergy Clinical Epidemiology Research Center and Division of Rheumatology, Allergy, and Immunology, and Mongan Institute, Department of Medicine, Massachusetts General Hospital Boston MA USA.
| |
Collapse
|
27
|
Ashida C, Nozaki Y, Li J, Akazawa H, Kishimoto K, Kinoshita K, Matsumura I. Urinary Kim-1 Correlates with Interstitial Nephritis Activity in Patients with Microscopic Polyangiitis. Curr Issues Mol Biol 2025; 47:196. [PMID: 40136450 PMCID: PMC11941514 DOI: 10.3390/cimb47030196] [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: 12/30/2024] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Microscopic polyangiitis (MPA) is a type of necrotizing vasculitis that primarily affects small vessels and belongs to the spectrum of anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAVs). While previous studies have identified potential prognostic biomarkers, further research is needed to validate a reliable marker for risk stratification in clinical practice. Kidney injury molecule-1 (Kim-1), a transmembrane protein expressed on proximal tubular epithelial cells, has been implicated in tubular damage. This study investigated the potential of Kim-1 as a biomarker in MPA. METHODS Kidney biopsy tissues, along with urine and blood samples, were retrospectively analyzed from 52 MPA patients and compared to urine samples from 7 healthy controls. Global disease activity was assessed using the Birmingham vasculitis activity score (BVAS) and vasculitis damage index, while renal disease activity was evaluated using renal BVAS (BVAS-R). RESULTS Urinary Kim-1 levels were significantly elevated in MPA patients compared to healthy controls. Urinary Kim-1 was positively correlated with the Mayo Clinic Chronicity Score (MCCS) but not with the ANCA Kidney Risk Score (AKRiS), whereas tubular Kim-1 was associated with AKRiS but not with MCCS, indicating their distinct pathological significance. Higher tubular Kim-1 expression was observed in patients with elevated BVAS-R. Urinary Kim-1 levels correlated with proteinuria and were associated with the Mayo Clinic Chronicity Score (MCCS) and ANCA Kidney Risk Score (AKRiS) but not with glomerular lesion severity. Unlike C-reactive protein (CRP), neither urinary nor tubular Kim-1 predicted MPA recurrence. CONCLUSIONS Urinary Kim-1 reflects histopathologic findings and renal impairment but does not predict systemic disease activity or recurrence in MPA, demonstrating its potential clinical utility as a biomarker for assessing chronic renal damage.
Collapse
Affiliation(s)
- Chisato Ashida
- Department of Hematology and Rheumatology, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan; (J.L.); (H.A.); (K.K.); (K.K.); (I.M.)
| | - Yuji Nozaki
- Department of Hematology and Rheumatology, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan; (J.L.); (H.A.); (K.K.); (K.K.); (I.M.)
| | | | | | | | | | | |
Collapse
|
28
|
Juto A, Martin M, Björk A, Padyukov L, Grönwall C, Antovic A, Bruchfeld A, Gunnarsson I, Blom AM. Association of C4d with disease activity in anti-neutrophil cytoplasmic antibody-associated vasculitis: evidence for classical/lectin complement pathway activation. Arthritis Res Ther 2025; 27:49. [PMID: 40045390 PMCID: PMC11881377 DOI: 10.1186/s13075-025-03503-0] [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: 11/25/2024] [Accepted: 02/10/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND We aimed to investigate the involvement of the classical/lectin complement pathway in anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) by exploring the complement activation fragment C4d in association to AAV activity. METHODS Forty patients with active AAV and twenty population-based controls were included. The study included 27 (67.5%) patients with a diagnosis of GPA and 13 (32.5%) with MPA. Twenty-four patients (60%) were anti-proteinase 3 (PR3)-ANCA positive and 16 (40%) anti-myeloperoxidase (MPO)-ANCA positive. Thirty-three (82.5%) patients had kidney involvement. A follow-up sample obtained after induction therapy (median 6 months) was available for 24 of the patients, of whom 20 were in remission. Plasma C4d was analysed by ELISA detecting an epitope that arises upon complement-mediated cleavage. Plasma complement factor 4 (C4) and the soluble terminal complement complex (sTCC) were analysed by ELISA. The C4d/C4 ratio was calculated. HLA-DRB1-typing and immunohistochemistry for C4d in kidney biopsies were performed. RESULTS Patients with active AAV had higher C4d, sTCC levels and C4d/C4 ratio than controls (p < 0.001, p = 0.004, p < 0.001). C4d, sTCC levels and C4d/C4 ratio all decreased from active disease to remission (p = 0.010, p = 0.009, p = 0.011). C4d levels in AAV patients in remission remained higher than population-based controls (p = 0.026). Active anti-PR3-ANCA patients had higher C4d levels and C4d/C4 ratio than anti-MPO-ANCA patients (p = 0.001, p = 0.007). Patients with active AAV and kidney involvement had lower C4d levels than patients without (p = 0.04). C4d levels and C4d/C4 ratio correlated positively with the percentage of normal glomeruli in kidney biopsies. The immunohistochemistry was negative for C4d in kidney biopsies. CONCLUSIONS The specific C4d assay revealed activity in the classical/lectin complement pathway in AAV, which reflected general disease activity, but was not associated specifically with kidney involvement. C4d levels differed depending on anti-PR3/MPO-ANCA subtypes suggesting differences in complement activation and underlying pathogenetic mechanisms. The findings imply that the classical/lectin complement pathway may play a more significant role in AAV pathogenesis than previously reported and that plasma C4d levels and C4d/C4 ratio may be biomarker candidates for disease activity and treatment outcome monitoring.
Collapse
Affiliation(s)
- Anna Juto
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden.
| | - Myriam Martin
- Department of Translational Medicine, Section of Medical Protein Chemistry, Lund University, Lund, Sweden
- Department of Clinical Chemistry and Pharmacology, Office for Medical Services, Region Skåne, Sweden
| | - Albin Björk
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center for Rheumatology, Academic Specialist Center, Stockholm, Sweden
| | - Leonid Padyukov
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Caroline Grönwall
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Aleksandra Antovic
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Annette Bruchfeld
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Department of Renal Medicine, Karolinska University Hospitaland, CLINTEC Karolinska Institutet, Stockholm, Sweden
| | - Iva Gunnarsson
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Anna M Blom
- Department of Translational Medicine, Section of Medical Protein Chemistry, Lund University, Lund, Sweden
- Department of Clinical Chemistry and Pharmacology, Office for Medical Services, Region Skåne, Sweden
| |
Collapse
|
29
|
Colina M, Campana G. Precision Medicine in Rheumatology: The Role of Biomarkers in Diagnosis and Treatment Optimization. J Clin Med 2025; 14:1735. [PMID: 40095875 PMCID: PMC11901317 DOI: 10.3390/jcm14051735] [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: 01/17/2025] [Revised: 02/23/2025] [Accepted: 02/24/2025] [Indexed: 03/19/2025] Open
Abstract
Rheumatic diseases encompass a wide range of autoimmune and inflammatory disorders, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), psoriatic arthritis (PsA), and systemic sclerosis (SSc). These conditions often result in chronic pain, disability, and reduced quality of life, with unpredictable disease courses that may lead to joint destruction, organ damage, or systemic complications. Biomarkers, defined as measurable indicators of biological processes or conditions, have the potential to transform clinical practice by improving disease diagnosis, monitoring, prognosis, and treatment decisions. While significant strides have been made in identifying and validating biomarkers in rheumatic diseases, challenges remain in their standardization, clinical utility, and integration into routine practice. This review provides an overview of the current state of biomarkers in rheumatic diseases, their roles in clinical settings, and the emerging advancements in the field.
Collapse
Affiliation(s)
- Matteo Colina
- Rheumatology Service, Section of Internal Medicine, Department of Medicine and Oncology, Ospedale Santa Maria della Scaletta, 40026 Imola, Italy
| | - Gabriele Campana
- Alma Mater Studiorum, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy;
| |
Collapse
|
30
|
Goldman S, Wilson B, Awan K, White T, Williams M. A Diagnostic Dilemma: Proteinase-3 (PR3)-Positive Anti-neutrophil Cytoplasmic Autoantibodies in Eosinophilic Granulomatosis With Polyangiitis. Cureus 2025; 17:e80365. [PMID: 40206908 PMCID: PMC11981696 DOI: 10.7759/cureus.80365] [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] [Accepted: 03/09/2025] [Indexed: 04/11/2025] Open
Abstract
Eosinophilic granulomatosis with polyangiitis (EGPA) is a multifaceted diseased vasculitis typically associated with myeloperoxidase-perinuclear-anti-neutrophil cytoplasmic antibody (+MPO-P-ANCA). Although rare, the diagnosis should be considered in patients with difficult-to-control or late-onset asthma and extrathoracic disease. We present the case of a 37-year-old male with adult-onset asthma and chronic rhinitis hospitalized with pancreatitis and hypoxemia. Blood investigations demonstrated eosinophilia with elevated lipase, and bronchoscopy demonstrated multiple endobronchial lesions with elevated eosinophils on bronchoalveolar lavage. In addition, labs showed proteinase 3-specific antineutrophil cytoplasmic antibody (PR3-C-ANCA) autoantibodies, and the patient was diagnosed with EGPA. Additional differential diagnoses of parasitic infection, sarcoidosis, chronic eosinophilic pneumonia, and granulomatosis with polyangiitis (GPA) were entertained, but ultimately, the multisystemic involvement, anti-neutrophil cytoplasmic autoantibody (ANCA) positivity, bronchoscopy, and imaging findings clinched the diagnosis. This case highlights the spectrum of possible EGPA presentations and a rare case of PR3-C-ANCA with gastrointestinal manifestations.
Collapse
Affiliation(s)
- Samuel Goldman
- Internal Medicine, Walter Reed National Military Medical Center, Bethesda, USA
| | - Benjamin Wilson
- Pulmonary Medicine, Walter Reed National Military Medical Center, Bethesda, USA
| | - Kanwal Awan
- Internal Medicine, Walter Reed National Military Medical Center, Bethesda, USA
| | - Tara White
- Pulmonary Medicine, Walter Reed National Military Medical Center, Bethesda, USA
| | - Matthias Williams
- Pulmonary Medicine, Walter Reed National Military Medical Center, Bethesda, USA
| |
Collapse
|
31
|
Minopoulou I, Wilhelm A, Albach F, Kleyer A, Wiebe E, Schallenberg S, Fleischmann A, Frick M, Damm F, Gogolok J, Serve S, Locher BN, Borie D, Casteleyn V, Biesen R, Dörner T, Alexander T, Zernicke J, Movassaghi K, Hütter-Krönke ML, Schrezenmeier E, Schreiber A, Schneider U, Bullinger L, Krönke G, Penack O, Simon D. Anti-CD19 CAR T cell therapy induces antibody seroconversion and complete B cell depletion in the bone marrow of a therapy-refractory patient with ANCA-associated vasculitis. Ann Rheum Dis 2025; 84:e4-e7. [PMID: 39893102 DOI: 10.1016/j.ard.2025.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/25/2024] [Accepted: 12/10/2024] [Indexed: 02/04/2025]
Affiliation(s)
- Ioanna Minopoulou
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Artur Wilhelm
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany; Deutsches Rheuma-Forschungszentrum Berlin, Berlin, Germany
| | - Fredrik Albach
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Arnd Kleyer
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Edgar Wiebe
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Simon Schallenberg
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Anja Fleischmann
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Mareike Frick
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Frederik Damm
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Julia Gogolok
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Sebastian Serve
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Benjamin Nick Locher
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | | | - Vincent Casteleyn
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Robert Biesen
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas Dörner
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany; Deutsches Rheuma-Forschungszentrum Berlin, Berlin, Germany
| | - Tobias Alexander
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany; Deutsches Rheuma-Forschungszentrum Berlin, Berlin, Germany
| | - Jan Zernicke
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kamran Movassaghi
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Marie Luise Hütter-Krönke
- Department of Hematology, Oncology and Cancer Immunology, Charité - Universitätsmedizin Berlin, Campus Steglitz, Berlin, Germany
| | - Eva Schrezenmeier
- Deutsches Rheuma-Forschungszentrum Berlin, Berlin, Germany; Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin, Berlin, Germany
| | - Adrian Schreiber
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin, Berlin, Germany
| | - Udo Schneider
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany; Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Gerhard Krönke
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany; Deutsches Rheuma-Forschungszentrum Berlin, Berlin, Germany; Department of Internal Medicine 3, University of Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), University of Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Olaf Penack
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - David Simon
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany; Department of Internal Medicine 3, University of Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), University of Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany.
| |
Collapse
|
32
|
Dunlap C, Zhao N, Ertl LS, Schall TJ, Sullivan KMC. C5aR expression in kidney tubules, macrophages and fibrosis. J Histotechnol 2025; 48:27-45. [PMID: 39607065 DOI: 10.1080/01478885.2024.2430041] [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: 06/20/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
The anaphylatoxin C5a and its receptor C5aR (CD88) are complement pathway effectors implicated in renal diseases, including ANCA-associated vasculitis. We investigated the kidney expression of C5aR and a second C5a receptor C5L2 by using immunohistochemistry, in situ hybridization, and spatial gene expression on formalin-fixed, paraffin-embedded human and mouse kidney. C5aR was detected on interstitial macrophages and in multiple tubular regions, including distal and proximal; C5L2 had a similar expression pattern. The 5/6 nephrectomy model of chronic kidney injury exhibited increased C5aR expression by infiltrating cells within the fibrotic regions. C5aR expression was confirmed on human leukocytes and in vitro differentiated macrophages by flow cytometry, and treatment with C5a induced the expression of chemokines and remodeling factors by macrophages, including CCL-3/-4/-7, -20, MMP-1/-3/-8/-12, and F3, and promoted leukocyte survival. C5a activity was C5aR dependent, as demonstrated by reversal with the C5aR inhibitor avacopan. Collectively, these results suggest that myeloid C5aR may induce excessive inflammation in the kidney via immune cell recruitment, extracellular matrix destruction, and remodeling, resulting in fibrotic tissue deposition.
Collapse
Affiliation(s)
- Carolyn Dunlap
- Department of Biology, ChemoCentryx Inc., San Carlos, CA, USA
| | - Niky Zhao
- Department of Biology, ChemoCentryx Inc., San Carlos, CA, USA
| | - Linda S Ertl
- Department of Biology, ChemoCentryx Inc., San Carlos, CA, USA
| | - Thomas J Schall
- Department of Biology, ChemoCentryx Inc., San Carlos, CA, USA
| | | |
Collapse
|
33
|
Sandel NJ, Wielzen H. Case report: Testicular manifestation of ANCA vasculitis. Urol Case Rep 2025; 59:102966. [PMID: 39981501 PMCID: PMC11840479 DOI: 10.1016/j.eucr.2025.102966] [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: 12/01/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/22/2025] Open
Abstract
ANCA-associated vasculitis is a rare autoimmune disorder affecting small to medium-sized vessels, often targeting the respiratory tract and kidneys. Testicular involvement is rare and can resemble malignancy, leading to unnecessary surgery. A 36-year-old male presented with painful fingers, oral and nasal ulcers, and knee arthritis. Elevated proteinase 3 (PR3) antibodies confirmed PR3 ANCA-associated vasculitis. During hospitalization, patient developed testicular pain, and an ultrasound raised suspicion of malignancy. An inguinal orchidectomy was performed, revealing inflammation consistent with vasculitis, but no malignancy. Testicular involvement in ANCA vasculitis can mimic cancer, and increased awareness may help prevent unnecessary surgical procedures.
Collapse
Affiliation(s)
- Noah J. Sandel
- Department of Urology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Henry Wielzen
- Department of Urology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
34
|
Odler B, Riedl R, Geetha D, Szpirt WM, Hawley C, Uchida L, Wallace ZS, Walters G, Muso E, Tesar V, Pusey CD, Little MA, Merkel PA, Walsh M, Jayne DRW, Kronbichler A. The effects of plasma exchange and glucocorticoids on early kidney function among patients with ANCA-associated vasculitis in the PEXIVAS trial. Kidney Int 2025; 107:558-567. [PMID: 39708998 DOI: 10.1016/j.kint.2024.11.029] [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: 04/21/2024] [Revised: 11/13/2024] [Accepted: 11/22/2024] [Indexed: 12/23/2024]
Abstract
Therapeutic plasma exchange (PLEX) is an adjunctive treatment for patients with anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis and kidney involvement. Little is known about the effect of PLEX on early changes in kidney function. This post-hoc analysis of the PEXIVAS trial investigated the effects of PLEX on changes in kidney function within 12 months. PEXIVAS was a randomized controlled trial recruiting 691 patients with ANCA-associated glomerulonephritis, of whom 349 underwent PLEX and 342 received no-PLEX. The primary outcomes of this post hoc study of PEXIVAS were change in estimated glomerular filtration rate (eGFR) from baseline and recovery of kidney function (defined as eGFR increase of 15ml/min/1.73m2 or more). Baseline eGFR was 21.7 ± 20.3 and 20.6 ± 18.7 ml/min/1.73m2 in the PLEX and no-PLEX groups, respectively. Mean improvements in eGFR at weeks two, four, and eight after initiation of therapy were greater for the PLEX vs. the no-PLEX groups. The greatest significant difference in recovery of kidney function in the PLEX compared to the no-PLEX groups was at week four (relative risk (RR): 1.41; 95% confidence interval:1.09-1.82). Increased eGFR or recovery of kidney function at week four were significantly associated with lower risk for end-stage kidney disease at week 52 (RR: 0.96: 0.95-0.97, and RR: 0.29: 0.16-0.52; respectively). Neither changes in eGFR nor recovery of kidney function differed by reduced- compared to standard-dose glucocorticoid group. Overall, our study indicates that PLEX improves early kidney function in patients with ANCA-associated glomerulonephritis.
Collapse
Affiliation(s)
- Balazs Odler
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Regina Riedl
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria
| | - Duvuru Geetha
- Division of Nephrology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Wladimir M Szpirt
- Department of Nephrology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Carmel Hawley
- The Australasian Kidney Trials Network (AKTN), Centre for Health Services Research, University of Queensland, Brisbane, Australia
| | - Lisa Uchida
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Zachary S Wallace
- Division of Rheumatology, Allergy, and Immunology, Mongan Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Giles Walters
- Department of Renal Medicine, Canberra Hospital, Canberra, Australian Capital Territory, Australia
| | - Eri Muso
- Department of Nephrology and Dialysis, Medical Research Institute Kitano Hospital, PIIF Tazuke-Kofukai, Osaka, Japan
| | - Vladimir Tesar
- Department of Nephrology, 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Charles D Pusey
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Mark A Little
- Trinity Kidney Centre, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Peter A Merkel
- Division of Rheumatology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Division of Epidemiology, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael Walsh
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada; Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - David R W Jayne
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Andreas Kronbichler
- Department of Internal Medicine IV, Nephrology and Hypertension, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
35
|
McGovern DP, Jones RB, Jayne DRW, Smith RM. The Expanding Antineutrophil Cytoplasmic Antibody-Associated Vasculitis Armamentarium. Drugs 2025; 85:325-341. [PMID: 39969779 DOI: 10.1007/s40265-024-02143-z] [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: 12/15/2024] [Indexed: 02/20/2025]
Abstract
The complex pathophysiology of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAV) is reflected in the heterogeneity of the presenting clinical syndromes caused by these diseases but also provides a variety of conceivable molecular and cellular targets that can be therapeutically manipulated. The last decade has seen an expansion of established and potential therapies for treating AAV, some of which target the dysfunctional autoreactive immune response and others aim to ameliorate the downstream consequences of local vascular inflammation and necrosis. The success and widespread adoption of the anti-CD20 monoclonal antibody, rituximab, as an agent to both induce and maintain remission, has heralded a change in the standard-of-care management of AAV, replacing the "old guard" combination of cyclophosphamide and high-dose corticosteroids established in the 1970s. The development and approval of avacopan, a first-in-class small-molecule antagonist to the main receptor for the complement anaphylatoxin C5a, has the potential to reduce the corticosteroid burden experienced by patients with AAV and may also improve outcomes for those with AAV kidney disease. It marks the culmination of almost 20 years of international collaboration, from understanding the pathological role of complement in basic murine models of AAV through to a phase III clinical trial, and emphasises the importance of following promising translational discoveries through to drug development and clinical deployment. This article summarises how recent progress in our understanding of the basic pathophysiology of AAV has resulted in the development of new and effective treatments and, reciprocally, how studying the impact of these treatments in patients has advanced our understanding of dysfunctional immunobiology in disease.
Collapse
Affiliation(s)
- Dominic P McGovern
- Vasculitis Research Group, Department of Medicine, Addenbrooke's Hospital Level 5, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK.
- Cambridge Lupus and Vasculitis Service, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - Rachel B Jones
- Vasculitis Research Group, Department of Medicine, Addenbrooke's Hospital Level 5, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
- Cambridge Lupus and Vasculitis Service, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - David R W Jayne
- Vasculitis Research Group, Department of Medicine, Addenbrooke's Hospital Level 5, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
- Cambridge Lupus and Vasculitis Service, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Rona M Smith
- Vasculitis Research Group, Department of Medicine, Addenbrooke's Hospital Level 5, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
- Cambridge Lupus and Vasculitis Service, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
36
|
Specks U, Marty PK. Interstitial lung disease, antineutrophil cystoplasmic antibodies and microscopic polyangiitis. Rheumatology (Oxford) 2025; 64:i42-i47. [PMID: 40071415 DOI: 10.1093/rheumatology/keae557] [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: 06/25/2024] [Accepted: 10/02/2024] [Indexed: 05/13/2025] Open
Abstract
This mini review explores the association of interstitial lung disease (ILD) with antineutrophil cystoplasmic antibodies (ANCA) and the clinical syndrome of microscopic polyangiitis (MPA). Reports on radiographic and histopathologic findings as well as genetic predispositions are reviewed. Based on this evidence a concept for the pathogenesis of the relationship of ILD, MPO-ANCA and MPA is proposed. Finally, a practical clinical management approach to patients presenting either with ILD and a positive ANCA test result, or to patients with MPA found to have pulmonary abnormalities qualifying as an ILD, is derived from the currently available literature. Treatment of these patients is based on up-to-date guidelines for the management of ILD as well as ANCA-associated vasculitis.
Collapse
Affiliation(s)
- Ulrich Specks
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | - Paige K Marty
- Division of Pulmonary Medicine, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
37
|
Bertuccio FR, Valente D, Baio N, Tomaselli S, Saracino L, Sciandrone G, Milanesi A, Delvino P, Codullo V, Corsico AG, Stella GM. A 28-Year-Old Man with Stridor and Dyspnea. J Clin Med 2025; 14:1532. [PMID: 40095461 PMCID: PMC11900471 DOI: 10.3390/jcm14051532] [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: 02/05/2025] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Background: Tracheobronchial stenosis is a significant complication in granulomatosis with polyangiitis (GPA), a systemic vasculitis that primarily affects the upper respiratory tract, kidneys, and lungs. The involvement of the tracheobronchial tree in GPA leads to airway narrowing, which can result in severe respiratory symptoms and increased morbidity, often requiring prompt diagnosis and management to prevent life-threatening airway obstruction. Method: We present the case of a 28-year-old male with mild exertional dyspnea, stridor, and retropharyngeal sputum. Clinical investigations revealed subglottic and bronchial concentric stenosis with granulomatous inflammation. A diagnosis of granulomatosis with polyangiitis (GPA) with isolated tracheobronchial stenosis (TBS) was confirmed. Results: Given the severity of airway obstruction, multidisciplinary management was initiated, combining rigid bronchoscopy with systemic immunosuppressive therapy. Post-intervention follow-up demonstrated significant airway improvement and maintained remission after two years. Conclusions: This case highlights TBS as a potentially debilitating GPA manifestation requiring a combination of systemic and endoscopic therapies. Further studies are needed to optimize therapeutic approaches and improve outcomes in GPA-associated TBS.
Collapse
Affiliation(s)
- Francesco Rocco Bertuccio
- Unit of Respiratory Disease, Cardiothoracic and Vascular Department, IRCCS Policlinico San Matteo, Viale Golgi 19, 27100 Pavia, Italy; (F.R.B.); (N.B.); (S.T.); (L.S.); (G.S.); (A.G.C.)
- Department of Internal Medicine and Pharmacology, University of Pavia, 27100 Pavia, Italy
| | - Davide Valente
- Radiology Institute, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Nicola Baio
- Unit of Respiratory Disease, Cardiothoracic and Vascular Department, IRCCS Policlinico San Matteo, Viale Golgi 19, 27100 Pavia, Italy; (F.R.B.); (N.B.); (S.T.); (L.S.); (G.S.); (A.G.C.)
- Department of Internal Medicine and Pharmacology, University of Pavia, 27100 Pavia, Italy
| | - Stefano Tomaselli
- Unit of Respiratory Disease, Cardiothoracic and Vascular Department, IRCCS Policlinico San Matteo, Viale Golgi 19, 27100 Pavia, Italy; (F.R.B.); (N.B.); (S.T.); (L.S.); (G.S.); (A.G.C.)
- Department of Internal Medicine and Pharmacology, University of Pavia, 27100 Pavia, Italy
| | - Laura Saracino
- Unit of Respiratory Disease, Cardiothoracic and Vascular Department, IRCCS Policlinico San Matteo, Viale Golgi 19, 27100 Pavia, Italy; (F.R.B.); (N.B.); (S.T.); (L.S.); (G.S.); (A.G.C.)
- Department of Internal Medicine and Pharmacology, University of Pavia, 27100 Pavia, Italy
| | - Gaetano Sciandrone
- Unit of Respiratory Disease, Cardiothoracic and Vascular Department, IRCCS Policlinico San Matteo, Viale Golgi 19, 27100 Pavia, Italy; (F.R.B.); (N.B.); (S.T.); (L.S.); (G.S.); (A.G.C.)
- Department of Internal Medicine and Pharmacology, University of Pavia, 27100 Pavia, Italy
| | - Alessandra Milanesi
- PhD Experimental Medicine, Università di Pavia, 27100 Pavia, Italy;
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy;
- Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Paolo Delvino
- School of Medicine, University of Milano-Bicocca, 20126 Milan, Italy;
- Rheumatology Unit, IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Veronica Codullo
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy;
- Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Angelo Guido Corsico
- Unit of Respiratory Disease, Cardiothoracic and Vascular Department, IRCCS Policlinico San Matteo, Viale Golgi 19, 27100 Pavia, Italy; (F.R.B.); (N.B.); (S.T.); (L.S.); (G.S.); (A.G.C.)
- Department of Internal Medicine and Pharmacology, University of Pavia, 27100 Pavia, Italy
| | - Giulia Maria Stella
- Unit of Respiratory Disease, Cardiothoracic and Vascular Department, IRCCS Policlinico San Matteo, Viale Golgi 19, 27100 Pavia, Italy; (F.R.B.); (N.B.); (S.T.); (L.S.); (G.S.); (A.G.C.)
- Department of Internal Medicine and Pharmacology, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
38
|
Duan N, Li Z, Li Z, Pang L, Du J, Chang L, Huang H, Li H. Evaluation of a tumor marker gastrin-releasing peptide precursor in the patients with kidney injuries. Am J Cancer Res 2025; 15:824-832. [PMID: 40084352 PMCID: PMC11897619 DOI: 10.62347/cbsp3728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025] Open
Abstract
Gastrin-releasing peptide precursor (ProGRP) is a bioactive precursor of GRP and might play an important role as an emerging tumor marker in early cancer diagnosis. It might also be abnormal in the nonmalignant disease and renal function abnormalities. The present study was undertaken to investigate the changes of ProGRP levels in patients with kidney injuries, especially with chronic kidney disease (CKD), determine the upper reference intervals and clinical diagnostic value of ProGRP in CKD, and thus help oncologists in interpreting ProGRP levels and making clinical judgments of malignances. 676 individuals were enrolled in this cross-sectional study and divided into five groups: healthy control (n=194), CKD (n=272), nephrotic syndrome (NS) (n=137), antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) (n=41), and urinary tract infection (UTI) (n=32). A total of 27 features including age, gender, and 25 laboratory markers were analyzed. Machine learning algorithms were built for the diagnostic models of CKD. Statistical analysis was performed by R software. It was shown that serum ProGRP level in CKD was significantly higher than that in healthy controls, UTI and NS (P < 0.01). The upper reference limit of ProGRP was 188.42 pg/ml for CKD, 245.40 pg/ml for CKD IV-V, and 97.25 pg/ml for NS. Compared with the healthy control, the level of serum ProGRP in CKD stages II, III, IV-V was significantly increased and elevated progressively with CKD grade (P < 0.01). Random Forest (RF) model works best among 4 building machine learning algorithms. 5 vital indicators, ProGRP, estimated glomerular filtration rate (eGFR), urea, albumin (ALB), and direct bilirubin (DBIL), were selected to establish RF model for diagnosing CKD with an area under the curve (AUC) of 0.96 (95% confidence interval [CI]: 0.94-0.97) and high sensitivity (0.89) and specificity (0.92). This study demonstrates that the level of ProGRP in patients with CKD, nephrotic syndrome or AAV, was significantly higher than that in the healthy population. The machine learning model of ProGRP with DBIL, eGFR, ALB, and urea, could provide good clinical value for CKD evaluation.
Collapse
Affiliation(s)
| | | | - Zhiyan Li
- Department of Clinical Laboratory, Peking University First HospitalBeijing 100034, China
| | - Lu Pang
- Department of Clinical Laboratory, Peking University First HospitalBeijing 100034, China
| | - Jialin Du
- Department of Clinical Laboratory, Peking University First HospitalBeijing 100034, China
| | - Le Chang
- Department of Clinical Laboratory, Peking University First HospitalBeijing 100034, China
| | - Haiming Huang
- Department of Clinical Laboratory, Peking University First HospitalBeijing 100034, China
| | - Haixia Li
- Department of Clinical Laboratory, Peking University First HospitalBeijing 100034, China
| |
Collapse
|
39
|
Zheng Z, Wang Y, Xie J, Chen Z, Jiang B, Xu Y. The association between serum lipids at diagnosis and renal outcome in microscopic polyangiitis patients. PeerJ 2025; 13:e18839. [PMID: 39950045 PMCID: PMC11823655 DOI: 10.7717/peerj.18839] [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: 08/28/2024] [Accepted: 12/18/2024] [Indexed: 02/16/2025] Open
Abstract
Objectives Microscopic polyangiitis (MPA) is a subgroup of ANCA-associated vasculitis (AAV), which is characterized by vascular endothelial cell damage caused by abnormally activated neutrophils. Dyslipidemia is associated with vascular endothelial cell injury, and the relationship between blood lipid levels and renal prognosis in MPA patients is not clear. We aim to investigate the correlation between blood lipid levels at diagnosis and renal prognosis in MPA patients. Methods Firstly, we retrospectively included 110 patients diagnosed with MPA and the primary endpoint was the occurrence of end stage renal disease (ESRD). The association between blood lipids at diagnosis and renal outcome was evaluated with Cox regression analysis and survival analysis. Secondly, we explored the potential underlying mechanism of poor renal prognosis in patients with high triglycerides (TG) levels at diagnosis using data independent acquisition (DIA) quantitative proteomics. Results During a median follow-up period of 23 months, 44 out of 110 patients (40%) developed ESRD. High serum TG at diagnosis was associated with ESRD development after adjusting for several confounding factors including age, gender, body mass index (BMI), hypertension, diabetes mellitus, estimated glomerular filtration rate (eGFR) and Birmingham Vasculitis Activity Score (BVAS). Serum very low-density lipoprotein (VLDL) demonstrated a marginal trend towards association with ESRD development. MPA patients with TG >1.45 mmol/L or VLDL > 0.66 mmol/L had significantly higher risk of ESRD development than those with TG ≤ 1.45 mmol/L or VLDL ≤ 0.66 mmol/L. DIA quantitative proteomics analysis suggested that patients with elevated TG levels and severe MPA had an upregulation of profibrotic pathways, inflammatory signaling, and complement and coagulation cascades, in contrast to those with lower TG levels and milder disease severity. Conclusions In MPA patients, high TG or VLDL at diagnosis is associated with an increased risk of ESRD development. The potential mechanisms may be associated with the upregulation of profibrotic and inflammatory signaling pathways, and the activation of complement and coagulation cascades.
Collapse
Affiliation(s)
- Zigui Zheng
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yujia Wang
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Jingzhi Xie
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhimin Chen
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Bingjing Jiang
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yanfang Xu
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
40
|
Chung J, Ha JW, Park YB, Lee SW. Serum glutathione peroxidase-3 concentration at diagnosis as a biomarker for assessing disease activity and damage of antineutrophil cytoplasmic antibody-associated vasculitis at diagnosis. Front Mol Biosci 2025; 12:1549454. [PMID: 39990869 PMCID: PMC11842223 DOI: 10.3389/fmolb.2025.1549454] [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: 12/24/2024] [Accepted: 01/27/2025] [Indexed: 02/25/2025] Open
Abstract
Background In this study, we investigated whether serum glutathione peroxidase-1 (GPX-3) concentration at diagnosis could be used to assess vasculitis activity and damage at diagnosis in immunosuppressive drug-naïve patients with antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). Methods We included 71 immunosuppressive drug-naïve patients newly diagnosed with AAV. Medical records were retrospectively reviewed and serum GPX-3 concentration was measured using serum samples collected and stored at diagnosis. The degree of vascular activity and extent of damage were assessed using the Birmingham vasculitis activity score (BVAS) and vasculitis damage index (VDI), respectively. Poor outcomes including all-cause mortality, end-stage kidney disease, and cerebrovascular and cardiovascular diseases were also investigated. Results The median age of the study subjects was 63.0 years, 26 and 45 patients were males and females, respectively. The median GPX-3 concentration was measured as 82.8 ng/mL. Serum GPX-3 concentration at diagnosis was inversely correlated with BVAS (r = -0.280), VDI (r = -0.263), and C-reactive protein (r = -0.261) at diagnosis, whereas, it was positively correlated with haemoglobin (r = 0.255), and serum albumin (r = 0.240) at diagnosis, respectively. However, serum GPX-3 concentration at diagnosis was not significantly associated with poor outcomes during follow-up in patients with AAV. Conclusion In this study, we demonstrated for the first time that serum GPX-3 concentration at diagnosis correlates with vasculitis activity and damage at diagnosis in patients with AAV, suggesting a possible role of serum GPX-3 as a complementary biomarker for assessing AAV activity in real clinical practice.
Collapse
Affiliation(s)
- Jihye Chung
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jang Woo Ha
- Division of Rheumatology, Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Gyeonggi-do, Republic of Korea
| | - Yong-Beom Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang-Won Lee
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
41
|
Qi M, Tang Y, Zhou H, Wang M, Yi Q, Liang Z, He JQ. Comprehensive characteristics of pulmonary antineutrophil cytoplasmic antibody-associated vasculitis and the development of a predictive nomogram for mortality. Int Immunopharmacol 2025; 147:113986. [PMID: 39755109 DOI: 10.1016/j.intimp.2024.113986] [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: 07/25/2024] [Revised: 12/10/2024] [Accepted: 12/28/2024] [Indexed: 01/06/2025]
Abstract
OBJECTIVES Antineutrophil cytoplasmic antibody (ANCA) associated vasculitis (AAV) is a rare and potential devastating disease with high mortality, frequently with pulmonary involvement. Our study aimed to explore the pulmonary features of AAV and identify predictors of long-term survival. METHODS We retrospectively analyzed 538 AAV patients diagnosed between January 2013 and July 2019, with follow-up data extending to August 2020. The Least absolute shrinkage and selection operator (LASSO) regression analysis was employed to identify variables predictive of mortality. Subsequently, a nomogram was developed, with its predictive accuracy and discrimination assessed by the concordance index and calibration curves, respectively. RESULTS A total of 460 (85.5 %) AAV patients presented with pulmonary involvement. The mortality was 36.8 %. Patients with pulmonary involvement more frequently exhibited respiratory symptoms, predominantly interstitial lung disease on radiographs, and were at higher risk for respiratory failure, diffuse alveolar hemorrhage, mechanic ventilation and death (All P < 0.05). The LASSO regression pinpointed 16 predictors of mortality, and the predictive model demonstrated an area under the curve of 0.810. The nomogram, based on these variables, achieved a concordance index of 0.825, with calibration curve indicating excellent predictive agreement. CONCLUSION The study establishes a predictive model for AAV mortality with high accuracy, offering insights crucial for patient care. Pulmonary involvement, prevalent and linked to higher mortality, underscores the need for precise predictive tools in AAV management.
Collapse
Affiliation(s)
- Min Qi
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China; State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yongjiang Tang
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China; State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Haixia Zhou
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China; State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Maoyun Wang
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China; State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Qun Yi
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China; Sichuan Cancer Hospital, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Zongan Liang
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China; State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Jian-Qing He
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China; State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China.
| |
Collapse
|
42
|
Tan MH, Jayne D. Top ten tips in managing ANCA vasculitis. Clin Kidney J 2025; 18:sfae389. [PMID: 39927255 PMCID: PMC11803310 DOI: 10.1093/ckj/sfae389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Indexed: 02/11/2025] Open
Abstract
Diagnosing and managing antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) remain a challenge for many clinicians, due to the complexity of the disease manifestations and its treatment. There has been a paradigm shift in ANCA vasculitis management, where treatment incorporates both emergency life- and organ-saving procedures and longer-term care to manage relapse and co-morbidity risk and the complications of organ damage. Here, we highlight 10 key tips for the management of ANCA-associated vasculitis based on current evidence and clinical experience. First, we advise making the diagnosis as early as possible, emphasizing the importance of using high-quality ANCA assays. Second, we recommend the use of glucocorticoids in combination with rituximab and/or cyclophosphamide as induction therapy. Third, plasma exchange should be considered in patients with severe renal impairment and diffuse alveolar haemorrhage. We advise the use of rapidly reducing glucocorticoid regimens and advocate consideration of avacopan early in the disease course. We recommend the use of rituximab as maintenance therapy and routine monitoring of kidney function, proteinuria, ANCA and immunoglobulin levels at baseline and during follow-up. The use of prophylactic antibiotics in susceptible patients and timely vaccination schedules is discussed. Rituximab is the preferred immune suppressive for treatment of relapse. Finally, we recommend switching treatment modalities in patients whose vasculitis is refractory to induction therapy and to consider plasma exchange in selected patients. These key tips aim to provide the necessary guidance to improve patient outcomes and reduce adverse events.
Collapse
Affiliation(s)
- Min Hui Tan
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Department of Nephrology, Hospital Kuala Lumpur, Kuala Lumpur, Malaysia
| | - David Jayne
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
43
|
Sondhi M, Qaiser I, Hayat S, Umer S, Muzaffar K. Unveiling the Shadow: Unraveling the Cause of Blindness. Arthritis Care Res (Hoboken) 2025; 77:157-162. [PMID: 39297277 DOI: 10.1002/acr.25435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 09/03/2024] [Accepted: 09/13/2024] [Indexed: 10/17/2024]
|
44
|
Casal Moura M, Fervenza FC, Specks U, Sethi S. "Normal" Glomerular Score Correlates With Outcomes in Antineutrophil Cytoplasmic Autoantibody-Associated Vasculitis With Glomerulonephritis. Kidney Int Rep 2025; 10:596-600. [PMID: 39990903 PMCID: PMC11843102 DOI: 10.1016/j.ekir.2024.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 02/25/2025] Open
Affiliation(s)
- Marta Casal Moura
- Division of Pulmonary and Critical Care, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Fernando C. Fervenza
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Ulrich Specks
- Division of Pulmonary and Critical Care, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Sanjeev Sethi
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
45
|
Sapkota N, Aryal Y, Basnet P. Avacopan as a Steroid-Sparing Therapy in Relapsing Granulomatosis With Polyangiitis. Cureus 2025; 17:e79072. [PMID: 40109798 PMCID: PMC11920855 DOI: 10.7759/cureus.79072] [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] [Accepted: 02/15/2025] [Indexed: 03/22/2025] Open
Abstract
Granulomatosis with polyangiitis (GPA) is a rare autoimmune disease that causes inflammation in small and medium-sized blood vessels, affecting organs such as the lungs, kidneys, and sinuses. We report the case of a 66-year-old man with relapsing GPA. He first presented with sinus and ear symptoms, which were managed with steroids and other immunosuppressive drugs. After many years of remission, he relapsed with nasal congestion, nosebleeds, and lung nodules. During his latest flare, he was treated with high-dose prednisone, rituximab, and avacopan, a new oral drug that blocks the C5a receptor. Avacopan helped reduce inflammation and allowed for a significant decrease in steroid use. The patient's rapid improvement supports the role of avacopan as a steroid-sparing agent in GPA management, offering a promising way to reduce the harmful side effects of long-term steroid therapy.
Collapse
Affiliation(s)
- Nisha Sapkota
- Medicine, One Brooklyn Health-Interfaith Medical Center, New York, USA
| | - Yubraj Aryal
- Hospital Medicine, Geisinger Medical Center, Danville, USA
| | | |
Collapse
|
46
|
Luo W, Liu C, Zhang L, Tang J, Chen J, Zhao Y, Huang X, Zheng X, Chen L, Xie C, Wei X, Luo X, Xiong A. Characteristics and risk factors for infection in patients with ANCA-associated vasculitis: A systematic review and meta-analysis. Autoimmun Rev 2025; 24:103713. [PMID: 39617249 DOI: 10.1016/j.autrev.2024.103713] [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: 05/31/2024] [Revised: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE To summarize the characteristics and risk factors for infection in patients with anti-neutrophil cytoplasmic antibody-associated vasculitis (AAV). METHODS PubMed, Embase, and Cochrane Library databases were searched for relevant articles from database inception to November 2023. The prevalence, odds ratio (OR), and mean difference (MD) with 95 % confidence intervals (CIs) were pooled using a random-effects model. Sensitivity and subgroup analysis were also performed. RESULTS Forty-one studies with 5343 patients with AAV were included, of whom 2890 patients experienced an infection. The pooled prevalence was 54.6 % (95 % CI, 48.4 % to 61.1 %) for all infections and 35.8 % (95 % CI, 31.0 % to 40.8 %) for severe infections; and prevalence of Pneumocystis jirovecii pneumonia, aspergillosis, candidiasis, cryptococcosis, herpes zoster, cytomegalovirus, and specific bacterial infections were pooled. The respiratory system was the most common infection site, followed by blood, urinary tract, skin and soft tissue, and digestive infections. Risk factors for infection included older age, end-stage renal disease, dialysis, diabetes, smoking, kidney and lung involvement, leukopenia; higher Birmingham Vasculitis Activity Score, and serum creatinine and C-reactive protein levels; and lower hemoglobin levels, and platelet and CD4 counts. In addition, use of cyclophosphamide, steroid pulse therapy, plasma exchange, and higher initial glucocorticoid dose were associated with significantly increased risk of infection. CONCLUSION In patients with AAV, therapy should take risk factors for infection into account. Risk factors should be modified wherever possible. Physicians should be familiar with the common infection sites and pathogens, and consider empiric therapy covering common pathogens for life-threatening infections.
Collapse
Affiliation(s)
- Wenxuan Luo
- Department of Rheumatology and Immunology, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, The Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Can Liu
- Department of Rheumatology and Immunology, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, The Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Lei Zhang
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jie Tang
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jie Chen
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yanzao Zhao
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xuemei Huang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaoli Zheng
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Long Chen
- Department of Rheumatology and Immunology, Suining Central Hospital, Suining, Sichuan, China
| | - Chuanmei Xie
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xin Wei
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Xiongyan Luo
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Anji Xiong
- Department of Rheumatology and Immunology, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, The Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, Sichuan, China; Inflammation and Immunology Key Laboratory of Nanchong City, Nanchong, Sichuan, China; Nanchong Central Hospital, (Nanchong Clinical Research Center), Nanchong, Sichuan, China.
| |
Collapse
|
47
|
Escoda T, Dehaene A, Velardocchio L, Deveze A, Terrier B, Chiche L. Rare association of granulomatosis with polyangiitis with an underdiagnosed spondyloarthritis effectively treated with rituximab: A case-report. Medicine (Baltimore) 2025; 104:e41366. [PMID: 39889193 PMCID: PMC11789892 DOI: 10.1097/md.0000000000041366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/06/2025] [Accepted: 01/09/2025] [Indexed: 02/02/2025] Open
Abstract
RATIONALE Associations of autoimmune diseases are rare but interesting and challenging situations from a diagnostic, pathophysiological, and therapeutic point of view. This article studies a rare association of autoimmune diseases by discussing the pathophysiological hypotheses and an original therapeutic management. The coexistence of antineutrophil cytoplasmic antibody-associated vasculitis and spondyloarthritis has rarely been described. PATIENT CONCERNS We present a patient with inflammatory back pain, stiffness, and enthesopathies followed by pulmonary and ear, nose and throat granulomatous involvement. DIAGNOSES A combination of spondyloarthritis and granulomatosis with polyangiitis, with spinal, enthesopathic, pulmonary, and ear, nose and throat involvement. INTERVENTIONS AND OUTCOMES Effective treatment with rituximab both on spondyloarthritis and vasculitis. LESSONS We discuss the pathogenic, diagnostic, and therapeutic implications of this rare but intriguing association between these 2 inflammatory conditions.
Collapse
Affiliation(s)
- Thomas Escoda
- Service de Médecine Interne, Hôpital Européen, Marseille, France
| | | | | | - Arnaud Deveze
- Service d’Oto-Rhino-Laryngologie, Chirurgie de la Face et du Cou, Clinique Clairval, Marseille, France
| | | | - Laurent Chiche
- Service de Médecine Interne, Hôpital Européen, Marseille, France
| |
Collapse
|
48
|
Kurts C, von Vietinghoff S, Krebs CF, Panzer U. Kidney immunology from pathophysiology to clinical translation. Nat Rev Immunol 2025:10.1038/s41577-025-01131-y. [PMID: 39885266 DOI: 10.1038/s41577-025-01131-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2025] [Indexed: 02/01/2025]
Abstract
Kidney diseases are widespread and represent a considerable medical, social and economic burden. However, there has been marked progress in understanding the immunological aspects of kidney disease. This includes the identification of distinct intrarenal immunological niches and characterization of kidney disease endotypes according to the underlying molecular immunopathology, as well as a better understanding of the pathological roles for T cells, mononuclear phagocytes and B cells and the renal elements they target. These insights have improved the diagnosis of kidney disease. Here, we discuss new developments in our understanding of kidney immunology, focusing on immune mechanisms of disease and their translational implications for the diagnosis and treatment of kidney disease. We also describe the immune-mediated crosstalk between the kidney and other organs that influences kidney disease and extrarenal inflammation.
Collapse
Affiliation(s)
- Christian Kurts
- Institute of Molecular Medicine and Experimental Immunology, University Hospital, Bonn, Germany.
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia.
| | - Sibylle von Vietinghoff
- Nephrology Section, University Hospital Bonn, Medical Clinic and Polyclinic I, Bonn, Germany
| | - Christian F Krebs
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulf Panzer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
49
|
Toki T, Mizunoya K, Itabashi M, Nishikawa N, Hoshino K, Saito H, Morimoto Y. Acute decompensated right heart failure potentially triggered by multiple factors including pulmonary vasodilator removal during plasma exchange: a case report. JA Clin Rep 2025; 11:5. [PMID: 39869255 PMCID: PMC11772900 DOI: 10.1186/s40981-025-00765-0] [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: 11/22/2024] [Revised: 12/28/2024] [Accepted: 01/07/2025] [Indexed: 01/28/2025] Open
Abstract
BACKGROUND Plasma exchange (PE) removes high-molecular-weight substances and is sometimes used for antineutrophil cytoplasmic antibody-associated vasculitis (AAV) with alveolar hemorrhage. Hypotension during PE is rare, except in allergic cases. We report a case of shock likely caused by increased pulmonary vascular resistance (PVR) during PE. CASE PRESENTATION A 66-year-old man with pulmonary hypertension (PH) and glomerulonephritis was admitted with dyspnea. He had discontinued sildenafil prior to admission. Alveolar hemorrhage associated with AAV was suspected, and PE was performed. Soon after, he developed circulatory failure and hyperlactatemia. Echocardiography revealed right ventricular dilation, suggesting increased PVR. Inhaled nitric oxide (iNO) was administered, rapidly improving hyperlactatemia and oxygenation. The shock observed during PE was attributed to multiple factors, including the potential removal of sildenafil, which may have led to an increase in PVR. CONCLUSIONS The shock was attributable to acute right heart failure caused by an exacerbation of PH, possibly due to sildenafil removal via PE, although other contributing factors could not be excluded.
Collapse
Affiliation(s)
- Takayuki Toki
- Department of Anesthesiology and Critical Care Medicine, Hokkaido University Hospital, N14W5, Kita-ku, Sapporo, 060-8648, Japan.
| | - Kazuyuki Mizunoya
- Department of Anesthesiology and Critical Care Medicine, Hokkaido University Hospital, N14W5, Kita-ku, Sapporo, 060-8648, Japan
| | - Misa Itabashi
- Department of Anesthesiology and Critical Care Medicine, Hokkaido University Hospital, N14W5, Kita-ku, Sapporo, 060-8648, Japan
| | - Naoki Nishikawa
- Department of Anesthesiology and Critical Care Medicine, Hokkaido University Hospital, N14W5, Kita-ku, Sapporo, 060-8648, Japan
| | - Koji Hoshino
- Department of Anesthesiology and Critical Care Medicine, Hokkaido University Hospital, N14W5, Kita-ku, Sapporo, 060-8648, Japan
| | - Hitoshi Saito
- Department of Anesthesiology and Critical Care Medicine, Hokkaido University Hospital, N14W5, Kita-ku, Sapporo, 060-8648, Japan
| | - Yuji Morimoto
- Department of Anesthesiology and Critical Care Medicine, Hokkaido University Hospital, N14W5, Kita-ku, Sapporo, 060-8648, Japan
| |
Collapse
|
50
|
Bonasia CG, Inrueangsri N, Bijma T, Borggrewe M, Post AI, Mennega KP, Abdulahad WH, Rutgers A, Bos NA, Heeringa P. Circulating B cells display differential immune regulatory molecule expression in granulomatosis with polyangiitis. Clin Exp Immunol 2025; 219:uxae096. [PMID: 39435875 PMCID: PMC11773817 DOI: 10.1093/cei/uxae096] [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: 07/03/2024] [Revised: 09/20/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024] Open
Abstract
Granulomatosis with polyangiitis (GPA) is a B-cell-mediated, relapsing, autoimmune disease. There is a need for novel therapeutic approaches and relapse markers to achieve durable remission. B cells express immune regulatory molecules that modulate their activation and maintain tolerance. While recent studies show dysregulation of these molecules in other autoimmune diseases, data on their expression in GPA are limited. This study aimed to map the expression of surface immune regulatory molecules on circulating B-cell subsets in GPA and correlate their expression with clinical parameters. Immune regulatory molecule expression on circulating B-cell subsets was comprehensively examined in active GPA (n = 16), GPA in remission (n = 16), and healthy controls (n = 16) cross-sectionally using a 35-color B-cell-specific spectral flow cytometry panel. Our supervised and unsupervised in-depth analysis revealed differential expression of inhibitory and stimulatory immune molecules on distinct B-cell populations in GPA, with the most notable differences observed in active GPA. These differences include the upregulation of FcγRIIB on nonmature B cells, downregulation of CD21 and upregulation of CD86 on antigen-experienced B cells, and elevated CD22 expression on various populations. Additionally, we found a strong association between FcγRIIB, BTLA, and CD21 expression on specific B-cell populations and disease activity in GPA. Together, these findings provide novel insights into the immune regulatory molecule expression profile of B cells in GPA and could potentially form the foundation for new therapeutic approaches and disease monitoring markers.
Collapse
Affiliation(s)
- Carlo G Bonasia
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Nanthicha Inrueangsri
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Theo Bijma
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Aline I Post
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Kevin P Mennega
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wayel H Abdulahad
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Abraham Rutgers
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Nicolaas A Bos
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|