1
|
Pugh D, Patel D, Macnaught G, Czopek A, Bruce L, Donachie J, Gallacher PJ, Tan S, Ahlman M, Grayson PC, Basu N, Dhaun N. 18F-FDG-PET/MR imaging to monitor disease activity in large vessel vasculitis. Nat Commun 2024; 15:7314. [PMID: 39183340 PMCID: PMC11345444 DOI: 10.1038/s41467-024-51613-1] [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: 11/23/2023] [Accepted: 08/14/2024] [Indexed: 08/27/2024] Open
Abstract
Disease-monitoring in large vessel vasculitis (LVV) is challenging. Simultaneous 18F-fluorodeoxyglucose positron emission tomography with magnetic resonance imaging (PET/MRI) provides functional assessment of vascular inflammation alongside high-definition structural imaging with a relatively low burden of radiation exposure. Here, we investigate the ability of PET/MRI to monitor LVV disease activity longitudinally in a prospective cohort of patients with active LVV. We demonstrate that both the PET and MRI components of the scan can distinguish active from inactive disease using established quantification methods. Using logistic-regression modelling of PET/MRI metrics, we devise a novel PET/MRI-specific Vasculitis Activity using MR PET (VAMP) score which is able to distinguish active from inactive disease with more accuracy than established methods and detects changes in disease activity longitudinally. These findings are evaluated in an independent validation cohort. Finally, PET/MRI improves clinicians' assessment of LVV disease activity and confidence in disease management, as assessed via clinician survey. In summary, PET/MRI may be useful in tracking disease activity and assessing treatment-response in LVV. Based on our findings, larger, prospective studies assessing PET/MRI in LVV are now warranted.
Collapse
Affiliation(s)
- Dan Pugh
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Dilip Patel
- Department of Radiology, Royal Infirmary of Edinburgh, Edinburgh, UK
| | | | - Alicja Czopek
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Lorraine Bruce
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - James Donachie
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Peter J Gallacher
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Sovira Tan
- National Institute of Arthritis & Musculoskeletal & Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mark Ahlman
- Department of Radiology & Imaging, Medical College of Georgia, Georgia, USA
| | - Peter C Grayson
- National Institute of Arthritis & Musculoskeletal & Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Neil Basu
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
| | - Neeraj Dhaun
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
2
|
The Soluble Fms-like Tyrosine Kinase-1 Contributes to Structural and Functional Changes in Endothelial Cells in Chronic Kidney Disease. Int J Mol Sci 2022; 23:ijms232416059. [PMID: 36555698 PMCID: PMC9787493 DOI: 10.3390/ijms232416059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Endothelial cells are a critical target of the soluble Fms-like tyrosine kinase-1 (sFlt-1), a soluble factor increased in different diseases with varying degrees of renal impairment and endothelial dysfunction, including chronic kidney disease (CKD). Although the mechanisms underlying endothelial dysfunction are multifactorial and complex, herein, we investigated the damaging effects of sFlt-1 on structural and functional changes in endothelial cells. Our results evidenced that sera from patients with CKD stiffen the endothelial cell cortex in vitro, an effect correlated with sFlt-1 levels and prevented by sFlt-1 neutralization. Besides, we could show that recombinant sFlt-1 leads to endothelial stiffening in vitro and in vivo. This was accompanied by cytoskeleton reorganization and changes in the endothelial barrier function, as observed by increased actin polymerization and endothelial cell permeability, respectively. These results depended on the activation of the p38 MAPK and were blocked by the specific inhibitor SB203580. However, sFlt-1 only minimally affected the expression of stiffness-sensitive genes. These findings bring new insight into the mechanism of action of sFlt-1 and its biological effects that cannot be exclusively ascribed to the regulation of angiogenesis.
Collapse
|
3
|
Scurt FG, Bose K, Hammoud B, Brandt S, Bernhardt A, Gross C, Mertens PR, Chatzikyrkou C. Old known and possible new biomarkers of ANCA-associated vasculitis. J Autoimmun 2022; 133:102953. [PMID: 36410262 DOI: 10.1016/j.jaut.2022.102953] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/06/2022] [Accepted: 11/06/2022] [Indexed: 11/19/2022]
Abstract
Antineutrophil cytoplasm antibody (ANCA)-associated vasculitis (AAV) comprises a group of multisystem disorders involving severe, systemic, small-vessel vasculitis with short- and long term serious and life-threating complications. Despite the simplification of treatment, fundamental aspects concerning assessment of its efficacy and its adaptation to encountered complications or to the relapsing/remitting/subclinical disease course remain still unknown. The pathogenesis of AAV is complex and unique, and despite the progress achieved in the last years, much has not to be learnt. Foremost, there is still no accurate marker enabling us to monitoring disease and guide therapy. Therefore, the disease management relays often on clinical judgment and follows a" trial and error approach". In the recent years, an increasing number of new molecules s have been explored and used for this purpose including genomics, B- and T-cell subpopulations, complement system factors, cytokines, metabolomics, biospectroscopy and components of our microbiome. The aim of this review is to discuss both the role of known historical and clinically established biomarkers of AAV, as well as to highlight potential new ones, which could be used for timely diagnosis and monitoring of this devastating disease, with the goal to improve the effectiveness and ameliorate the complications of its demanding therapy.
Collapse
Affiliation(s)
- Florian G Scurt
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany.
| | - K Bose
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany
| | - Ben Hammoud
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany
| | - S Brandt
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany
| | - A Bernhardt
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany
| | - C Gross
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany
| | - Peter R Mertens
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany
| | | |
Collapse
|
4
|
Néel A, Degauque N, Bruneau S, Braudeau C, Bucchia M, Caristan A, De Mornac D, Genin V, Glemain A, Oriot C, Rimbert M, Brouard S, Josien R, Hamidou M. [Pathogenesis of ANCA-associated vasculitides in 2021: An update]. Rev Med Interne 2022; 43:89-97. [PMID: 35033384 DOI: 10.1016/j.revmed.2021.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 10/22/2021] [Accepted: 11/04/2021] [Indexed: 10/19/2022]
Abstract
Anticytoplasmic neutrophil antibodies (ANCA)-associated vasculitis (AAV) are rare systemic immune-mediated diseases characterized by small vessel necrotizing vasculitis and/or respiratory tract inflammation. Over the last 2 decades, anti-MPO vasculitis mouse model has enlightened the role of ANCA, neutrophils, complement activation, T helper cells (Th1, Th17) and microbial agents. In humans, CD4T cells have been extensively studied, while the dramatic efficacy of rituximab demonstrated the key role of B cells. Many areas of uncertainty remain, such as the driving force of GPA extra-vascular granulomatous inflammation and the relapse risk of anti-PR3 AAV pathogenesis. Animal models eventually led to identify complement activation as a promising therapeutic target. New investigation tools, which permit in depth immune profiling of human blood and tissues, may open a new era for the studying of AAV pathogenesis.
Collapse
Affiliation(s)
- A Néel
- Service de médecine interne, CHU de Nantes, Nantes, France; Inserm, centre de recherche en transplantation et immunologie, UMR 1064, université de Nantes, Nantes, France; Centre de référence maladies auto-immunes systémiques Rares, hôpital Cochin, AP-HP, Paris, France.
| | - N Degauque
- Inserm, centre de recherche en transplantation et immunologie, UMR 1064, université de Nantes, Nantes, France
| | - S Bruneau
- Inserm, centre de recherche en transplantation et immunologie, UMR 1064, université de Nantes, Nantes, France
| | - C Braudeau
- Inserm, centre de recherche en transplantation et immunologie, UMR 1064, université de Nantes, Nantes, France; Laboratoire d'immunologie, CHU de Nantes, Nantes, France
| | - M Bucchia
- Inserm, centre de recherche en transplantation et immunologie, UMR 1064, université de Nantes, Nantes, France; Service de pédiatrie, CHU de Nantes, Nantes, France
| | - A Caristan
- Service de médecine interne, CHD Vendée, La-Roche-Sur-Yon, France
| | - D De Mornac
- Service de médecine interne, CHU de Nantes, Nantes, France; Inserm, centre de recherche en transplantation et immunologie, UMR 1064, université de Nantes, Nantes, France
| | - V Genin
- Service de médecine interne, CHU de Nantes, Nantes, France; Inserm, centre de recherche en transplantation et immunologie, UMR 1064, université de Nantes, Nantes, France
| | - A Glemain
- Inserm, centre de recherche en transplantation et immunologie, UMR 1064, université de Nantes, Nantes, France
| | - C Oriot
- Inserm, centre de recherche en transplantation et immunologie, UMR 1064, université de Nantes, Nantes, France; Service de pédiatrie, CHU de Nantes, Nantes, France
| | - M Rimbert
- Inserm, centre de recherche en transplantation et immunologie, UMR 1064, université de Nantes, Nantes, France; Laboratoire d'immunologie, CHU de Nantes, Nantes, France
| | - S Brouard
- Inserm, centre de recherche en transplantation et immunologie, UMR 1064, université de Nantes, Nantes, France
| | - R Josien
- Inserm, centre de recherche en transplantation et immunologie, UMR 1064, université de Nantes, Nantes, France; Laboratoire d'immunologie, CHU de Nantes, Nantes, France
| | - M Hamidou
- Service de médecine interne, CHU de Nantes, Nantes, France; Inserm, centre de recherche en transplantation et immunologie, UMR 1064, université de Nantes, Nantes, France
| |
Collapse
|
5
|
Wewers TM, Schulz A, Nolte I, Pavenstädt H, Brand M, Di Marco GS. Circulating Soluble Fms-like Tyrosine Kinase in Renal Diseases Other than Preeclampsia. J Am Soc Nephrol 2021; 32:1853-1863. [PMID: 34155060 PMCID: PMC8455271 DOI: 10.1681/asn.2020111579] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/20/2021] [Indexed: 02/04/2023] Open
Abstract
Soluble Fms-like tyrosine kinase (sFlt-1/sVEGFR1) is a naturally occurring antagonist of vascular endothelial growth factor (VEGF). Despite being a secreted, soluble protein lacking cytoplasmic and transmembrane domains, sFlt-1 can act locally and be protective against excessive microenvironmental VEGF concentration or exert autocrine functions independently of VEGF. Circulating sFlt-1 may indiscriminately affect endothelial function and the microvasculature of distant target organs. The clinical significance of excess sFlt-1 in kidney disease was first shown in preeclampsia, a major renal complication of pregnancy. However, circulating sFlt-1 levels appear to be increased in various diseases with varying degrees of renal impairment. Relevant clinical associations between circulating sFlt-1 and severe outcomes (e.g., endothelial dysfunction, renal impairment, cardiovascular disease, and all-cause mortality) have been observed in patients with CKD and after kidney transplantation. However, sFlt-1 appears to be protective against renal dysfunction-associated aggravation of atherosclerosis and diabetic nephropathy. Therefore, in this study, we provide an update on sFlt-1 in several kidney diseases other than preeclampsia, discuss clinical findings and experimental studies, and briefly consider its use in clinical practice.
Collapse
Affiliation(s)
- Theresa M. Wewers
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany,Small Animal Hospital, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Annika Schulz
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany
| | - Ingo Nolte
- Small Animal Hospital, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Hermann Pavenstädt
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany
| | - Marcus Brand
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany
| | - Giovana S. Di Marco
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany,Correspondence: Giovana S. Di Marco, Albert-Schweitzer-Campus 1, Building A14, 48149 Münster, Germany.
| |
Collapse
|
6
|
Vegting Y, Vogt L, Anders HJ, de Winther MPJ, Bemelman FJ, Hilhorst ML. Monocytes and macrophages in ANCA-associated vasculitis. Autoimmun Rev 2021; 20:102911. [PMID: 34298153 DOI: 10.1016/j.autrev.2021.102911] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 06/05/2021] [Indexed: 12/15/2022]
Abstract
Anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitides (AAV) are characterized by inflammation of small-to-medium-sized blood vessels and the presence of autoantibodies against cytoplasmic proteases sited in neutrophils and monocytes. Increasing evidence indicates a substantial role of monocytes and macrophages in the pathogenesis of AAV. Activated monocytes and macrophages contribute to necroinflammation in peripheral vasculitic lesions as well as to central and peripheral mechanisms of autoimmunity. The intermediate monocyte subset (CD14++CD16+) is increased and monocytes show elevated expression of CD14, Toll-like receptor 2/4, MHCII and integrins, likely reflecting activation and increased monocyte extravasation. Monocytes differentiate locally predominantly into alternatively activated (M2) macrophages, which are known for cell-clearance and phagocytosis, but may ultimately lead to fibrosis. Phagocytotic function of macrophages can be impaired by surface expression of cytoplasmic proteases on apoptotic neutrophils and causes release of inflammatory cytokines and immunogenic contents, presumably resulting in a vicious circle of increased neutrophil, T and B cell activation and consequent ANCA production. Considering their crucial role in initiating necroinflammation as well as fibrogenesis, monocytes and macrophages may represent a logic first-line target for new treatment options in AAV.
Collapse
Affiliation(s)
- Yosta Vegting
- Department of Internal Medicine, Section of Nephrology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
| | - Liffert Vogt
- Department of Internal Medicine, Section of Nephrology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Hans-Joachim Anders
- Department of Internal Medicine IV, Division of Nephrology, University Hospital of the Ludwig Maximilians University Munich, Munich, Germany
| | - Menno P J de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Frederike J Bemelman
- Department of Internal Medicine, Section of Nephrology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Marc L Hilhorst
- Department of Internal Medicine, Section of Nephrology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
7
|
Immunopathogenesis of ANCA-Associated Vasculitis. Int J Mol Sci 2020; 21:ijms21197319. [PMID: 33023023 PMCID: PMC7584042 DOI: 10.3390/ijms21197319] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/26/2020] [Accepted: 09/30/2020] [Indexed: 12/11/2022] Open
Abstract
Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis is an autoimmune disorder which affects small- and, to a lesser degree, medium-sized vessels. ANCA-associated vasculitis encompasses three disease phenotypes: granulomatosis with polyangiitis (GPA), microscopic polyangiitis (MPA), and eosinophilic granulomatosis with polyangiitis (EGPA). This classification is largely based on clinical presentations and has several limitations. Recent research provided evidence that genetic background, risk of relapse, prognosis, and co-morbidities are more closely related to the ANCA serotype, proteinase 3 (PR3)-ANCA and myeloperoxidase (MPO)-ANCA, compared to the disease phenotypes GPA or MPA. This finding has been extended to the investigation of biomarkers predicting disease activity, which again more closely relate to the ANCA serotype. Discoveries related to the immunopathogenesis translated into clinical practice as targeted therapies are on the rise. This review will summarize the current understanding of the immunopathogenesis of ANCA-associated vasculitis and the interplay between ANCA serotype and proposed disease biomarkers and illustrate how the extending knowledge of the immunopathogenesis will likely translate into development of a personalized medicine approach in the management of ANCA-associated vasculitis.
Collapse
|
8
|
Neves KB, Montezano AC, Lang NN, Touyz RM. Vascular toxicity associated with anti-angiogenic drugs. Clin Sci (Lond) 2020; 134:2503-2520. [PMID: 32990313 DOI: 10.1042/cs20200308] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
Over the past two decades, the treatment of cancer has been revolutionised by the highly successful introduction of novel molecular targeted therapies and immunotherapies, including small-molecule kinase inhibitors and monoclonal antibodies that target angiogenesis by inhibiting vascular endothelial growth factor (VEGF) signaling pathways. Despite their anti-angiogenic and anti-cancer benefits, the use of VEGF inhibitors (VEGFi) and other tyrosine kinase inhibitors (TKIs) has been hampered by potent vascular toxicities especially hypertension and thromboembolism. Molecular processes underlying VEGFi-induced vascular toxicities still remain unclear but inhibition of endothelial NO synthase (eNOS), reduced nitric oxide (NO) production, oxidative stress, activation of the endothelin system, and rarefaction have been implicated. However, the pathophysiological mechanisms still remain elusive and there is an urgent need to better understand exactly how anti-angiogenic drugs cause hypertension and other cardiovascular diseases (CVDs). This is especially important because VEGFi are increasingly being used in combination with other anti-cancer dugs, such as immunotherapies (immune checkpoint inhibitors (ICIs)), other TKIs, drugs that inhibit epigenetic processes (histone deacetylase (HDAC) inhibitor) and poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitors, which may themselves induce cardiovascular injury. Here, we discuss vascular toxicities associated with TKIs, especially VEGFi, and provide an up-to-date overview on molecular mechanisms underlying VEGFi-induced vascular toxicity and cardiovascular sequelae. We also review the vascular effects of VEGFi when used in combination with other modern anti-cancer drugs.
Collapse
Affiliation(s)
- Karla B Neves
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| | - Ninian N Lang
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| |
Collapse
|
9
|
Fukui S, Ichinose K, Sada KE, Miyamoto J, Harigai M, Amano K, Atsumi T, Takasaki Y, Dobashi H, Arimura Y, Hasegawa H, Yuzawa Y, Yamagata K, Tsuboi N, Maruyama S, Matsuo S, Makino H, Maeda T, Kawakami A. Complement profile in microscopic polyangiitis and granulomatosis with polyangiitis: analysis using sera from a nationwide prospective cohort study. Scand J Rheumatol 2020; 49:301-311. [PMID: 32286129 DOI: 10.1080/03009742.2019.1695927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVE The complement cascade, especially the alternative pathway of complement, has been shown in basic research to be associated with anti-neutrophil cytoplasmic autoantibody (ANCA)-associated vasculitis (AAV). We aimed to elucidate relationships between serum complement components and clinical characteristics in AAV. METHOD In a nationwide prospective cohort study (RemIT-JAV-RPGN), we measured the serum levels of C1q, C2, C3, C3b/iC3b, C4, C4b, C5, C5a, C9, factor B, factor D, factor H, factor I, mannose-binding lectin, and properdin in 52 patients with microscopic polyangiitis (MPA) and 39 patients with granulomatosis with polyangiitis (GPA). RESULTS The properdin level of MPA and GPA was significantly lower than that of healthy donors. The properdin level was negatively correlated with the Birmingham Vasculitis Activity Score (BVAS) (ρ = -0.2148, p = 0.0409). The factor D level at 6 months was significantly positively correlated with the Vasculitis Damage Index (VDI) at 6, 12, and 24 months (ρ = 0.4207, 0.4132, and 0.3115, respectively). Patients with a higher ratio of C5a to C5 had higher neutrophil percentage and serum immunoglobulin G levels, and significantly lower creatinine levels. Cluster analysis divided the MPA and GPA patients into three subgroups. A principal component (PC) analysis aggregated 15 types of complements into alternative pathway-related PC 1 and complement classical pathway and common pathway-related PC 2. CONCLUSIONS The serum levels of properdin and factor D were correlated with the BVAS and the VDI in MPA and GPA, respectively. Our analyses suggested the pathological heterogeneity of MPA and GPA from the aspect of complement components.
Collapse
Affiliation(s)
- S Fukui
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences , Nagasaki, Japan.,Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences , Nagasaki, Japan
| | - K Ichinose
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences , Nagasaki, Japan
| | - K-E Sada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Okayama, Japan
| | - J Miyamoto
- Nagasaki University Hospital Clinical Research Center , Nagasaki, Japan
| | - M Harigai
- Division of Epidemiology and Pharmacoepidemiology of Rheumatic Diseases, Department of Rheumatology, School of Medicine, Tokyo Women's Medical University , Tokyo, Japan
| | - K Amano
- Department of Rheumatology and Clinical Immunology, Saitama Medical Center, Saitama Medical University , Kawagoe, Japan
| | - T Atsumi
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University , Sapporo, Japan
| | - Y Takasaki
- Department of Rheumatology, Graduate School of Medicine, Juntendo University , Tokyo, Japan
| | - H Dobashi
- Division of Endocrinology and Metabolism, Haematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University , Miki-cho, Japan
| | - Y Arimura
- Nephrology and Rheumatology, First Department of Internal Medicine, Kyorin University School of Medicine , Tokyo, Japan
| | - H Hasegawa
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine , Toon, Japan
| | - Y Yuzawa
- Department of Nephrology, Fujita Health University School of Medicine , Toyoake, Japan
| | - K Yamagata
- Department of Nephrology, Faculty of Medicine, University of Tsukuba , Tsukuba, Japan
| | - N Tsuboi
- Department of Nephrology, Internal Medicine, Nagoya University Graduate School of Medicine , Nagoya, Japan
| | - S Maruyama
- Department of Nephrology, Internal Medicine, Nagoya University Graduate School of Medicine , Nagoya, Japan
| | - S Matsuo
- Department of Nephrology, Internal Medicine, Nagoya University Graduate School of Medicine , Nagoya, Japan
| | - H Makino
- Okayama University , Okayama, Japan
| | - T Maeda
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences , Nagasaki, Japan.,Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences , Nagasaki, Japan
| | - A Kawakami
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences , Nagasaki, Japan
| |
Collapse
|
10
|
Abstract
The kidney harbours different types of endothelia, each with specific structural and functional characteristics. The glomerular endothelium, which is highly fenestrated and covered by a rich glycocalyx, participates in the sieving properties of the glomerular filtration barrier and in the maintenance of podocyte structure. The microvascular endothelium in peritubular capillaries, which is also fenestrated, transports reabsorbed components and participates in epithelial cell function. The endothelium of large and small vessels supports the renal vasculature. These renal endothelia are protected by regulators of thrombosis, inflammation and complement, but endothelial injury (for example, induced by toxins, antibodies, immune cells or inflammatory cytokines) or defects in factors that provide endothelial protection (for example, regulators of complement or angiogenesis) can lead to acute or chronic renal injury. Moreover, renal endothelial cells can transition towards a mesenchymal phenotype, favouring renal fibrosis and the development of chronic kidney disease. Thus, the renal endothelium is both a target and a driver of kidney and systemic cardiovascular complications. Emerging therapeutic strategies that target the renal endothelium may lead to improved outcomes for both rare and common renal diseases.
Collapse
|
11
|
Zsengellér ZK, Lo A, Tavasoli M, Pernicone E, Karumanchi SA, Rosen S. Soluble fms-Like Tyrosine Kinase 1 Localization in Renal Biopsies of CKD. Kidney Int Rep 2019; 4:1735-1741. [PMID: 31844810 PMCID: PMC6895657 DOI: 10.1016/j.ekir.2019.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 07/19/2019] [Accepted: 08/06/2019] [Indexed: 01/15/2023] Open
Abstract
Introduction Soluble fms-like tyrosine kinase 1 (sFLT1) is a splice variant of the vascular endothelial growth factor (VEGF) receptor lacking the transmembrane and cytoplasmic domains and acts as a powerful antagonist of VEGF signaling. Plasma sFLT1 levels are higher in patients with chronic kidney disease (CKD) and correlate with renal dysfunction. The source of plasma sFLT1 in CKD is unclear. Methods Fifty-two renal biopsies were studied for sFLT1 expression using immunohistochemistry and evaluated on a 0–4 grading scale of positive cells within inflammatory infiltrates. These included drug-induced interstitial nephritis (6); allografts (12), with polyomavirus nephritis (3); diabetes mellitus (10); lupus glomerulonephritis (6); pauci-immune vasculitis (7); IgA nephropathy (6); and miscellaneous CKD (5). Results Forty-seven biopsies had inflammatory infiltrates of which 37 had sFLT1-positive cells: of these biopsies, 3 were grade 4, i.e., had cells that constituted more than 50% of the inflammatory infiltrate, 9 were grade 3 (25%–50%), 5 were grade 2 (10%–25%), 3 were grade 1 (10%), and 17 were grade 0.5 (<10%). There was a robust correlation (r2 = 0.89) between degree of inflammation and sFLT1-positive cells. CD68/sFLT1 co-immunostaining studies indicated that sFLT1-positive cells were histiocytes. The surrounding capillary network was reduced. Conclusion sFLT1-positive histiocytes are generally part of the inflammatory infiltrates noted in CKD and are particularly abundant in forms of interstitial nephritis. Their presence promotes an anti-angiogenic state locally in the tubulointerstitium that could inhibit capillary repair, contribute to peritubular capillary loss, and enhance fibrosis in CKD.
Collapse
Affiliation(s)
- Zsuzsanna K Zsengellér
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachussetts, USA
| | - Agnes Lo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachussetts, USA
| | - Mahtab Tavasoli
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachussetts, USA
| | - Elizabeth Pernicone
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachussetts, USA
| | - S Ananth Karumanchi
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachussetts, USA.,Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Seymour Rosen
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachussetts, USA
| |
Collapse
|
12
|
Wewers TM, Mayer AB, Pfleiderer A, Beul K, Schmidt R, Heitplatz B, Van Marck V, Nolte I, Pavenstädt H, Reuter S, Brand M, Di Marco GS. Increased soluble fms-like tyrosine kinase 1 after ischemia reperfusion contributes to adverse clinical outcomes following kidney transplantation. Kidney Int 2019; 95:1091-1102. [PMID: 30824181 DOI: 10.1016/j.kint.2018.11.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/05/2018] [Accepted: 11/02/2018] [Indexed: 12/20/2022]
Abstract
Renal ischemia reperfusion injury (IRI) adversely affects clinical outcomes following kidney transplantation. Understanding the cellular mechanisms and the changes in gene/protein expression following IRI may help to improve these outcomes. Serum soluble fms-like tyrosine kinase 1 (sFlt-1), a circulating antiangiogenic protein, is increased in the first week following kidney transplantation. We evaluated the casual relationship of elevated sFlt-1 levels with renal microvascular dysfunction following IRI in a longitudinal study of 93 kidney transplant recipients and in several animal models. Transplant recipients with higher sFlt-1 levels had higher odds of delayed graft function, graft rejection, impaired graft function, and death. In a subgroup of 25 participants who underwent kidney biopsy within 4 months of kidney transplantation, peritubular capillary area was lower in those with elevated serum sFtl-1 levels. The administration of recombinant sFlt-1 into rodents resulted in significant structural and functional changes of the renal microvasculature, including reduced peritubular capillary density and intracapillary blood volume, and lead to increased expression of inflammatory genes and increased fibrosis. In a murine model of IRI, the kidney was a site of sFlt-1 production, and systemic neutralization of sFlt-1 preserved peritubular capillary density and alleviated renal fibrosis. Our data indicate that high sFlt-1 levels after IRI play an important role in the pathogenesis of microvascular dysfunction, thereby contributing to adverse clinical outcomes following kidney transplantation.
Collapse
Affiliation(s)
- Theresa M Wewers
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany; Small Animal Hospital, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Anna B Mayer
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Alexander Pfleiderer
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Katrin Beul
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Rene Schmidt
- Institute for Biostatistics and Clinical Research, University Hospital Münster, Münster, Germany
| | - Barbara Heitplatz
- Department of Pathology, University Hospital Münster, Münster, Germany
| | - Veerle Van Marck
- Department of Pathology, University Hospital Münster, Münster, Germany
| | - Ingo Nolte
- Small Animal Hospital, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Hermann Pavenstädt
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Stefan Reuter
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Marcus Brand
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Giovana S Di Marco
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany.
| |
Collapse
|
13
|
Sun XJ, Chen M, Zhao MH. Thrombin Contributes to Anti-myeloperoxidase Antibody Positive IgG-Mediated Glomerular Endothelial Cells Activation Through SphK1-S1P-S1PR3 Signaling. Front Immunol 2019; 10:237. [PMID: 30891029 PMCID: PMC6413724 DOI: 10.3389/fimmu.2019.00237] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/28/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Activation of coagulation system plays an important role in antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) pathogenesis. Thrombin, generated during coagulation could disrupt endothelial barrier integrity through protease-activated receptor 1 (PAR1). Our previous study found that sphingosine-1-phosphate (S1P) contributed to myeloperoxidase (MPO)-ANCA-positive IgG-induced glomerular endothelial cell (GEnC) activation through a S1P receptor (S1PR)-dependent route. In recent years, S1P signaling was reported to be involved in thrombin effects on endothelial cells. This current study investigated whether the interaction between thrombin-PAR and S1P-S1PR signaling contributed to MPO-ANCA-positive IgG-induced GEnC dysfunction. Methods: The effect of thrombin on GEnC activation was analyzed from three aspects. First, morphological alteration of GEnCs was observed. Second, permeability assay was performed to determine GEnC monolayer activation quantitatively. Third, endothelin-1 (ET-1) levels were measured. Expression levels of sphingosine kinases (SphKs) and S1PRs were detected. In addition, antagonists of PAR1 and S1PR3 were employed to determine their roles. Eventually, PAR1 and tissue factor (TF) expression levels as well as TF procoagulant activity were analyzed. Results: Thrombin induced further damage of tight junction, increase in endothelial monolayer permeability as well as upregulation of ET-1 levels in GEnCs stimulated with MPO-ANCA-positive IgG. Blocking PAR1 downregulated ET-1 levels in the supernatants of GEnCs treated by thrombin plus MPO-ANCA-positive IgG. Expression levels of SphK1, S1PR3 increased significantly in GEnCs treated with thrombin plus MPO-ANCA-positive IgG. S1P upregulated PAR1 and TF expression, and enhanced procoagulant activity of TF in MPO-ANCA-positive IgG-stimulated GEnCs. Conclusion: Thrombin synergized with SphK1-S1P-S1PR3 signaling pathway to enhance MPO-ANCA-positive IgG-mediated GEnC activation.
Collapse
Affiliation(s)
- Xiao-Jing Sun
- Renal Division, Department of Medicine, Peking University, First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Min Chen
- Renal Division, Department of Medicine, Peking University, First Hospital, Peking University Institute of Nephrology, Beijing, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| | - Ming-Hui Zhao
- Renal Division, Department of Medicine, Peking University, First Hospital, Peking University Institute of Nephrology, Beijing, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China.,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| |
Collapse
|
14
|
Jourde-Chiche N, Fakhouri F, Dou L, Bellien J, Burtey S, Frimat M, Jarrot PA, Kaplanski G, Le Quintrec M, Pernin V, Rigothier C, Sallée M, Fremeaux-Bacchi V, Guerrot D, Roumenina LT. Endothelium structure and function in kidney health and disease. Nat Rev Nephrol 2019. [PMID: 30607032 DOI: 10.1038/s4158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
The kidney harbours different types of endothelia, each with specific structural and functional characteristics. The glomerular endothelium, which is highly fenestrated and covered by a rich glycocalyx, participates in the sieving properties of the glomerular filtration barrier and in the maintenance of podocyte structure. The microvascular endothelium in peritubular capillaries, which is also fenestrated, transports reabsorbed components and participates in epithelial cell function. The endothelium of large and small vessels supports the renal vasculature. These renal endothelia are protected by regulators of thrombosis, inflammation and complement, but endothelial injury (for example, induced by toxins, antibodies, immune cells or inflammatory cytokines) or defects in factors that provide endothelial protection (for example, regulators of complement or angiogenesis) can lead to acute or chronic renal injury. Moreover, renal endothelial cells can transition towards a mesenchymal phenotype, favouring renal fibrosis and the development of chronic kidney disease. Thus, the renal endothelium is both a target and a driver of kidney and systemic cardiovascular complications. Emerging therapeutic strategies that target the renal endothelium may lead to improved outcomes for both rare and common renal diseases.
Collapse
Affiliation(s)
- Noemie Jourde-Chiche
- Aix-Marseille University, Centre de Nephrologie et Transplantation Renale, AP-HM Hopital de la Conception, Marseille, France.
- Aix-Marseille University, C2VN, INSERM 1263, Institut National de la Recherche Agronomique (INRA) 1260, Faculte de Pharmacie, Marseille, France.
| | - Fadi Fakhouri
- Centre de Recherche en Transplantation et Immunologie, INSERM, Université de Nantes and Department of Nephrology, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Laetitia Dou
- Aix-Marseille University, C2VN, INSERM 1263, Institut National de la Recherche Agronomique (INRA) 1260, Faculte de Pharmacie, Marseille, France
| | - Jeremy Bellien
- Department of Pharmacology, Rouen University Hospital and INSERM, Normandy University, Université de Rouen Normandie, Rouen, France
| | - Stéphane Burtey
- Aix-Marseille University, Centre de Nephrologie et Transplantation Renale, AP-HM Hopital de la Conception, Marseille, France
- Aix-Marseille University, C2VN, INSERM 1263, Institut National de la Recherche Agronomique (INRA) 1260, Faculte de Pharmacie, Marseille, France
| | - Marie Frimat
- Université de Lille, INSERM, Centre Hospitalier Universitaire de Lille, U995, Lille Inflammation Research International Center (LIRIC), Lille, France
- Nephrology Department, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Pierre-André Jarrot
- Aix-Marseille University, C2VN, INSERM 1263, Institut National de la Recherche Agronomique (INRA) 1260, Faculte de Pharmacie, Marseille, France
- Assistance Publique-Hôpitaux de Marseille, Service de Médecine Interne et d'Immunologie Clinique, Hôpital de La Conception, Marseille, France
| | - Gilles Kaplanski
- Aix-Marseille University, C2VN, INSERM 1263, Institut National de la Recherche Agronomique (INRA) 1260, Faculte de Pharmacie, Marseille, France
- Assistance Publique-Hôpitaux de Marseille, Service de Médecine Interne et d'Immunologie Clinique, Hôpital de La Conception, Marseille, France
| | - Moglie Le Quintrec
- Centre Hospitalier Universitaire de Lapeyronie, Département de Néphrologie Dialyse et Transplantation Rénale, Montpellier, France
- Institute for Regenerative Medicine and Biotherapy (IRMB), Montpellier, France
| | - Vincent Pernin
- Centre Hospitalier Universitaire de Lapeyronie, Département de Néphrologie Dialyse et Transplantation Rénale, Montpellier, France
- Institute for Regenerative Medicine and Biotherapy (IRMB), Montpellier, France
| | - Claire Rigothier
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France
- Service de Néphrologie Transplantation, Dialyse et Aphérèse, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Marion Sallée
- Aix-Marseille University, Centre de Nephrologie et Transplantation Renale, AP-HM Hopital de la Conception, Marseille, France
- Aix-Marseille University, C2VN, INSERM 1263, Institut National de la Recherche Agronomique (INRA) 1260, Faculte de Pharmacie, Marseille, France
| | - Veronique Fremeaux-Bacchi
- Assistance Publique-Hôpitaux de Paris, Service d'Immunologie Biologique, Hôpital Européen Georges Pompidou, Paris, France
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Dominique Guerrot
- Normandie Université, Université de Rouen Normandie, Rouen University Hospital, Department of Nephrology, Rouen, France
| | - Lubka T Roumenina
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France.
- Sorbonne Universités, Paris, France.
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
15
|
Hutton HL, Holdsworth SR, Kitching AR. ANCA-Associated Vasculitis: Pathogenesis, Models, and Preclinical Testing. Semin Nephrol 2018; 37:418-435. [PMID: 28863790 DOI: 10.1016/j.semnephrol.2017.05.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Our understanding of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis has developed greatly since the discovery of ANCA, directed against neutrophil components, in 1982. Observations in human disease, and increasingly sophisticated studies in vitro and in rodent models in vivo, have allowed a nuanced understanding of many aspects of the immunopathogenesis of disease, including the significance of ANCA as a diagnostic and monitoring tool as well as a mediator of microvascular injury. The mechanisms of leukocyte recruitment and tissue injury, and the role of T cells increasingly are understood. Unexpected findings, such as the role of complement, also have been uncovered through experimental studies and human observations. This review focusses on the pathogenesis of ANCA-associated vasculitis, highlighting the challenges in finding new, less-toxic treatments and potential therapeutic targets in this disease. The current suite of rodent models is reviewed, and future directions in the study of this complex and fascinating disease are suggested.
Collapse
Affiliation(s)
- Holly L Hutton
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia; Department of Nephrology, Monash Health, Clayton, Victoria, Australia
| | - Stephen R Holdsworth
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia; Department of Nephrology, Monash Health, Clayton, Victoria, Australia
| | - A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia; Department of Nephrology, Monash Health, Clayton, Victoria, Australia; Department of Pediatric Nephrology, Monash Children's Hospital, Clayton, Victoria, Australia.
| |
Collapse
|
16
|
Lamprecht P, Kerstein A, Klapa S, Schinke S, Karsten CM, Yu X, Ehlers M, Epplen JT, Holl-Ulrich K, Wiech T, Kalies K, Lange T, Laudien M, Laskay T, Gemoll T, Schumacher U, Ullrich S, Busch H, Ibrahim S, Fischer N, Hasselbacher K, Pries R, Petersen F, Weppner G, Manz R, Humrich JY, Nieberding R, Riemekasten G, Müller A. Pathogenetic and Clinical Aspects of Anti-Neutrophil Cytoplasmic Autoantibody-Associated Vasculitides. Front Immunol 2018; 9:680. [PMID: 29686675 PMCID: PMC5900791 DOI: 10.3389/fimmu.2018.00680] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 03/20/2018] [Indexed: 12/11/2022] Open
Abstract
Anti-neutrophil cytoplasmic autoantibodies (ANCA) targeting proteinase 3 (PR3) and myeloperoxidase expressed by innate immune cells (neutrophils and monocytes) are salient diagnostic and pathogenic features of small vessel vasculitis, comprising granulomatosis with polyangiitis (GPA), microscopic polyangiitis, and eosinophilic GPA. Genetic studies suggest that ANCA-associated vasculitides (AAV) constitute separate diseases, which share common immunological and pathological features, but are otherwise heterogeneous. The successful therapeutic use of anti-CD20 antibodies emphasizes the prominent role of ANCA and possibly other autoantibodies in the pathogenesis of AAV. However, to elucidate causal effects in AAV, a better understanding of the complex interplay leading to the emergence of B lymphocytes that produce pathogenic ANCA remains a challenge. Different scenarios seem possible; e.g., the break of tolerance induced by a shift from non-pathogenic toward pathogenic autoantigen epitopes in inflamed tissue. This review gives a brief overview on current knowledge about genetic and epigenetic factors, barrier dysfunction and chronic non-resolving inflammation, necro-inflammatory auto-amplification of cellular death and inflammation, altered autoantigen presentation, alternative complement pathway activation, alterations within peripheral and inflamed tissue-residing T- and B-cell populations, ectopic lymphoid tissue neoformation, the characterization of PR3-specific T-cells, properties of ANCA, links between autoimmune disease and infection-triggered pathology, and animal models in AAV.
Collapse
Affiliation(s)
- Peter Lamprecht
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Anja Kerstein
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Sebastian Klapa
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Susanne Schinke
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Christian M Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Xinhua Yu
- Xiamen-Borstel Joint Laboratory of Autoimmunity, Medical College of Xiamen University, Xiamen, China.,Priority Area Asthma and Allergy, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Marc Ehlers
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutrition Medicine, University of Lübeck and University Medical Center Schleswig Holstein, Lübeck, Germany
| | - Jörg T Epplen
- Department of Human Genetics, Ruhr-University, Bochum, Germany.,University of Witten/Herdecke, ZBAF, Witten, Germany
| | | | - Thorsten Wiech
- Institute of Pathology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Kathrin Kalies
- Institute of Anatomy, University of Lübeck, Lübeck, Germany
| | - Tanja Lange
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Martin Laudien
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Kiel, Kiel, Germany
| | - Tamas Laskay
- Department for Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Timo Gemoll
- Department of Surgery, Section for Translational Surgical Oncology and Biobanking, University of Lübeck, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, Center for Experimental Medicine, University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sebastian Ullrich
- Institute of Anatomy and Experimental Morphology, Center for Experimental Medicine, University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Medical Department 3, Gastroenterology/Rheumatology, Municipal Hospital Kiel, Kiel, Germany
| | - Hauke Busch
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Saleh Ibrahim
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Nicole Fischer
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Ralph Pries
- Department of Otorhinolaryngology, University of Lübeck, Lübeck, Germany
| | - Frank Petersen
- Priority Area Asthma and Allergy, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Gesche Weppner
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Rudolf Manz
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Jens Y Humrich
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Relana Nieberding
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Antje Müller
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| |
Collapse
|
17
|
Brown KL, Lubieniecka JM, Armaroli G, Kessel K, Gibson KM, Graham J, Liu D, Hancock REW, Ross CJ, Benseler SM, Luqmani RA, Cabral DA, Foell D, Kessel C. S100A12 Serum Levels and PMN Counts Are Elevated in Childhood Systemic Vasculitides Especially Involving Proteinase 3 Specific Anti-neutrophil Cytoplasmic Antibodies. Front Pediatr 2018; 6:341. [PMID: 30533405 PMCID: PMC6266798 DOI: 10.3389/fped.2018.00341] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/22/2018] [Indexed: 11/29/2022] Open
Abstract
Objectives: Chronic primary systemic vasculitidies (CPV) are a collection of rare diseases involving inflammation in blood vessels, often in multiple organs. CPV can affect adults and children and may be life- or organ-threatening. Treatments for adult CPV, although effective, have known severe potential toxicities; safety and efficacy of these drugs in pediatric patients is not fully understood. There is an unmet need for biologic measures to assess the level of disease activity and, in turn, inform treatment choices for stopping, starting, or modifying therapy. This observational study determines if S100 calcium-binding protein A12 (S100A12) and common inflammatory indicators are sensitive markers of disease activity in children and adolescents with CPV that could be used to inform a minimal effective dose of therapy. Methods: Clinical data and sera were collected from 56 participants with CPV at study visits from diagnosis to remission. Serum concentrations of S100A12, C-reactive protein (CRP) and hemoglobin (Hb) as well as whole blood cell counts and erythrocyte sedimentation rate (ESR) were measured. Disease activity was inferred by physician's global assessment (PGA) and the pediatric vasculitis activity score (PVAS). Results: Serum concentrations of standard markers of inflammation (ESR, CRP, Hb, absolute blood neutrophil count), and S100A12 track with clinically assessed disease activity. These measures-particularly neutrophil counts and sera concentrations of S100A12-had the most significant correlation with clinical scores of disease activity in those children with vasculitis that is associated with anti-neutrophil cytoplasmic antibodies (ANCA) against proteinase 3. Conclusions: S100A12 and neutrophil counts should be considered in the assessment of disease activity in children with CPV particularly the most common forms of the disease that involve proteinase 3 ANCA. Key messages: - In children with chronic primary systemic vasculitis (CPV), classical measures of inflammation are not formally considered in scoring of disease activity. - Inflammatory markers-specifically S100A12 and neutrophil count-track preferentially with the most common forms of childhood CPV which affect small to medium sized vessels and involve anti neutrophil cytoplasmic antibodies (ANCA) against proteinase-3.
Collapse
Affiliation(s)
- Kelly L Brown
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.,British Columbia Children's Hospital, Vancouver, BC, Canada
| | - Joanna M Lubieniecka
- Department of Statistics and Actuarial Science, Simon Fraser University, Burnaby, BC, Canada
| | - Giulia Armaroli
- Department of Pediatric Rheumatology and Immunology, University Hospital Muenster, Muenster, Germany
| | - Katharina Kessel
- Department of Pediatric Rheumatology and Immunology, University Hospital Muenster, Muenster, Germany
| | - Kristen M Gibson
- British Columbia Children's Hospital, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Jinko Graham
- Department of Statistics and Actuarial Science, Simon Fraser University, Burnaby, BC, Canada
| | - Dongmeng Liu
- Department of Statistics and Actuarial Science, Simon Fraser University, Burnaby, BC, Canada
| | - Robert E W Hancock
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada.,Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Colin J Ross
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.,Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Susanne M Benseler
- Department of Pediatrics, Alberta Children's Hospital, Calgary, AB, Canada
| | - Raashid A Luqmani
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - David A Cabral
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.,British Columbia Children's Hospital, Vancouver, BC, Canada
| | - Dirk Foell
- Department of Pediatric Rheumatology and Immunology, University Hospital Muenster, Muenster, Germany
| | - Christoph Kessel
- Department of Pediatric Rheumatology and Immunology, University Hospital Muenster, Muenster, Germany
| |
Collapse
|
18
|
Pathogenesis and treatment of ANCA-associated vasculitis-a role for complement. Pediatr Nephrol 2018; 33:1-11. [PMID: 27596099 DOI: 10.1007/s00467-016-3475-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 05/18/2016] [Accepted: 05/25/2016] [Indexed: 01/20/2023]
Abstract
The antineutrophil cytoplasm autoantibody (ANCA)-associated vasculitides (AAV), although rare in childhood, can have devastating effects on affected organs, especially the kidney. In this review we present an update on the pathogenesis and treatment of ANCA vasculitis, with a particular emphasis on the role of the alternative pathway of complement. The rationale and evidence for the current treatment strategies are summarized. Targeting the activation of neutrophils by the anaphylatoxin C5a may serve as an additional therapeutic strategy, however the results of clinical studies are awaited.
Collapse
|
19
|
Ma TKW, McAdoo SP, Tam FWK. Targeting the tyrosine kinase signalling pathways for treatment of immune-mediated glomerulonephritis: from bench to bedside and beyond. Nephrol Dial Transplant 2017; 32:i129-i138. [PMID: 28391340 PMCID: PMC5410974 DOI: 10.1093/ndt/gfw336] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 08/18/2016] [Indexed: 12/25/2022] Open
Abstract
Glomerulonephritis (GN) affects patients of all ages and is an important cause of morbidity and mortality. Non-selective immunosuppressive drugs have been used in immune-mediated GN but often result in systemic side effects and occasionally fatal infective complications. There is increasing evidence from both preclinical and clinical studies that abnormal activation of receptor and non-receptor tyrosine kinase signalling pathways are implicated in the pathogenesis of immune-mediated GN. Activation of spleen tyrosine kinase (SYK), Bruton's tyrosine kinase (BTK), platelet-derived growth factor receptor (PDGFR), epidermal growth factor receptor (EGFR) and discoidin domain receptor 1 (DDR1) have been demonstrated in anti-GBM disease. SYK is implicated in the pathogenesis of ANCA-associated GN. SYK, BTK, PDGFR, EFGR, DDR1 and Janus kinase are implicated in the pathogenesis of lupus nephritis. A representative animal model of IgA nephropathy (IgAN) is lacking. Based on the results from in vitro and human renal biopsy study results, a phase II clinical trial is ongoing to evaluate the efficacy and safety of fostamatinib (an oral SYK inhibitor) in high-risk IgAN patient. Various tyrosine kinase inhibitors (TKIs) have been approved for cancer treatment. Clinical trials of TKIs in GN may be justified given their long-term safety data. In this review we will discuss the current unmet medical needs in GN treatment and research as well as the current stage of development of TKIs in GN treatment and propose an accelerated translational research approach to investigate whether selective inhibition of tyrosine kinase provides a safer and more efficacious option for GN treatment.
Collapse
Affiliation(s)
- Terry King-Wing Ma
- Renal and Vascular Inflammation Section, Department of Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK.,Carol and Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine and Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Stephen P McAdoo
- Renal and Vascular Inflammation Section, Department of Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK
| | - Frederick Wai Keung Tam
- Renal and Vascular Inflammation Section, Department of Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK
| |
Collapse
|
20
|
Monocytes Promote Crescent Formation in Anti-Myeloperoxidase Antibody–Induced Glomerulonephritis. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1908-1915. [DOI: 10.1016/j.ajpath.2017.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/27/2017] [Accepted: 05/02/2017] [Indexed: 11/20/2022]
|
21
|
Choi M, Schreiber A, Eulenberg-Gustavus C, Scheidereit C, Kamps J, Kettritz R. Endothelial NF- κB Blockade Abrogates ANCA-Induced GN. J Am Soc Nephrol 2017; 28:3191-3204. [PMID: 28687535 DOI: 10.1681/asn.2016060690] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 05/18/2017] [Indexed: 01/15/2023] Open
Abstract
ANCA-associated vasculitis (AAV) is a highly inflammatory condition in which ANCA-activated neutrophils interact with the endothelium, resulting in necrotizing vasculitis. We tested the hypothesis that endothelial NF-κB mediates necrotizing crescentic GN (NCGN) and provides a specific treatment target. Reanalysis of kidneys from previously examined murine NCGN disease models revealed NF-κB activation in affected kidneys, mostly as a p50/p65 heterodimer, and increased renal expression of NF-κB-dependent tumor necrosis factor α (TNF-α). NF-κB activation positively correlated with crescent formation, and nuclear phospho-p65 staining showed NF-κB activation within CD31-expressing endothelial cells (ECs) in affected glomeruli. Therefore, we studied the effect of ANCA on NF-κB activation in neutrophil/EC cocultures in vitro ANCA did not activate NF-κB in primed human neutrophils, but ANCA-stimulated primed neutrophils activated NF-κB in ECs, at least in part via TNF-α release. This effect increased endothelial gene transcription and protein production of NF-κB-regulated interleukin-8. Moreover, upregulation of endothelial NF-κB promoted neutrophil adhesion to EC monolayers, an effect that was inhibited by a specific IKKβ inhibitor. In a murine NCGN model, prophylactic application of E-selectin-targeted immunoliposomes packed with p65 siRNA to downregulate endothelial NF-κB significantly reduced urine abnormalities, renal myeloid cell influx, and NCGN. Increased glomerular endothelial phospho-p65 staining in patients with AAV indicated that NF-κB is activated in human NCGN also. We suggest that ANCA-stimulated neutrophils activate endothelial NF-κB, which contributes to NCGN and provides a potential therapeutic target in AAV.
Collapse
Affiliation(s)
- Mira Choi
- Experimental and Clinical Research Center, the Charité Universitätsmedizin Berlin and the Max-Delbrück Center for Molecular Medicine at the Charité, Berlin, Germany; .,Nephrology and Internal Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Adrian Schreiber
- Experimental and Clinical Research Center, the Charité Universitätsmedizin Berlin and the Max-Delbrück Center for Molecular Medicine at the Charité, Berlin, Germany.,Nephrology and Internal Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Eulenberg-Gustavus
- Experimental and Clinical Research Center, the Charité Universitätsmedizin Berlin and the Max-Delbrück Center for Molecular Medicine at the Charité, Berlin, Germany
| | | | - Jan Kamps
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ralph Kettritz
- Experimental and Clinical Research Center, the Charité Universitätsmedizin Berlin and the Max-Delbrück Center for Molecular Medicine at the Charité, Berlin, Germany.,Nephrology and Internal Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
22
|
Braudeau C, Néel A, Amouriaux K, Martin JC, Rimbert M, Besançon A, Giraudet S, Terrien C, Aliaga M, Salabert-Le Guen N, Hémont C, Hamidou M, Josien R. Dysregulated Responsiveness of Circulating Dendritic Cells to Toll-Like Receptors in ANCA-Associated Vasculitis. Front Immunol 2017; 8:102. [PMID: 28232832 PMCID: PMC5298972 DOI: 10.3389/fimmu.2017.00102] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/20/2017] [Indexed: 02/02/2023] Open
Abstract
Objective Dendritic cells (DCs) are critical effectors of innate and adaptive immunity playing crucial roles in autoimmune responses. We previously showed that blood DC numbers were reduced in autoimmune antineutrophil cytoplasmic autoantibody-associated vasculitis (AAV). Here, we assessed toll-like receptor (TLR) responsiveness of blood DCs from patients with granulomatosis with polyangiitis (GPA) or microscopic polyangiitis (MPA). Methods Blood samples from healthy controls (HCs), GPA, or MPA patients, without treatment, during acute phase (AP) or remission phase (RP) were analyzed. Cytokine production by DCs and T cells was assessed on whole blood by flow cytometry after TLRs or polyclonal stimulation, respectively. Results We first showed that GPA and MPA are associated with a decreased blood DC number during AP. Conventional DCs (cDCs) from patients with GPA and MPA in AP exhibited a profound decrease of IL-12/IL-23p40 production after TLR3, 4, or 7/8 stimulation compared to patients in remission and HC, with a return to normal values in RP. TNFα secretion was also affected, with a decrease in cDCs from GPA patients in AP after TLR3 stimulation but an increase after TLR7/8 stimulation. By contrast, the responsiveness of plasmacytoid DCs to TLR7 and 9 was only marginally affected. Finally, we observed that IFNγ-producing CD4+ T cell frequency was significantly lower in AP-GPA patients than in HC. Conclusion We describe, for the first time, a dysregulated response to TLRs of circulating DCs in AAV patients mostly affecting cDCs that exhibit an unexpected reduced inflammatory cytokine secretion possibly contributing to an altered Th cell response.
Collapse
Affiliation(s)
- Cécile Braudeau
- CIMNA, Laboratoire d'Immunologie, CHU Nantes, Nantes, France; Institut de Transplantation-Urologie-Néphrologie (ITUN), CHU Nantes, Nantes, France; Centre de Recherche en Transplantation et Immunologie (UMR1064), INSERM, Université de Nantes, Nantes, France
| | - Antoine Néel
- Centre de Recherche en Transplantation et Immunologie (UMR1064), INSERM, Université de Nantes, Nantes, France; Faculté de Médecine, Université de Nantes, Nantes, France; Service de Médecine Interne, CHU Nantes, Nantes, France
| | - Karine Amouriaux
- CIMNA, Laboratoire d'Immunologie, CHU Nantes, Nantes, France; LabEx Immunotherapy Graft Oncology (IGO), Nantes, France
| | - Jérôme C Martin
- CIMNA, Laboratoire d'Immunologie, CHU Nantes, Nantes, France; Institut de Transplantation-Urologie-Néphrologie (ITUN), CHU Nantes, Nantes, France; Centre de Recherche en Transplantation et Immunologie (UMR1064), INSERM, Université de Nantes, Nantes, France; Faculté de Médecine, Université de Nantes, Nantes, France
| | - Marie Rimbert
- CIMNA, Laboratoire d'Immunologie, CHU Nantes, Nantes, France; Institut de Transplantation-Urologie-Néphrologie (ITUN), CHU Nantes, Nantes, France; Centre de Recherche en Transplantation et Immunologie (UMR1064), INSERM, Université de Nantes, Nantes, France
| | - Audrey Besançon
- CIMNA, Laboratoire d'Immunologie, CHU Nantes , Nantes , France
| | | | | | - Marine Aliaga
- CIMNA, Laboratoire d'Immunologie, CHU Nantes , Nantes , France
| | - Nina Salabert-Le Guen
- CIMNA, Laboratoire d'Immunologie, CHU Nantes, Nantes, France; Centre de Recherche en Transplantation et Immunologie (UMR1064), INSERM, Université de Nantes, Nantes, France; LabEx Immunotherapy Graft Oncology (IGO), Nantes, France
| | - Caroline Hémont
- CIMNA, Laboratoire d'Immunologie, CHU Nantes, Nantes, France; Institut de Transplantation-Urologie-Néphrologie (ITUN), CHU Nantes, Nantes, France; Centre de Recherche en Transplantation et Immunologie (UMR1064), INSERM, Université de Nantes, Nantes, France
| | - Mohamed Hamidou
- Centre de Recherche en Transplantation et Immunologie (UMR1064), INSERM, Université de Nantes, Nantes, France; Faculté de Médecine, Université de Nantes, Nantes, France; Service de Médecine Interne, CHU Nantes, Nantes, France
| | - Régis Josien
- CIMNA, Laboratoire d'Immunologie, CHU Nantes, Nantes, France; Institut de Transplantation-Urologie-Néphrologie (ITUN), CHU Nantes, Nantes, France; Centre de Recherche en Transplantation et Immunologie (UMR1064), INSERM, Université de Nantes, Nantes, France; Faculté de Médecine, Université de Nantes, Nantes, France; LabEx Immunotherapy Graft Oncology (IGO), Nantes, France
| |
Collapse
|
23
|
Goland S, Weinstein JM, Zalik A, Kuperstein R, Zilberman L, Shimoni S, Arad M, Ben Gal T, George J. Angiogenic Imbalance and Residual Myocardial Injury in Recovered Peripartum Cardiomyopathy Patients. Circ Heart Fail 2016; 9:CIRCHEARTFAILURE.116.003349. [DOI: 10.1161/circheartfailure.116.003349] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/29/2016] [Indexed: 01/08/2023]
Abstract
Background—
Recent studies suggest that angiogenic imbalance during pregnancy may lead to acute peripartum cardiomyopathy (PPCM). We propose that angiogenic imbalance and residual cardiac dysfunction may exist even after recovery from PPCM.
Methods and Results—
Twenty-nine women at least 12 months after presentation with PPCM, who exhibited recovery of left ventricular (LV) ejection fraction (≥50%), were included in the study (mean age 35±6 years, LV ejection fraction 61.0±3.9%). The number of circulating endothelial progenitor cells (EPCs) and plasma levels of proangiogenic vascular endothelial growth factor and of soluble vascular endothelial growth factor receptor Flt1 (sFlt1) were measured. All patients underwent comprehensive cardiac function assessment, including tissue Doppler imaging and 2-dimensional (2D) strain echocardiography. All measurements were compared with healthy controls. Patients with a history of PPCM have significantly higher sFlt1 concentrations (median [25th–75th percentile]; 149.57, [63.14–177.89] versus 20.29, [15.00–53.89] pg/mL,
P
<0.001) and significantly decreased vascular endothelial growth factor/sFlt1 ratio (
P
=0.012) compared with controls, with a trend toward lower concentration of circulating CD34
+
/KDR
+
levels. In addition, patients with PPCM had lower early velocities E′ septal (9.9±2.1 versus 11.0±1.5 cm/s,
P
=0.02), with a significantly lower systolic velocity S′ septal (7.6±1.2 versus 8.5±1.2 cm/s,
P
=0.003) by tissue Doppler imaging. Significantly lower LV global longitudinal (−19.1±3.3 versus −22.7±2.2%,
P
<0.001) and apical circumferential 2D strain (−16.6±4.9 versus −21.2±7.9,
P
=0.02) were present in patients with PPCM compared with controls.
Conclusions—
Higher concentration of sFlt1 with concomitant decreased circulating endothelial progenitor cell levels along with inappropriate attenuated vascular endothelial growth factor levels may imply an angiogenic imbalance that exists even after recovery and may thus predispose to PPCM. In addition, tissue Doppler imaging and 2D strain were able to identify residual myocardial injury in post-PPCM women with apparent recovery of LV systolic function. Both angiogenic imbalance and residual myocardial injury may play an important role in the recurrence of LV dysfunction during subsequent pregnancies.
Collapse
Affiliation(s)
- Sorel Goland
- From the Heart Institute, Kaplan Medical Center, Rehovot, Israel (S.G., A.Z., L.Z., S.S., J.G.); Heart Insitute, Sheba Medical Center, Ramat Gan, Israel (R.K., M.A.); Heart Insitute, Soroka Medical Center, Beersheba, Israel (J.M.W.); and Heart Insitute, Rabin Medical Center, Petah Tikva, Israel (T.B.G.)
| | - Jean Marc Weinstein
- From the Heart Institute, Kaplan Medical Center, Rehovot, Israel (S.G., A.Z., L.Z., S.S., J.G.); Heart Insitute, Sheba Medical Center, Ramat Gan, Israel (R.K., M.A.); Heart Insitute, Soroka Medical Center, Beersheba, Israel (J.M.W.); and Heart Insitute, Rabin Medical Center, Petah Tikva, Israel (T.B.G.)
| | - Adi Zalik
- From the Heart Institute, Kaplan Medical Center, Rehovot, Israel (S.G., A.Z., L.Z., S.S., J.G.); Heart Insitute, Sheba Medical Center, Ramat Gan, Israel (R.K., M.A.); Heart Insitute, Soroka Medical Center, Beersheba, Israel (J.M.W.); and Heart Insitute, Rabin Medical Center, Petah Tikva, Israel (T.B.G.)
| | - Rafael Kuperstein
- From the Heart Institute, Kaplan Medical Center, Rehovot, Israel (S.G., A.Z., L.Z., S.S., J.G.); Heart Insitute, Sheba Medical Center, Ramat Gan, Israel (R.K., M.A.); Heart Insitute, Soroka Medical Center, Beersheba, Israel (J.M.W.); and Heart Insitute, Rabin Medical Center, Petah Tikva, Israel (T.B.G.)
| | - Liaz Zilberman
- From the Heart Institute, Kaplan Medical Center, Rehovot, Israel (S.G., A.Z., L.Z., S.S., J.G.); Heart Insitute, Sheba Medical Center, Ramat Gan, Israel (R.K., M.A.); Heart Insitute, Soroka Medical Center, Beersheba, Israel (J.M.W.); and Heart Insitute, Rabin Medical Center, Petah Tikva, Israel (T.B.G.)
| | - Sara Shimoni
- From the Heart Institute, Kaplan Medical Center, Rehovot, Israel (S.G., A.Z., L.Z., S.S., J.G.); Heart Insitute, Sheba Medical Center, Ramat Gan, Israel (R.K., M.A.); Heart Insitute, Soroka Medical Center, Beersheba, Israel (J.M.W.); and Heart Insitute, Rabin Medical Center, Petah Tikva, Israel (T.B.G.)
| | - Michael Arad
- From the Heart Institute, Kaplan Medical Center, Rehovot, Israel (S.G., A.Z., L.Z., S.S., J.G.); Heart Insitute, Sheba Medical Center, Ramat Gan, Israel (R.K., M.A.); Heart Insitute, Soroka Medical Center, Beersheba, Israel (J.M.W.); and Heart Insitute, Rabin Medical Center, Petah Tikva, Israel (T.B.G.)
| | - Tuvia Ben Gal
- From the Heart Institute, Kaplan Medical Center, Rehovot, Israel (S.G., A.Z., L.Z., S.S., J.G.); Heart Insitute, Sheba Medical Center, Ramat Gan, Israel (R.K., M.A.); Heart Insitute, Soroka Medical Center, Beersheba, Israel (J.M.W.); and Heart Insitute, Rabin Medical Center, Petah Tikva, Israel (T.B.G.)
| | - Jacob George
- From the Heart Institute, Kaplan Medical Center, Rehovot, Israel (S.G., A.Z., L.Z., S.S., J.G.); Heart Insitute, Sheba Medical Center, Ramat Gan, Israel (R.K., M.A.); Heart Insitute, Soroka Medical Center, Beersheba, Israel (J.M.W.); and Heart Insitute, Rabin Medical Center, Petah Tikva, Israel (T.B.G.)
| |
Collapse
|
24
|
Allipour Birgani S, Mailänder M, Wasle I, Dietrich H, Gruber J, Distler O, Sgonc R. Efficient therapy of ischaemic lesions with VEGF121-fibrin in an animal model of systemic sclerosis. Ann Rheum Dis 2016; 75:1399-406. [PMID: 26362758 PMCID: PMC4766736 DOI: 10.1136/annrheumdis-2015-207548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 07/01/2015] [Accepted: 08/01/2015] [Indexed: 11/17/2022]
Abstract
BACKGROUND In systemic sclerosis (SSc), chronic and uncontrolled overexpression of vascular endothelial growth factor (VEGF) results in chaotic vessels, and intractable fingertip ulcers. Vice versa, VEGF is a potent mediator of angiogenesis if temporally and spatially controlled. We have addressed this therapeutic dilemma in SSc by a novel approach using a VEGF121 variant that covalently binds to fibrin and gets released on demand by cellular enzymatic activity. Using University of California at Davis (UCD)-206 chickens, we tested the hypothesis that cell-demanded release of fibrin-bound VEGF121 leads to the formation of stable blood vessels, and clinical improvement of ischaemic lesions. METHODS Ninety-one early and late ischaemic comb and neck skin lesions of UCD-206 chickens were treated locally with VEGF121-fibrin, fibrin alone, or left untreated. After 1 week of treatment the clinical outcome was assessed. Angiogenesis was studied by immunofluorescence staining of vascular markers quantitatively analysed using TissueQuest. RESULTS Overall, 79.3% of the lesions treated with VEGF121-fibrin showed clinical improvement, whereas 71.0% of fibrin treated controls, and 93.1% of untreated lesions deteriorated. This was accompanied by significantly increased growth of stable microvessels, upregulation of the proangiogenic VEGFR-2 and its regulator TAL-1, and increase of endogenous endothelial VEGF expression. CONCLUSIONS Our findings in the avian model of SSc suggest that cell-demanded release of VEGF121 from fibrin matrix induces controlled angiogenesis by differential regulation of VEGFR-1 and VEGFR-2 expression, shifting the balance towards the proangiogenic VEGFR-2. The study shows the potential of covalently conjugated VEGF-fibrin matrices for the therapy of ischaemic lesions such as fingertip ulcers.
Collapse
Affiliation(s)
- Shadab Allipour Birgani
- Division of Experimental Pathophysiology and Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Marion Mailänder
- Division of Experimental Pathophysiology and Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Ines Wasle
- Division of Experimental Pathophysiology and Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Hermann Dietrich
- Central Laboratory Animal Facilities, Medical University of Innsbruck, Innsbruck, Austria
| | - Johann Gruber
- Department of Internal Medicine VI, Medical University of Innsbruck, Innsbruck, Austria
| | - Oliver Distler
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Roswitha Sgonc
- Division of Experimental Pathophysiology and Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
25
|
Schreiber A, Eulenberg-Gustavus C, Bergmann A, Jerke U, Kettritz R. Lessons from a double-transgenic neutrophil approach to induce antiproteinase 3 antibody-mediated vasculitis in mice. J Leukoc Biol 2016; 100:1443-1452. [PMID: 27365530 DOI: 10.1189/jlb.5a0116-037r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/25/2016] [Accepted: 06/15/2016] [Indexed: 12/17/2022] Open
Abstract
ANCA to either PR3 or MPO are found in patients with necrotizing vasculitis and glomerulonephritis. ANCA binding to their target antigens on neutrophils and subsequent neutrophil activation are pivotal disease mechanisms that lead to vascular inflammation and necrosis. ANCA interaction with PR3 is more complex than with MPO as the neutrophil-specific CD177 receptor is involved in PR3 surface expression and PR3-ANCA-induced neutrophil activation. Modeling human disease is important to clinical research. Highly successful mouse models of MPO-ANCA vasculitis exist; however, recapitulating PR3-ANCA vasculitis has not been successful. We generated double-transgenic (DT) mice that expressed human PR3 and CD177 under a myeloid-specific huMRP8 promoter in an attempt to model PR3-ANCA vasculitis. DT mice strongly expressed the human transgenes in and on murine neutrophils and bound murine and human anti-PR3 antibodies. Nevertheless, passive transfer of these antibodies into LPS-primed DT mice or immunization of C57BL/6 mice with human PR3 followed by irradiation and transplantation of DT bone marrow failed to induce glomerulonephritis. Further analyses revealed that anti-PR3 antibodies did not activate DT neutrophils as shown by superoxide generation. Moreover, we found that mice did not properly process human pro-PR3 into mature PR3 and, consequently, the signaling complex between PR3, CD177, and CD11b, which promotes neutrophil activation by anti-PR3 antibodies, failed to form. We conclude that important species differences in PR3 and CD177 exist between men and mice that prevented successful generation of a murine anti-PR3 antibody model.
Collapse
Affiliation(s)
- Adrian Schreiber
- Experimental and Clinical Research Center, Charité, Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Nephrology and Intensive Care Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Eulenberg-Gustavus
- Experimental and Clinical Research Center, Charité, Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Astrid Bergmann
- Experimental and Clinical Research Center, Charité, Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Uwe Jerke
- Experimental and Clinical Research Center, Charité, Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Ralph Kettritz
- Experimental and Clinical Research Center, Charité, Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; .,Nephrology and Intensive Care Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
26
|
Kronbichler A, Kerschbaum J, Gründlinger G, Leierer J, Mayer G, Rudnicki M. Evaluation and validation of biomarkers in granulomatosis with polyangiitis and microscopic polyangiitis. Nephrol Dial Transplant 2015; 31:930-6. [PMID: 26410887 DOI: 10.1093/ndt/gfv336] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 08/20/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Studies in anti-neutrophil cytoplasm antibody (ANCA)-associated vasculitis (AAV) have revealed promising biomarkers. The aim of our study was to validate the most encouraging markers of granulomatosis with polyangiitis and microscopic polyangiitis identified by literature search and to create biomarker panels. METHODS A systematic literature review was performed and we identified 161 marker molecules that were ranked by their quantitative differential expression between active and inactive disease. Enzyme-linked immunosorbent assays were used to validate the results in a cross-sectional cohort of patients with renal involvement. Active vasculitis as assessed by the Birmingham Vasculitis Score version 3 (BVAS v3) was defined as BVAS v3 ≥1 and inactive disease as BVAS v3 = 0. Statistical analysis was performed with SPSS version 21 and the Salford Predictive Modeler 7.0 was used to generate a predictive biomarker panel. RESULTS The review indicated abundant expression of sC5bC9, C3a, C5a and monocyte chemotactic protein (MCP)-1 in urine, whereas granulocyte macrophage colony-stimulating factor, C-reactive protein (CRP), soluble fms-like tyrosine kinase-1, interleukin-17A (IL-17A), C5a, hyaluronan, C3a and interleukin-18 binding protein (IL-18BP) were identified to be highly diverse in active and inactive disease in blood samples. Our cross-sectional analysis revealed significant up-regulation of CRP, C5a, C3a, IL-18BP in blood and C5a and MCP-1 in urine samples during active AAV (all P < 0.05). Creation of a biomarker panel comprising CRP and urinary MCP-1 yielded a sensitivity and specificity of 76% (area under the curve 0.89). CONCLUSIONS We identified promising biomarkers in a literature-based review that were in part corroborated as has been shown for CRP, C3a, C5a, IL-18BP in blood and MCP-1 and C5a in urine samples. Moreover, we propose a biomarker panel comprising CRP and urinary MCP-1 in patients with AAV and renal involvement. Further investigations to confirm our preliminary results are clearly warranted, including the reliability to predict disease relapses.
Collapse
Affiliation(s)
- Andreas Kronbichler
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University of Innsbruck, Innsbruck, Austria Vasculitis and Lupus Clinic, Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, UK
| | - Julia Kerschbaum
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University of Innsbruck, Innsbruck, Austria
| | - Georg Gründlinger
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University of Innsbruck, Innsbruck, Austria
| | - Johannes Leierer
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University of Innsbruck, Innsbruck, Austria
| | - Gert Mayer
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University of Innsbruck, Innsbruck, Austria
| | - Michael Rudnicki
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
27
|
Hilhorst M, van Paassen P, Tervaert JWC. Proteinase 3-ANCA Vasculitis versus Myeloperoxidase-ANCA Vasculitis. J Am Soc Nephrol 2015; 26:2314-27. [PMID: 25956510 DOI: 10.1681/asn.2014090903] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In patients with GN or vasculitis, ANCAs are directed against proteinase 3 (PR3) or myeloperoxidase (MPO). The differences between PR3-ANCA-associated vasculitis (AAV) and MPO-AAV described in the past have been supplemented during the last decade. In this review, we discuss the differences between these two small-vessel vasculitides, focusing especially on possible etiologic and pathophysiologic differences. PR3-AAV is more common in northern parts of the world, whereas MPO-AAV is more common in southern regions of Europe, Asia, and the Pacific, with the exception of New Zealand and Australia. A genetic contribution has been extensively studied, and there is a high prevalence of the HLA-DPB1*04:01 allele in patients with PR3-AAV as opposed to patients with MPO-AAV and/or healthy controls. Histologically, MPO-AAV and PR3-AAV are similar but show qualitative differences when analyzed carefully. Clinically, both serotypes are difficult to distinguish, but quantitative differences are present. More organs are affected in PR3-AAV, whereas renal limited vasculitis occurs more often in patients with MPO-AAV. For future clinical trials, we advocate classifying patients by ANCA serotype as opposed to the traditional disease type classification.
Collapse
Affiliation(s)
- Marc Hilhorst
- Clinical and Experimental Immunology, Maastricht University, Maastricht, The Netherlands
| | - Pieter van Paassen
- Clinical and Experimental Immunology, Maastricht University, Maastricht, The Netherlands
| | - Jan Willem Cohen Tervaert
- Clinical and Experimental Immunology, Maastricht University, Maastricht, The Netherlands jw.cohentervaert@maastrichtuniversity
| | | |
Collapse
|
28
|
Bertram A, Lovric S, Engel A, Beese M, Wyss K, Hertel B, Park JK, Becker JU, Kegel J, Haller H, Haubitz M, Kirsch T. Circulating ADAM17 Level Reflects Disease Activity in Proteinase-3 ANCA-Associated Vasculitis. J Am Soc Nephrol 2015; 26:2860-70. [PMID: 25788529 DOI: 10.1681/asn.2014050477] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 01/04/2015] [Indexed: 12/14/2022] Open
Abstract
ANCA-associated vasculitides are characterized by inflammatory destruction of small vessels accompanied by enhanced cleavage of membrane-bound proteins. One of the main proteases responsible for ectodomain shedding is disintegrin and metalloproteinase domain-containing protein 17 (ADAM17). Given its potential role in aggravating vascular dysfunction, we examined the role of ADAM17 in active proteinase-3 (PR3)-positive ANCA-associated vasculitis (AAV). ADAM17 concentration was significantly increased in plasma samples from patients with active PR3-AAV compared with samples from patients in remission or from other controls with renal nonvascular diseases. Comparably, plasma levels of the ADAM17 substrate syndecan-1 were significantly enhanced in active AAV. We also observed that plasma-derived ADAM17 retained its specific proteolytic activity and was partly located on extracellular microparticles. Transcript levels of ADAM17 were increased in blood samples of patients with active AAV, but those of ADAM10 or tissue inhibitor of metalloproteinases 3, which inhibits ADAMs, were not. We also performed a microRNA (miR) screen and identified miR-634 as significantly upregulated in blood samples from patients with active AAV. In vitro, miR-634 mimics induced a proinflammatory phenotype in monocyte-derived macrophages, with enhanced expression and release of ADAM17 and IL-6. These data suggest that ADAM17 has a prominent role in AAV and might account for the vascular complications associated with this disease.
Collapse
Affiliation(s)
- Anna Bertram
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| | - Svjetlana Lovric
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| | - Alissa Engel
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| | - Michaela Beese
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| | - Kristin Wyss
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| | - Barbara Hertel
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| | - Joon-Keun Park
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| | - Jan U Becker
- Institute for Forensic Medicine, Hannover Medical School, Hannover, Germany; and
| | - Johanna Kegel
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| | - Hermann Haller
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| | - Marion Haubitz
- Department of Nephrology and Hypertension, Center for Internal Medicine and Medical Clinic III, Klinikum Fulda, Fulda, Germany
| | - Torsten Kirsch
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| |
Collapse
|
29
|
|
30
|
Lavainne F, Meffray E, Pepper RJ, Néel M, Delcroix C, Salama AD, Fakhouri F. Heparin use during dialysis sessions induces an increase in the antiangiogenic factor soluble Flt1. Nephrol Dial Transplant 2014; 29:1225-31. [DOI: 10.1093/ndt/gft517] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
31
|
Holwerda KM, Burke SD, Faas MM, Zsengeller Z, Stillman IE, Kang PM, van Goor H, McCurley A, Jaffe IZ, Karumanchi SA, Lely AT. Hydrogen sulfide attenuates sFlt1-induced hypertension and renal damage by upregulating vascular endothelial growth factor. J Am Soc Nephrol 2013; 25:717-25. [PMID: 24335973 DOI: 10.1681/asn.2013030291] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Soluble fms-like tyrosine kinase 1 (sFlt1), a circulating antiangiogenic protein, is elevated in kidney diseases and contributes to the development of preeclampsia. Hydrogen sulfide is a vasorelaxant and proangiogenic gas with therapeutic potential in several diseases. Therefore, we evaluated the potential therapeutic effect and mechanisms of action of hydrogen sulfide in an animal model of sFlt1-induced hypertension, proteinuria, and glomerular endotheliosis created by adenovirus-mediated overexpression of sFlt1 in Sprague-Dawley rats. We injected sFlt1-overexpressing animals intraperitoneally with the hydrogen sulfide-donor sodium hydrosulfide (NaHS) (50 µmol/kg, twice daily) or vehicle (n=7 per group). Treatment with NaHS for 8 days significantly reduced sFlt1-induced hypertension, proteinuria, and glomerular endotheliosis. Measurement of plasma protein concentrations with ELISA revealed a reduction of free plasma sFlt1 and an increase of free plasma vascular endothelial growth factor (VEGF) after treatment with NaHS. Renal VEGF-A mRNA expression increased significantly with NaHS treatment. In vitro, NaHS was proangiogenic in an endothelial tube assay and attenuated the antiangiogenic effects of sFlt1. Stimulation of podocytes with NaHS resulted in both short-term VEGF release (120 minutes) and upregulation of VEGF-A mRNA levels (24 hours). Furthermore, pretreatment of mesenteric vessels with a VEGF receptor 2-neutralizing antibody significantly attenuated NaHS-induced vasodilation. These results suggest that hydrogen sulfide ameliorates sFlt1-induced hypertension, proteinuria, and glomerular endotheliosis in rats by increasing VEGF expression. Further studies are warranted to evaluate the role of hydrogen sulfide as a novel therapeutic agent for vascular disorders such as preeclampsia.
Collapse
|
32
|
Increased Soluble Flt-1 Correlates With Delayed Graft Function and Early Loss of Peritubular Capillaries in the Kidney Graft. Transplantation 2013; 96:739-44. [DOI: 10.1097/tp.0b013e31829f4772] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
33
|
Schreiber A, Kettritz R. The neutrophil in antineutrophil cytoplasmic autoantibody-associated vasculitis. J Leukoc Biol 2013; 94:623-31. [DOI: 10.1189/jlb.1012525] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
34
|
|
35
|
Chronic kidney disease may be differentially diagnosed from preeclampsia by serum biomarkers. Kidney Int 2013; 83:177-81. [DOI: 10.1038/ki.2012.348] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
36
|
Yuan J, Gou SJ, Huang J, Hao J, Chen M, Zhao MH. C5a and its receptors in human anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis. Arthritis Res Ther 2012; 14:R140. [PMID: 22691190 PMCID: PMC3446523 DOI: 10.1186/ar3873] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 06/12/2012] [Indexed: 11/27/2022] Open
Abstract
Introduction The complement system is crucial for the development of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). In particular, C5a plays a central role. In this study, plasma and urinary levels of C5a as well as renal C5a receptors (CD88 and C5L2) expression were investigated in patients with AAV. Methods Twenty-four patients with AAV in the active phase, 19 patients with AAV in the remission phase, and 20 patients with lupus nephritis (LN) were included. Plasma and urinary levels of C5a were measured with enzyme-linked immunosorbent assay (ELISA). The staining of CD88 and C5L2 in renal specimens was detected with immunohistochemistry. Results The level of plasma C5a was significantly higher in patients with AAV in the active phase than that in patients in remission, that in patients with LN, and that in normal controls. The urinary C5a level was significantly higher in patients with AAV in the active phase than that in patients in remission and that in normal controls, but not significantly different between patients with active AAV and patients with LN. The mean optical density of CD88 staining in the tubulointerstitium was significantly lower in AAV patients than that in normal controls (0.0052 ± 0.0011 versus 0.029 ± 0.0042; P = 0.005). The mean optical density of C5L2 in glomeruli was significantly higher in AAV patients than that in normal controls (0.013 ± 0.0027 versus 0.0032 ± 0.0006; P < 0.001). The mean optical density of CD88 staining closely correlated with the initial eGFR (r = 0.835; P < 0.001) in AAV patients. Double-labeling immunofluorescence assay suggested that CD88 did not express on neutrophils, monocytes, or macrophages, but C5L2 expressed on neutrophils (or monocytes) and macrophages. Conclusion The elevated plasma and urinary C5a levels indicated complement activation in human AAV. The level of renal CD88 expression could reflect the disease severity of ANCA-associated glomerulonephritis. CD88 expression was downregulated, and C5L2 was upregulated in ANCA-associated glomerulonephritis.
Collapse
Affiliation(s)
- Jun Yuan
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Beijing 100034, China
| | | | | | | | | | | |
Collapse
|
37
|
|