1
|
Ramasamy A, Mohan C. Molecular and Cellular Mediators of Renal Fibrosis in Lupus Nephritis. Int J Mol Sci 2025; 26:2621. [PMID: 40141260 PMCID: PMC11942537 DOI: 10.3390/ijms26062621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/28/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Lupus nephritis (LN), a significant complication of systemic lupus erythematosus (SLE), represents a challenging manifestation of the disease. One of the prominent pathophysiologic mechanisms targeting the renal parenchyma is fibrosis, a terminal process resulting in irreversible tissue damage that eventually leads to a decline in renal function and/or end-stage kidney disease (ESKD). Both glomerulosclerosis and interstitial fibrosis emerge as reliable prognostic indicators of renal outcomes. This article reviews the hallmarks of renal fibrosis in lupus nephritis, including the known and putative drivers of fibrogenesis. A better understanding of the cellular and molecular processes driving fibrosis in LN may help inform the development of therapeutic strategies for this disease, as well as the identification of individuals at higher risk of developing ESKD.
Collapse
Affiliation(s)
| | - Chandra Mohan
- Biomedical Engineering Department, University of Houston, 3517 Cullen Blvd, Room 2027, Houston, TX 77204, USA;
| |
Collapse
|
2
|
Mostmans Y, De Smedt K, Feoli F, Waelput W, De Maertelaer V, Olemans C, Meiers I, Cielen T, Corazza F, Michel O, Richert B. Elevated cutaneous expression of stem cell factor in chronic spontaneous urticaria: a prospective cohort study. Clin Exp Dermatol 2024; 49:1659-1667. [PMID: 38963799 DOI: 10.1093/ced/llae252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Tissue expression of endothelial cell markers of microcirculatory changes in chronic spontaneous urticaria (CSU) is poorly understood. OBJECTIVES To explore the expression of specific endothelial cell markers [stem cell factor (SCF), vascular endothelial growth factor (VEGF) and membrane attack complex (MAC)] in lesional and nonlesional CSU skin through immunohistochemistry (IHC) and in serum. METHODS Lesional and nonlesional skin biopsies from patients with CSU (n = 23) and healthy controls (n = 9) were studied by IHC for expression of SCF, VEGF and MAC. In this population, we also investigated blood levels of VEGF and SCF. Patients were also assessed for clinical characteristics, disease activity and markers of autoimmune CSU. The study was registered at ClinicalTrials.gov (NCT03443362). RESULTS Epidermal SCF reactivity was significantly higher in CSU lesional skin than in healthy skin (P = 0.026). In the dermis, SCF immunoreactivity was seen particularly in endothelial, perivascular and epithelial cells. In CSU lesional skin, the mean perivascular SCF staining was significantly more intense than in healthy controls (P < 0.001). Furthermore, CSU nonlesional skin also showed significantly higher SCF staining in dermal perivascular cells than in healthy controls (P < 0.001). Patients with CSU had the highest SCF immunoreactivity scores in the epidermis and/or on dermal endothelial cells. These patients did not have significantly higher SCF serum levels. CONCLUSIONS To our knowledge, this is the first study to show elevated cutaneous expression of SCF in CSU. These findings underline the potential therapeutic possibilities of anti-Kit antibodies in CSU treatment.
Collapse
Affiliation(s)
- Yora Mostmans
- Department of Immunology-Allergology, CHU Brugmann, Université Libre de Bruxelles, Laken, Belgium
- Department of Dermatology, CHU Brugmann, Université Libre de Bruxelles, Laken, Belgium
| | - Katleen De Smedt
- Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Department of Pathology, Brussels, Belgium
| | - Francesco Feoli
- Department of Dermatopathology, CHU Saint-Pierre, Brussels, Belgium
| | - Wim Waelput
- Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Department of Pathology, Brussels, Belgium
| | - Viviane De Maertelaer
- IRIBHM, Statistical Unit, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Isabelle Meiers
- Department of Dermatopathology, CHU Saint-Pierre, Brussels, Belgium
| | - Thierry Cielen
- Department of Anatomopathology, Institut Jules Bordet, Brussels, Belgium
| | - Francis Corazza
- Department of Immunology, Laboratoire Hospitalier Universitaire de Bruxelles - Universitair Laboratorium Brussels, Brussels, Belgium
| | - Olivier Michel
- Department of Immunology-Allergology, CHU Brugmann, Université Libre de Bruxelles, Laken, Belgium
- Hôpital Delta (CHIREC) and Université Libre de Bruxelles, Brussels, Belgium
| | - Bertrand Richert
- Department of Dermatology, CHU Brugmann, Université Libre de Bruxelles, Laken, Belgium
| |
Collapse
|
3
|
Kissling S, Schwotzer N, Moser M, Froissart M, Fakhouri F. Urinary sC5b-9 is Better Linked to Albuminuria Than to Intrarenal Inflammation in Common Kidney Disease. Kidney Int Rep 2024; 9:3335-3337. [PMID: 39534214 PMCID: PMC11551109 DOI: 10.1016/j.ekir.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/09/2024] [Accepted: 08/05/2024] [Indexed: 11/16/2024] Open
Affiliation(s)
- Sébastien Kissling
- Service of Nephrology and Hypertension, CHUV, Lausanne University, Lausanne, Switzerland
| | - Nora Schwotzer
- Service of Nephrology and Hypertension, CHUV, Lausanne University, Lausanne, Switzerland
| | - Mireille Moser
- Clinical Research Center, University Hospital of Lausanne and University of Lausanne, Switzerland
| | - Marc Froissart
- Research and Education Department, University Hospital of Lausanne and University of Lausanne, Switzerland
| | - Fadi Fakhouri
- Service of Nephrology and Hypertension, CHUV, Lausanne University, Lausanne, Switzerland
| |
Collapse
|
4
|
Meuleman MS, Petitprez F, Pickering MC, Le Quintrec M, Artero MR, Duval A, Rabant M, Gilmore A, Boyer O, Hogan J, Servais A, Provot F, Gnemmi V, Eloudzeri M, Grunenwald A, Buob D, Boffa JJ, Moktefi A, Audard V, Goujon JM, Bridoux F, Thervet E, Karras A, Roumenina LT, Frémeaux Bacchi V, Duong Van Huyen JP, Chauvet S. Complement Terminal Pathway Activation and Intrarenal Immune Response in C3 Glomerulopathy. J Am Soc Nephrol 2024; 35:1034-1044. [PMID: 38709564 PMCID: PMC11377803 DOI: 10.1681/asn.0000000000000373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/29/2024] [Indexed: 05/08/2024] Open
Abstract
Key Points
We evidenced terminal pathway activation (C5b-9 deposits) in most of the glomeruli on kidney biopsy of C3 glomerulopathy.The amount of C5b-9 deposits correlated with disease prognosis in C3 glomerulopathy.Increased terminal pathway activation was found predominantly in a subgroup exhibiting an immuno-fibroblastic signature.
Background
C3 glomerulopathy is a rare disease resulting from an overactivation of the complement alternative pathway. Although there is also evidence of terminal pathway activation, its occurrence and consequences on the disease have been poorly studied.
Methods
We retrospectively studied a cohort of 42 patients diagnosed with C3 glomerulopathy. We performed centralized extensive characterization of histological parameters. Kidney C5b-9 staining was performed as a marker of terminal pathway activation; intrarenal immune response was characterized through transcriptomic analysis.
Results
Eighty-eight percent of biopsies showed C5b-9 deposits in glomeruli. Biopsies were grouped according to the amount of C5b-9 deposits (no or low n=15/42, 36%; intermediate n=15/42, 36%; and high n=12/42, 28%). Patients with high C5b-9 deposits significantly differed from the two other groups of patients and were characterized by a significant higher histological chronicity score (P = 0.005) and lower outcome-free survival (P = 0.001). In multivariable analysis, higher glomerular C5b-9 remained associated with poor kidney prognosis after adjustment. One third of the 847 studied immune genes were upregulated in C3 glomerulopathy biopsies compared with controls. Unsupervised clustering on differentially expressed genes identified a group of kidney biopsies enriched in high glomerular C5b-9 with high immune and fibroblastic signature and showed high chronicity scores on histological examination.
Conclusions
In a cohort of patients with C3 glomerulopathy, intrarenal terminal pathway activation was associated with specific histological phenotype and disease prognosis.
Collapse
Affiliation(s)
- Marie-Sophie Meuleman
- Inflammation, Complement and Cancer Team, Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Paris, France
| | - Florent Petitprez
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Matthew C Pickering
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Imperial College, London, United Kingdom
| | - Moglie Le Quintrec
- Department of Nephrology, Montpellier University Hospital, Montpellier, France
| | - Mikel Rezola Artero
- Inflammation, Complement and Cancer Team, Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Paris, France
| | - Anna Duval
- Inflammation, Complement and Cancer Team, Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Paris, France
- Department of Nephrology, Strasbourg University Hospital, Strasbourg, France
| | - Marion Rabant
- Department of Anathomopathology, Necker Hospital, APHP, Paris, France
- Département Croissance et Signalisation, INSERM U1151, CNRS UMR 8253, Institut Necker-Enfants Malades, Paris, France
| | - Alyssa Gilmore
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Imperial College, London, United Kingdom
| | - Olivia Boyer
- Pediatric Nephrology, Necker-Enfants Malades University Hospital, MARHEA reference center, APHP, Institut Imagine, Université Paris Cité, Paris, France
| | - Julien Hogan
- Department of pediatric Nephrology, Robert Debré Hospital, APHP, Paris, France
| | - Aude Servais
- Department of Nephrology, Necker-Enfants Malades Hospital, APHP, Paris, France
| | - François Provot
- Department of Nephrology, Lille University Hospital, Lille, France
| | - Vivianne Gnemmi
- Department of Pathology, Lille University Hospital, Lille, France
| | - Maeva Eloudzeri
- Département Croissance et Signalisation, INSERM U1151, CNRS UMR 8253, Institut Necker-Enfants Malades, Paris, France
| | - Anne Grunenwald
- Inflammation, Complement and Cancer Team, Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Paris, France
- Department of Nephrology, Poissy Intercommunal Hospital, Poissy, France
| | - David Buob
- Department of Pathology, Tenon Hospital, APHP, Paris, France
| | | | - Anissa Moktefi
- Department of Pathology, Henri Mondor Hospital, APHP, Créteil, France
| | - Vincent Audard
- Assistance Publique des Hôpitaux de Paris (AP-HP), Nephrology and Renal Transplantation Department, Henri Mondor Hospital University, Centre de Référence Maladie Rare Syndrome Néphrotique Idiopathique, Fédération Hospitalo-Universitaire Innovative therapy for immune disorders, Créteil, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U955, Institut Mondor de Recherche Biomédicale (IMRB), Univ Paris Est Créteil, Créteil, France
| | | | - Frank Bridoux
- Department of Nephrology, Poitiers University Hospital, Poitiers, France
| | - Eric Thervet
- Department of Nephrology, European Hospital Georges Pompidou, APHP, Paris, France
- Paris Cité University, Paris, France
| | - Alexandre Karras
- Department of Nephrology, European Hospital Georges Pompidou, APHP, Paris, France
- Paris Cité University, Paris, France
| | - Lubka T Roumenina
- Inflammation, Complement and Cancer Team, Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Paris, France
| | - Véronique Frémeaux Bacchi
- Inflammation, Complement and Cancer Team, Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Paris, France
- Department of Immunology, European Hospital Georges Pompidou, APHP, Paris, France
| | - Jean-Paul Duong Van Huyen
- Inflammation, Complement and Cancer Team, Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Paris, France
- Department of Anathomopathology, Necker Hospital, APHP, Paris, France
- Paris Cité University, Paris, France
| | - Sophie Chauvet
- Inflammation, Complement and Cancer Team, Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Paris, France
- Department of Nephrology, European Hospital Georges Pompidou, APHP, Paris, France
- Paris Cité University, Paris, France
| |
Collapse
|
5
|
Alkaff FF, Lammerts RGM, Daha MR, Berger SP, van den Born J. Apical tubular complement activation and the loss of kidney function in proteinuric kidney diseases. Clin Kidney J 2024; 17:sfae215. [PMID: 39135935 PMCID: PMC11318052 DOI: 10.1093/ckj/sfae215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Indexed: 08/15/2024] Open
Abstract
Many kidney diseases are associated with proteinuria. Since proteinuria is independently associated with kidney function loss, anti-proteinuric medication, often in combination with dietary salt restriction, comprises a major cornerstone in the prevention of progressive kidney failure. Nevertheless, complete remission of proteinuria is very difficult to achieve, and most patients with persistent proteinuria slowly progress toward kidney failure. It is well-recognized that proteinuria leads to kidney inflammation and fibrosis via various mechanisms. Among others, complement activation at the apical side of the proximal tubular epithelial cells is suggested to play a crucial role as a cause of progressive loss of kidney function. However, hitherto limited attention is given to the pathophysiological role of tubular complement activation relative to glomerular complement activation. This review aims to summarize the evidence for tubular epithelial complement activation in proteinuric kidney diseases in relation to loss of kidney function.
Collapse
Affiliation(s)
- Firas F Alkaff
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Division of Pharmacology and Therapy, Department of Anatomy, Histology, and Pharmacology, Faculty of Medicine Universitas Airlangga, Surabaya, Indonesia
| | - Rosa G M Lammerts
- Transplantation Immunology, Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mohamed R Daha
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Stefan P Berger
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jacob van den Born
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
6
|
Reddy H, Kashiv P, Kumar S, Sejpal KN, Acharya S. Overlap of Scleroderma and Class V Lupus Nephritis: A Rare Autoimmune Confluence. Cureus 2024; 16:e56990. [PMID: 38681456 PMCID: PMC11046166 DOI: 10.7759/cureus.56990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 03/26/2024] [Indexed: 05/01/2024] Open
Abstract
The autoimmune connective tissue disease scleroderma is characterized by fibrosis of the skin, blood vessels, and visceral organs. Overlap syndromes are conditions in which a patient has characteristics from two or more autoimmune disorders. The coexistence of scleroderma and lupus nephritis is rare but documented. However, it is crucial to note that both scleroderma and lupus are complex autoimmune diseases with diverse clinical presentations and can affect multiple organ systems, including the kidneys. This case report presents a unique clinical scenario of a patient with the coexistence of scleroderma and systemic lupus erythematosus with Class V lupus nephritis, highlighting the challenges in diagnosis, treatment, and the need for a multidisciplinary approach.
Collapse
Affiliation(s)
- Harshitha Reddy
- Internal Medicine, Jawaharlal Nehru Medical College, Wardha, IND
| | - Pranjal Kashiv
- Nephrology, Jawaharlal Nehru Medical College, Wardha, IND
| | - Sunil Kumar
- Internal Medicine, Jawaharlal Nehru Medical College, Wardha, IND
| | - Kapil N Sejpal
- Nephrology, Jawaharlal Nehru Medical College, Wardha, IND
| | - Sourya Acharya
- Internal Medicine, Jawaharlal Nehru Medical College, Wardha, IND
| |
Collapse
|
7
|
Lyu X, Li M, Zhang PL, Wei W, Werth VP, Liu ML. Neutrophil extracellular traps drive lupus flares with acute skin and kidney inflammation triggered by ultraviolet irradiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.23.572573. [PMID: 38187639 PMCID: PMC10769371 DOI: 10.1101/2023.12.23.572573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Sunlight triggers lupus flares causing both local skin and systemic inflammation, including lupus nephritis, through poorly understood mechanisms. To address this knowledge gap, we found that UVB irradiation of asymptomatic, young female lupus-prone mice induced skin and kidney inflammation with proteinuria, accompanied by neutrophil infiltration and neutrophil extracellular trap (NET) formation. Furthermore, UVB irradiation induced co-expression of CXCR4 and cytokines/C3 by neutrophils in vitro and in vivo, in the skin and kidneys of lupus-prone mice, indicating their transmigratory and pro-inflammatory potentials. A causality study demonstrated that inhibiting CXCR4 attenuated renal neutrophil infiltration, accumulation of NETs, NET-associated cytokines/C3, and proteinuria in UVB-irradiated lupus-prone mice. Remarkably, inhibiting NETosis through a novel strategy targeting nuclear envelope integrity reduced deposition of NET-associated cytokines/C3 in skin and kidneys, attenuating proteinuria in UVB-irradiated MRL/lpr·lmnB1 Tg mice. Our investigation unveils a new mechanism by which neutrophil NETs drive the early onset of lupus flares triggered by UVB-irradiation. Targeting neutrophil transmigration and NETosis could be promising therapeutic strategies.
Collapse
|
8
|
Anderson M, Magro C, Belmont HM. Microvascular C5b-9 deposition in non-lesional skin in patients with SLE and its correlation with active lupus nephritis: a prospective observational study. Lupus Sci Med 2023; 10:e000996. [PMID: 37879755 PMCID: PMC10603335 DOI: 10.1136/lupus-2023-000996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/14/2023] [Indexed: 10/27/2023]
Abstract
OBJECTIVE Tissue damage in lupus nephritis (LN) is mediated by activation of the classical complement pathway. Complement-mediated upregulation of endothelial cell adhesion molecules is seen in dermal blood vessels of non-lesional skin of patients with active lupus. In diseases with systemic complement activation, extensive microvascular C5b-9 deposition is seen in non-lesional skin. In this study, we assess the presence of systemic complement pathway activation as determined by non-lesional skin microvascular C5b-9 deposition in patients with LN. METHODS Eight patients with active LN and eight patients without active LN underwent non-lesional skin biopsies. Using a diaminobenzidine technique, specimens were evaluated for microvascular C5b-9 consistent with systemic complement pathway activation. RESULTS Five of eight patients with active LN and one of eight patients without active LN demonstrated positive C5b-9 staining in non-lesional skin (p=0.04). Positive non-lesional C5b-9 staining has greater specificity, 87.5%, for active LN than pyuria, low complements, elevated double-stranded DNA (dsDNA) and proteinuria. Urine protein creatinine ratio was significantly higher in patients with positive non-lesional C5b-9 deposition (5.18 vs 1.20; p=0.04). C5b-9 deposition was not associated with a higher NIH Activity Index, interstitial fibrosis, dsDNA or lower complements. CONCLUSION This is the first study to demonstrate evidence in non-lesional skin of microvascular C5b-9 indicative of systemic complement pathway activation in LN. C5b-9 deposition is statistically more common and demonstrated greater specificity than most historical biomarkers for active LN. The findings support a potential role for microvascular C5b-9 assessment in non-lesional skin as a biomarker for LN activity.
Collapse
Affiliation(s)
- Meghan Anderson
- Department of Rheumatology, NYU Grossman School of Medicine, New York City, New York, USA
| | - Cynthia Magro
- Department of Pathology, Weill Cornell Medicine, New York City, New York, USA
| | - H Michael Belmont
- Department of Rheumatology, NYU Grossman School of Medicine, New York City, New York, USA
| |
Collapse
|
9
|
Cheung CK, Dormer JP, Barratt J. The role of complement in glomerulonephritis-are novel therapies ready for prime time? Nephrol Dial Transplant 2023; 38:1789-1797. [PMID: 36307926 DOI: 10.1093/ndt/gfac296] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Indexed: 08/01/2023] Open
Abstract
The complement system plays a key pathogenic role in glomerular diseases with a diverse range of aetiologies, including C3 glomerulopathy, immunoglobulin A nephropathy, membranous nephropathy, ANCA-associated vasculitis and lupus nephritis. Several novel therapies targeting complement activity have recently been developed, which have now been approved or are in the late stages of clinical development. In this review, potential benefits and challenges of targeting the complement system in glomerular disease are discussed. We summarize current understanding of the role of complement, and the novel targeted therapies that are being developed for the treatment of glomerular disease.
Collapse
Affiliation(s)
- Chee Kay Cheung
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- John Walls Renal Unit, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - John P Dormer
- Department of Histopathology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Jonathan Barratt
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- John Walls Renal Unit, University Hospitals of Leicester NHS Trust, Leicester, UK
| |
Collapse
|
10
|
Tomaszewski K, Herlitz L. Complement detection in kidney biopsies - utility and challenges. Curr Opin Nephrol Hypertens 2023; 32:241-248. [PMID: 36811623 DOI: 10.1097/mnh.0000000000000872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
PURPOSE OF REVIEW This review discusses the important role of staining for components of the complement cascade in both native and transplant kidney biopsies. The use of complement staining as a marker of prognosis, disease activity, and as a potential future tool in identifying patients who may benefit from complement-targeted therapies is discussed. RECENT FINDINGS While staining for C3, C1q and C4d can yield valuable information about complement activation in kidney biopsies, to adequately assess complement activation and potential therapeutic targets, expanded staining panels looking at multiple split products and complement regulatory proteins are needed. Recent progress has been made in identifying markers of disease severity in C3 glomerulonephritis and IgA nephropathy, such as Factor H-related Protein-5, which may serve as future tissue biomarkers. In the transplant setting, the limitation of relying on C4d staining to identify antibody mediated rejection is giving way to molecular diagnostics, including The Banff Human Organ Transplant (B-HOT) panel, which includes numerous complement complement-related transcripts, with the classical, lectin, alternative, and common pathways. SUMMARY Staining for complement components in kidney biopsies to understand how complement is activated in individual cases may help to identify patients who may benefit from complement-targeted therapies.
Collapse
|
11
|
Abstract
Cerebrovascular accidents (CVAs) or strokes are part of the common thrombotic manifestations of Systemic Lupus Erythematosus (SLEs) and Antiphospholipid syndrome (APS). Such neurological thrombotic events tend to occur in patients with SLE at a higher frequency when Antiphospholipid antibodies (aPLs) are present, and tend to involve the large cerebral vessels. The mechanism of stroke in SLE can be driven by complement deposition and neuroinflammation involving the blood-brain barrier although the traditional cardiovascular risk factors remain major contributing factors. Primary prevention with antiplatelet therapy and disease activity controlling agent is the basis of the management. Anticoagulation via warfarin had been a tool for secondary prevention, especially in stroke recurrence, although the debate continues regarding the target international normalized ratio (INR). The presence of either of the three criteria antiphospholipid antibodies (aPLs) and certain non-criteria aPL can be an independent risk factor for stroke. The exact mechanism for the involvement of the large cerebral arteries, especially in lupus anticoagulant (LAC) positive cases, is still to be deciphered. The data on the role of non-criteria aPL remain very limited and heterogenous, but IgA antibodies against β2GPI and the D4/5 subunit as well as aPS/PT IgG might have a contribution. Anticoagulation with warfarin has been recommended although the optimal dosing or the utility of combination with antiplatelet agents is still unknown. Minimal data is available for direct oral anticoagulants (DOACs).
Collapse
Affiliation(s)
- Georges El Hasbani
- Department of Internal Medicine, St Vincent's Medical Center, Bridgeport, CT, USA
| | - Imad Uthman
- Department of Internal Medicine, 11238American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
12
|
Wijaya C, Burns C, Hall S, Farmer M, Jones D, Rowlandson M, Choi P, Formby M, de Malmanche T. Measurement of Complement Activation via Plasma-Soluble C5b-9 Comparison with Terminal Complement Complex Staining in a Series of Kidney Biopsies. Kidney Blood Press Res 2023; 48:220-230. [PMID: 36917968 PMCID: PMC10124756 DOI: 10.1159/000529734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 02/09/2023] [Indexed: 03/16/2023] Open
Abstract
INTRODUCTION With the emergence of therapeutic complement inhibitors, there is a need to identify patients with complement-driven inflammation. C5b-9 is the terminal product of the three complement pathways and therefore a marker of total complement activation. We present a pilot study which aims to assess whether plasma soluble C5b-9 (sC5b-9) correlates with terminal complement complex (TCC) staining in kidney tissue. The secondary aim was to assess the utility of plasma sC5b-9 as part of routine workup in kidney patients undergoing kidney biopsy. METHODS Thirty-seven patients undergoing kidney biopsy had plasma sC5b-9 and TCC staining on kidney tissue performed. Additional blood markers including creatinine, haemoglobin, CRP, factor H, factor I, and midkine levels were also taken. These parameters were correlated with the histological diagnoses. Patients were divided into a diseased group (n = 31) and a control group (n = 6) consisting of transplanted kidneys with minor or no changes. Of the biopsies in the control group, 50% were performed as per protocol, and the other 50% were performed due to clinical need. RESULTS There was no correlation found between plasma sC5b-9 and TCC kidney staining. Elevated sC5b-9 levels were found in a heterogeneous group of patients but were associated with higher CRP and lower haemoglobin levels. Overall, there was more TCC kidney staining in the diseased group compared with the control group, and a trend was observed of diabetic, primary membranous nephropathy, and amyloidosis patients having more intense glomerular and peritubular/interstitial staining. CONCLUSION Plasma sC5b-9 as a marker of total complement activation does not correlate with TCC kidney staining. This discordance suggests that plasma sC5b-9 and TCC staining are distinct markers of disease. TCC staining reflects chronicity and tissue deposition of complement over time. Conversely, plasma sC5b-9 concentrations change rapidly and reflect systemic complement activation. Complement activation was present in a heterogeneous group of kidney disease, indicating the underlying role of complement in many disorders.
Collapse
Affiliation(s)
- Carolyn Wijaya
- Immunopathology, NSW Health Pathology, John Hunter Hospital, New Lambton Heights, NSW, Australia
- University of Newcastle, University Drive, Callaghan, NSW, Australia
| | - Christine Burns
- Immunopathology, NSW Health Pathology, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Sharron Hall
- Immunopathology, NSW Health Pathology, John Hunter Hospital, New Lambton Heights, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Melissa Farmer
- Anatomical Pathology, NSW Health Pathology, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Denise Jones
- Renal Department, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Matthew Rowlandson
- Renal Department, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Peter Choi
- Renal Department, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Mark Formby
- Anatomical Pathology, NSW Health Pathology, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Theo de Malmanche
- Immunopathology, NSW Health Pathology, John Hunter Hospital, New Lambton Heights, NSW, Australia
| |
Collapse
|
13
|
Labib HS, Salman MI, Halim MI, Fawaz SI. Apoptosis in lupus nephritis patients: a study of Bcl-2 to assess glomerular and tubular damage. EGYPTIAN RHEUMATOLOGY AND REHABILITATION 2023. [DOI: 10.1186/s43166-023-00186-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2023] Open
Abstract
Abstract
Background
Systemic lupus erythematosus (SLE) is an immune-mediated disease, due to exposure of self-antigens, through impairment of apoptosis and failure of lymphocytic tolerance. Impaired regulation of the pro- and anti-apoptotic gene products which coordinate programmed cell death may result in autoreactive B and T cells and autoimmunity. Genetically engineered mice that over-express the anti-apoptotic molecule Bcl-2, B cell lymphoma 2 (Bcl2) in B-lymphocytes advance a lupus-like illness. Lupus nephritis (LN) is one of the most serious manifestations of this autoimmune disorder. Glomerulonephritis (GN) is caused by either impaired regulation of apoptosis and/or clearance of apoptotic cells leading to a T cell-mediated autoimmune reaction with initiation of pathological immune complex deposits.
Objective
To evaluate the correlation between Bcl2 glomerular and tubular expression and pathological findings and laboratory data in different types of SLE GN.
Results
Compared to the control group, patients with lupus nephritis have significantly higher glomerular, interstitial and tubular expression level (P value < 0.001). BCL2 expression was positively correlated with serum anti-ds-DNA, urine 24-h protein and with the chronicity index. All LN patients had significant glomerular, interstitial and tubular deposits of BCL2, P value < 0.001, P value 0.004, and P value 0.03, respectively.
Conclusion
The intrinsic pathway of apoptosis interferes not only with the pathogenesis of lupus glomerulonephritis but also interferes with the pathogenesis of tubulointerstitial lupus nephritis. tubulointerstitial lesions may not only be a result of glomerular injury but also a significant factor in lupus nephritis.
Collapse
|
14
|
Zarantonello A, Revel M, Grunenwald A, Roumenina LT. C3-dependent effector functions of complement. Immunol Rev 2023; 313:120-138. [PMID: 36271889 PMCID: PMC10092904 DOI: 10.1111/imr.13147] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
C3 is the central effector molecule of the complement system, mediating its multiple functions through different binding sites and their corresponding receptors. We will introduce the C3 forms (native C3, C3 [H2 O], and intracellular C3), the C3 fragments C3a, C3b, iC3b, and C3dg/C3d, and the C3 expression sites. To highlight the important role that C3 plays in human biological processes, we will give an overview of the diseases linked to C3 deficiency and to uncontrolled C3 activation. Next, we will present a structural description of C3 activation and of the C3 fragments generated by complement regulation. We will proceed by describing the C3a interaction with the anaphylatoxin receptor, followed by the interactions of opsonins (C3b, iC3b, and C3dg/C3d) with complement receptors, divided into two groups: receptors bearing complement regulatory functions and the effector receptors without complement regulatory activity. We outline the molecular architecture of the receptors, their binding sites on the C3 activation fragments, the cells expressing them, the diversity of their functions, and recent advances. With this review, we aim to give an up-to-date analysis of the processes triggered by C3 activation fragments on different cell types in health and disease contexts.
Collapse
Affiliation(s)
- Alessandra Zarantonello
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Margot Revel
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Anne Grunenwald
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Lubka T Roumenina
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| |
Collapse
|
15
|
Obrișcă B, Vornicu A, Procop A, Herlea V, Terinte-Balcan G, Gherghiceanu M, Ismail G. A Histology-Guided Approach to the Management of Patients with Lupus Nephritis: Are We There Yet? Biomedicines 2022; 10:biomedicines10061409. [PMID: 35740431 PMCID: PMC9220241 DOI: 10.3390/biomedicines10061409] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/06/2022] [Accepted: 06/13/2022] [Indexed: 11/16/2022] Open
Abstract
Renal involvement is a frequent complication of systemic lupus erythematosus (SLE). It occurs in up to two-thirds of patients, often early during the disease course, and is the most important predictor of the morbidity and mortality of SLE patients. Despite tremendous improvements in the approach of the lupus nephritis (LN) therapy, including the recent approval of two new disease-modifying therapies, up to 50% of patients do not obtain a renal response and up to 25% will eventually progress to end-stage renal disease (ESRD) within 10 years of diagnosis. Given the lack of correlation between clinical features and histological lesions, there is an increasing need for a histology-guided approach to the management of patients with LN. Apart from the initial diagnosis of type and severity of renal injury in SLE, the concept of a repeat kidney biopsy (either in a for-cause or a per-protocol scenario) has begun to gain increasing popularity in the nephrology community. Herein, we will provide a comprehensive overview of the most important areas of utility of the kidney biopsy in patients with LN.
Collapse
Affiliation(s)
- Bogdan Obrișcă
- Department of Nephrology, Fundeni Clinical Institute, 022328 Bucharest, Romania; (B.O.); (A.V.)
- Department of Nephrology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Alexandra Vornicu
- Department of Nephrology, Fundeni Clinical Institute, 022328 Bucharest, Romania; (B.O.); (A.V.)
- Department of Nephrology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Alexandru Procop
- Department of Pathology, Fundeni Clinical Institute, 022328 Bucharest, Romania; (A.P.); (V.H.)
| | - Vlad Herlea
- Department of Pathology, Fundeni Clinical Institute, 022328 Bucharest, Romania; (A.P.); (V.H.)
| | - George Terinte-Balcan
- Ultrastructural Pathology, “Victor Babes” National Institute of Pathology, 050097 Bucharest, Romania; (G.T.-B.); (M.G.)
| | - Mihaela Gherghiceanu
- Ultrastructural Pathology, “Victor Babes” National Institute of Pathology, 050097 Bucharest, Romania; (G.T.-B.); (M.G.)
| | - Gener Ismail
- Department of Nephrology, Fundeni Clinical Institute, 022328 Bucharest, Romania; (B.O.); (A.V.)
- Department of Nephrology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Correspondence:
| |
Collapse
|
16
|
Fakhouri F, Schwotzer N, Golshayan D, Frémeaux-Bacchi V. The Rational Use of Complement Inhibitors in Kidney Diseases. Kidney Int Rep 2022; 7:1165-1178. [PMID: 35685323 PMCID: PMC9171628 DOI: 10.1016/j.ekir.2022.02.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/15/2022] [Accepted: 02/21/2022] [Indexed: 12/14/2022] Open
Abstract
The development of complement inhibitors represented one of the major breakthroughs in clinical nephrology in the last decade. Complement inhibition has dramatically transformed the outcome of one of the most severe kidney diseases, the atypical hemolytic uremic syndrome (aHUS), a prototypic complement-mediated disorder. The availability of complement inhibitors has also opened new promising perspectives for the management of several other kidney diseases in which complement activation is involved to a variable extent. With the rapidly growing number of complement inhibitors tested in a rapidly increasing number of indications, a rational use of this innovative and expensive new therapeutic class has become crucial. The present review aims to summarize what we know, and what we still ignore, regarding complement activation and therapeutic inhibition in kidney diseases. It also provides some clues and elements of thoughts for a rational approach of complement modulation in kidney diseases.
Collapse
Affiliation(s)
- Fadi Fakhouri
- Service de Néphrologie et d'hypertension, Département de Médecine, Centre Hospitalier Universitaire Vaudois (CHUV), Université de Lausanne, Lausanne, Switzerland
| | - Nora Schwotzer
- Service de Néphrologie et d'hypertension, Département de Médecine, Centre Hospitalier Universitaire Vaudois (CHUV), Université de Lausanne, Lausanne, Switzerland
| | - Déla Golshayan
- Centre de Transplantation d'organes, Département de Médecine, Centre Hospitalier Universitaire Vaudois (CHUV), Université de Lausanne, Lausanne, Switzerland
| | - Véronique Frémeaux-Bacchi
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service d'Immunologie, Paris University, Paris, France
| |
Collapse
|
17
|
Urinary C5b-9 as a Prognostic Marker in IgA Nephropathy. J Clin Med 2022; 11:jcm11030820. [PMID: 35160271 PMCID: PMC8836759 DOI: 10.3390/jcm11030820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/24/2022] [Accepted: 02/02/2022] [Indexed: 01/27/2023] Open
Abstract
C5b-9 plays an important role in the pathogenesis of immunoglobin A nephropathy (IgAN). We evaluated C5b-9 as a prognostic marker for IgAN. We prospectively enrolled 33 patients with biopsy-proven IgAN. We analyzed the correlation between baseline urinary C5b-9 levels, posttreatment changes in their levels, and clinical outcomes, including changes in proteinuria, estimated glomerular filtration rate (eGFR), and treatment response. Baseline urinary C5b-9 levels were positively correlated with proteinuria (r = 0.548, p = 0.001) at the time of diagnosis. Changes in urinary C5b-9 levels were positively correlated with changes in proteinuria (r = 0.644, p < 0.001) and inversely correlated with changes in eGFR (r = −0.410, p = 0.018) at 6 months after treatment. Changes in urinary C5b-9 levels were positively correlated with time-averaged proteinuria during the follow-up period (r= 0.461, p = 0.007) but were not correlated with the mean annual rate of eGFR decline (r = −0.282, p = 0.112). Baseline urinary C5b-9 levels were not a significant independent factor that could predict the treatment response in logistic regression analyses (odds ratio 0.997; 95% confidence interval, 0.993 to 1.000; p = 0.078). Currently, urinary C5b-9 is not a promising prognostic biomarker for IgAN, and further studies are needed.
Collapse
|
18
|
Wang S, Wu M, Chiriboga L, Zeck B, Goilav B, Wang S, Jimenez AL, Putterman C, Schwartz D, Pullman J, Broder A, Belmont HM. Membrane attack complex (MAC) deposition in renal tubules is associated with interstitial fibrosis and tubular atrophy: a pilot study. Lupus Sci Med 2022; 9:9/1/e000576. [PMID: 34996855 PMCID: PMC8744090 DOI: 10.1136/lupus-2021-000576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/23/2021] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Treatment failures for lupus nephritis (LN) are high with 10%-30% of patients progressing to end-stage renal disease (ESRD) within 10 years. Interstitial fibrosis/tubular atrophy (IFTA) is a predictor of progression to ESRD. Prior studies suggest that tubulointerstitial injury secondary to proteinuria in LN is mediated by complement activation in the tubules, specifically through the membrane attack complex (MAC). This study aimed to investigate the associations between tubular MAC deposition with IFTA and proteinuria. METHODS In this cross-sectional study, LN kidney biopsies were assessed for MAC deposition by staining for Complement C9, a component of the MAC. Chromogenic immunohistochemistry was performed on paraffin-embedded human renal biopsy sections using unconjugated, murine anti-human Complement C9 (Hycult Biotech, clone X197). Tubular C9 staining intensity was analysed as present versus absent. IFTA was defined as minimal (<10%), mild (10%-24%), moderate (25%-50%) and severe (>50%). RESULTS Renal biopsies from 30 patients with LN were studied. There were 24 (80%) female sex, mean age (SD) was 33 (12) years old and 23 (77%) had pure/mixed proliferative LN. Tubular C9 staining was present in 7 (23%) biopsies. 27 patients had minimal-to-mild IFTA and 3 patients had moderate IFTA. Among the C9 + patients, 3 (43%) had moderate IFTA as compared with none in the C9- group, p=0.009. C9 + patients had higher median (IQR) proteinuria as compared with C9- patients: 6.2 g (3.3-13.1) vs 2.4 g (1.3-4.6), p=0.001 at the time of biopsy. There was no difference in estimated glomerular filtration rate (eGFR) between the C9 + and C9- groups. CONCLUSION This study demonstrated that tubular MAC deposition is associated with higher degree of IFTA and proteinuria, which are predictors of progression to ESRD. These results suggest that tubular MAC deposition may be useful in classification of LN. Understanding the role of complement in tubulointerstitial injury will also identify new avenues for LN treatment.
Collapse
Affiliation(s)
- Shudan Wang
- Rheumatology, Montefiore Medical Center, Bronx, New York, USA .,Rheumatology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Ming Wu
- Pathology, NYU Langone Health, New York, New York, USA
| | | | - Briana Zeck
- Pathology, NYU Langone Health, New York, New York, USA
| | - Beatrice Goilav
- Nephrology, The Children's Hospital at Montefiore, Bronx, New York, USA
| | - Shuwei Wang
- Rheumatology, Morristown Medical Center, Morristown, New Jersey, USA
| | - Alejandra Londono Jimenez
- Rheumatology, Montefiore Medical Center, Bronx, New York, USA.,Rheumatology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Chaim Putterman
- Rheumatology, Albert Einstein College of Medicine, Bronx, New York, USA.,Bar-Ilan University, Ramat Gan, Tel Aviv, Israel
| | | | - James Pullman
- Pathology, Montefiore Medical Center, Bronx, New York, USA
| | - Anna Broder
- Rheumatology, Montefiore Medical Center, Bronx, New York, USA.,Rheumatology, Albert Einstein College of Medicine, Bronx, New York, USA
| | | |
Collapse
|
19
|
Nithagon P, Cortazar F, Shah SI, Weins A, Laliberte K, Jeyabalan A, Niles J, Zonozi R. Eculizumab and Complement Activation in Anti-glomerular Basement Membrane Disease. Kidney Int Rep 2021; 6:2713-2717. [PMID: 34622110 PMCID: PMC8484114 DOI: 10.1016/j.ekir.2021.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/30/2021] [Accepted: 07/03/2021] [Indexed: 11/25/2022] Open
Affiliation(s)
- Pravarut Nithagon
- Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Frank Cortazar
- New York Nephrology Vasculitis and Glomerular Center, Albany, New York, USA
| | - Sujal I Shah
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Astrid Weins
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Karen Laliberte
- Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Anushya Jeyabalan
- Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - John Niles
- Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Reza Zonozi
- Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Trachtman H, Laskowski J, Lee C, Renner B, Feemster A, Parikh S, Panzer SE, Zhong W, Cravedi P, Cantarelli C, Kulik L, You Z, Satchell S, Rovin B, Liu F, Kalled SL, Holers VM, Jalal D, Thurman JM. Natural antibody and complement activation characterize patients with idiopathic nephrotic syndrome. Am J Physiol Renal Physiol 2021; 321:F505-F516. [PMID: 34459222 DOI: 10.1152/ajprenal.00041.2021] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) and minimal change disease (MCD) are common forms of idiopathic nephrotic syndrome. The causes of these diseases are incompletely understood, but the response of patients to immunosuppressive therapies suggests that their pathogenesis is at least in part immune mediated. Preclinical and clinical research indicates that activation of the classical pathway of complement contributes to glomerular injury in FSGS. Glomerular IgM deposits are also prominent in some patients, raising the possibility that IgM is a trigger of classical pathway activation. In the present study, we examined the pattern of complement activation in the glomeruli and plasma of patients with nephrotic syndrome. We also tested whether patients with FSGS and MCD have elevated levels of natural IgM reactive with epitopes on glomerular endothelial cells and cardiolipin. We found evidence of classical pathway activation in patients with idiopathic nephrotic syndrome compared with healthy control subjects. We also detected higher levels of self-reactive IgM to both targets. Based on these results, IgM and classical pathway activation may contribute to disease pathogenesis in some patients with FSGS and MCD.NEW & NOTEWORTHY IgM is detected in biopsies from some patients with nephrotic syndrome, although this has been attributed to passive trapping of the protein. We found, however, that IgM colocalizes with complement activation fragments in some glomeruli. We also found that affected patients had higher levels of IgM reactive to glomerular endothelial cell epitopes. Thus, IgM activates the complement system in the glomeruli of some patients with nephrotic syndrome and may contribute to injury.
Collapse
Affiliation(s)
- Howard Trachtman
- Department of Pediatrics, Langone Medical Center, New York University School of Medicine, New York, New York
| | - Jennifer Laskowski
- Department of Medicine, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, Colorado
| | - Cameron Lee
- Department of Medicine, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, Colorado
| | - Brandon Renner
- Department of Medicine, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, Colorado
| | - Andrew Feemster
- Department of Pediatrics, Langone Medical Center, New York University School of Medicine, New York, New York
| | - Samir Parikh
- Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Sarah E Panzer
- Department of Medicine, University of Wisconsin-Madison Hospital and Clinics, Madison, Wisconsin
| | - Weixiong Zhong
- Department of Medicine, University of Wisconsin-Madison Hospital and Clinics, Madison, Wisconsin
| | - Paolo Cravedi
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Chiara Cantarelli
- Dipartimento di Medicina e Chirurgia, Università di Parma, UO Nefrologia, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Liudmila Kulik
- Department of Medicine, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, Colorado
| | - Zhiying You
- Department of Medicine, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, Colorado
| | - Simon Satchell
- Bristol Renal, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Brad Rovin
- Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Fei Liu
- Q32 Bio, Incorporated, Cambridge, Massachusetts
| | | | - V Michael Holers
- Department of Medicine, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, Colorado
| | - Diana Jalal
- Department of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Joshua M Thurman
- Department of Medicine, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
21
|
Bomback AS, Appel GB, Gipson DS, Hladunewich MA, Lafayette R, Nester CM, Parikh SV, Smith RJH, Trachtman H, Heeger PS, Ram S, Rovin BH, Ali S, Arceneaux N, Ashoor I, Bailey-Wickins L, Barratt J, Beck L, Cattran DC, Cravedi P, Erkan E, Fervenza F, Frazer-Abel AA, Fremeaux-Bacchi V, Fuller L, Gbadegesin R, Hogan JJ, Kiryluk K, le Quintrec-Donnette M, Licht C, Mahan JD, Pickering MC, Quigg R, Rheault M, Ronco P, Sarwal MM, Sethna C, Spino C, Stegall M, Vivarelli M, Feldman DL, Thurman JM. Improving Clinical Trials for Anticomplement Therapies in Complement-Mediated Glomerulopathies: Report of a Scientific Workshop Sponsored by the National Kidney Foundation. Am J Kidney Dis 2021; 79:570-581. [PMID: 34571062 DOI: 10.1053/j.ajkd.2021.07.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/13/2021] [Indexed: 12/25/2022]
Abstract
Blocking the complement system as a therapeutic strategy has been proposed for numerous glomerular diseases but presents myriad questions and challenges, not the least of which is demonstrating efficacy and safety. In light of these potential issues and because there are an increasing number of anticomplement therapy trials either planned or under way, the National Kidney Foundation facilitated an all-virtual scientific workshop entitled "Improving Clinical Trials for Anti-Complement Therapies in Complement-Mediated Glomerulopathies." Attended by patient representatives and experts in glomerular diseases, complement physiology, and clinical trial design, the aim of this workshop was to develop standards applicable for designing and conducting clinical trials for anticomplement therapies across a wide spectrum of complement-mediated glomerulopathies. Discussions focused on study design, participant risk assessment and mitigation, laboratory measurements and biomarkers to support these studies, and identification of optimal outcome measures to detect benefit, specifically for trials in complement-mediated diseases. This report summarizes the discussions from this workshop and outlines consensus recommendations.
Collapse
Affiliation(s)
- Andrew S Bomback
- Division of Nephrology, Columbia University Irving Medical Center, New York.
| | - Gerald B Appel
- Division of Nephrology, New York University Langone Health, New York
| | - Debbie S Gipson
- Department of Pediatrics, Division of Nephrology, University of Michigan, Ann Arbor, Michigan
| | | | | | - Carla M Nester
- Division of Nephrology, University of Iowa, Iowa City, Iowa
| | - Samir V Parikh
- Division of Nephrology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Richard J H Smith
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Howard Trachtman
- Division of Nephrology, New York University Langone Health, New York
| | - Peter S Heeger
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York
| | - Sanjay Ram
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Brad H Rovin
- Division of Nephrology, The Ohio State University College of Medicine, Columbus, Ohio
| | | | | | - Isa Ashoor
- Division of Nephrology, Louisiana State University Health, New Orleans, Louisiana
| | | | | | - Laurence Beck
- Division of Nephrology, Boston University School of Medicine, Boston, Massachusetts
| | - Daniel C Cattran
- Division of Nephrology, University of Toronto, Toronto, ON, Canada
| | - Paolo Cravedi
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York
| | - Elif Erkan
- Division of Nephrology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | | | - Ashley A Frazer-Abel
- Division of Nephrology, University of Colorado School of Medicine, Aurora, Colorado
| | | | | | | | - Jonathan J Hogan
- Division of Nephrology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Krzysztof Kiryluk
- Division of Nephrology, Columbia University Irving Medical Center, New York
| | | | - Christoph Licht
- Division of Nephrology, University of Toronto, Toronto, ON, Canada
| | - John D Mahan
- Division of Nephrology, The Ohio State University College of Medicine, Columbus, Ohio
| | | | - Richard Quigg
- Division of Nephrology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Michelle Rheault
- Division of Nephrology, University of Minnesota, Minneapolis, Minnesota
| | - Pierre Ronco
- Division of Nephrology, Sorbonne Université, Université Pierre et Marie Curie, Paris
| | - Minnie M Sarwal
- Division of Nephrology, University of California, San Francisco, California
| | - Christine Sethna
- Division of Nephrology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
| | - Cathie Spino
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | | | - Marina Vivarelli
- Division of Nephrology, Bambino Gesu Children's Hospital, Rome, Italy
| | | | - Joshua M Thurman
- Division of Nephrology, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
22
|
Medjeral-Thomas NR, Cook HT, Pickering MC. Complement activation in IgA nephropathy. Semin Immunopathol 2021; 43:679-690. [PMID: 34379175 PMCID: PMC8551128 DOI: 10.1007/s00281-021-00882-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 07/23/2021] [Indexed: 11/25/2022]
Abstract
IgA nephropathy pathogenesis is incompletely understood, and this limits the development of disease-specific biomarkers and effective therapies. Evidence of complement activity in IgA nephropathy is well established. However, a growing body of research indicates complement activity is an important contributor to IgA nephropathy pathology. In particular, multiple associations have been identified between complement alternative, lectin and terminal pathway proteins and IgA nephropathy severity. Recently, we have also gained insight into possible mechanisms that could link glomerular IgA deposition, complement activity, glomerular inflammation and disease severity. Ongoing clinical trials of therapeutic complement inhibitors will provide insight into the importance of complement activity to IgA nephropathy pathogenesis. Further research into mechanisms of complement activity is essential to improving our understanding and management of patients with IgA nephropathy.
Collapse
Affiliation(s)
- Nicholas R Medjeral-Thomas
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, W12 0NN, UK.
| | - H Terence Cook
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, W12 0NN, UK
| | - Matthew C Pickering
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, W12 0NN, UK
| |
Collapse
|
23
|
Ding Y, Yu X, Wu L, Tan Y, Qu Z, Yu F. The Spectrum of C4d Deposition in Renal Biopsies of Lupus Nephritis Patients. Front Immunol 2021; 12:654652. [PMID: 34276649 PMCID: PMC8281350 DOI: 10.3389/fimmu.2021.654652] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 06/08/2021] [Indexed: 11/29/2022] Open
Abstract
Objectives This study aimed to determine the prevalence and localization of complement factor C4d in renal biopsies from patients with lupus nephritis (LN), as well as its associations with the disease's clinico-pathological features. The correlation between arteriolar C4d deposition and renal microvascular lesions (RVLs) was further analyzed. Methods A total of 325 biopsy-proven LN patients were enrolled, and their clinico-pathological data were collected. C4d staining of renal biopsies was performed by immunohistochemistry. The associations between C4d deposition and the clinico-pathological features were further analyzed. Results C4d deposition was present in most (98.8%) renal specimens in our cohort. These deposits were localized in the glomeruli (98.2%), tubular basement membrane (TBM) (43.7%), arterioles (31.4%), and peritubular capillary (33.8%). Patients with TBM C4d staining had higher disease activity (measured with the Systemic Lupus Erythematous Disease Activity Index) and higher National Institutes of Health pathological activity and chronicity indices (all P < 0.01). Patients with arteriolar C4d deposition were more likely to develop RVLs (91.2%) compared to those with no arteriolar C4d deposition (78.0%; P = 0.004), especially with two or more types of RVLs (P < 0.001). During the mean follow-up of 55.8 months, arteriolar C4d was related to worse renal outcomes [hazard ration (HR): 2.074, 95% confidence interval (CI) 1.056-4.075, P = 0.034]. Multivariate Cox hazard analysis showed that co-deposition of arteriolar C4d and C3c was an independent risk factor (HR: 3.681, 95% CI 1.519-8.921, P = 0.004) for predicting renal outcomes. Conclusions C4d deposition was common in renal tissues from LN patients. TBM C4d deposition was related to the disease activity, and arteriolar C4d deposition was associated with RVLs and worse renal outcomes.
Collapse
Affiliation(s)
- Ying Ding
- Renal Division, Department of Medicine, Peking University First Hospital; Institute of Nephrology, Peking University; Key Laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
- Department of Nephrology, Peking University International Hospital, Beijing, China
| | - Xiaojuan Yu
- Renal Division, Department of Medicine, Peking University First Hospital; Institute of Nephrology, Peking University; Key Laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
| | - Lihua Wu
- Department of Nephrology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ying Tan
- Renal Division, Department of Medicine, Peking University First Hospital; Institute of Nephrology, Peking University; Key Laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
| | - Zhen Qu
- Department of Nephrology, Peking University International Hospital, Beijing, China
| | - Feng Yu
- Renal Division, Department of Medicine, Peking University First Hospital; Institute of Nephrology, Peking University; Key Laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
- Department of Nephrology, Peking University International Hospital, Beijing, China
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW In this review, we discuss recent studies showing the importance of the complement pathway in kidney disease. RECENT FINDINGS Recent findings in C3 glomerulopathy (C3G) include: acute postinfectious glomerulonephritis is characterised by the presence of antifactor B antibodies; human leukocyte antigen type, but not rare complement gene variation, is associated with primary immunoglobulin-associated membranoproliferative GN and C3G. Immunohistochemistry in C3G shows that factor H related protein 5 (FHR5) is the most prevalent complement protein and correlates with kidney function. A multicentre study supported the use of mycophenolate mofetil (MMF) in C3G even after a propensity matching analysis. In immunoglobulin A nephropathy (IgAN) several studies have emphasised the importance of complement. Imbalances of circulating FH and FHR1 and FHR5, which interfere with the regulatory functions of FH, associate with IgAN. Immunohistochemistry has shown associations between glomerular FHR5 deposition and C3 activation; glomerular FHR5 associated with clinical markers of IgAN severity. Data also suggest the lectin complement pathway contributes to IgAN severity. We also discuss complement activation in thrombotic microangiopathy and other kidney diseases. SUMMARY Complement activity can be detected in a wide range of kidney diseases and this provides pathogenic insight and potential for therapy with the ongoing development of several drugs directed at complement activation.
Collapse
|
25
|
Koopman JJE, van Essen MF, Rennke HG, de Vries APJ, van Kooten C. Deposition of the Membrane Attack Complex in Healthy and Diseased Human Kidneys. Front Immunol 2021; 11:599974. [PMID: 33643288 PMCID: PMC7906018 DOI: 10.3389/fimmu.2020.599974] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/21/2020] [Indexed: 12/11/2022] Open
Abstract
The membrane attack complex-also known as C5b-9-is the end-product of the classical, lectin, and alternative complement pathways. It is thought to play an important role in the pathogenesis of various kidney diseases by causing cellular injury and tissue inflammation, resulting in sclerosis and fibrosis. These deleterious effects are, consequently, targeted in the development of novel therapies that inhibit the formation of C5b-9, such as eculizumab. To clarify how C5b-9 contributes to kidney disease and to predict which patients benefit from such therapy, knowledge on deposition of C5b-9 in the kidney is essential. Because immunohistochemical staining of C5b-9 has not been routinely conducted and never been compared across studies, we provide a review of studies on deposition of C5b-9 in healthy and diseased human kidneys. We describe techniques to stain deposits and compare the occurrence of deposits in healthy kidneys and in a wide spectrum of kidney diseases, including hypertensive nephropathy, diabetic nephropathy, membranous nephropathy, IgA nephropathy, lupus nephritis, C3 glomerulopathy, and thrombotic microangiopathies such as the atypical hemolytic uremic syndrome, vasculitis, interstitial nephritis, acute tubular necrosis, kidney tumors, and rejection of kidney transplants. We summarize how these deposits are related with other histological lesions and clinical characteristics. We evaluate the prognostic relevance of these deposits in the light of possible treatment with complement inhibitors.
Collapse
Affiliation(s)
- Jacob J E Koopman
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Mieke F van Essen
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Helmut G Rennke
- Division of Renal Pathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Aiko P J de Vries
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Cees van Kooten
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
26
|
Expanding the Role of Complement Therapies: The Case for Lupus Nephritis. J Clin Med 2021; 10:jcm10040626. [PMID: 33562189 PMCID: PMC7915321 DOI: 10.3390/jcm10040626] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/27/2021] [Accepted: 02/03/2021] [Indexed: 12/14/2022] Open
Abstract
The complement system is an innate immune surveillance network that provides defense against microorganisms and clearance of immune complexes and cellular debris and bridges innate and adaptive immunity. In the context of autoimmune disease, activation and dysregulation of complement can lead to uncontrolled inflammation and organ damage, especially to the kidney. Systemic lupus erythematosus (SLE) is characterized by loss of tolerance, autoantibody production, and immune complex deposition in tissues including the kidney, with inflammatory consequences. Effective clearance of immune complexes and cellular waste by early complement components protects against the development of lupus nephritis, while uncontrolled activation of complement, especially the alternative pathway, promotes kidney damage in SLE. Therefore, complement plays a dual role in the pathogenesis of lupus nephritis. Improved understanding of the contribution of the various complement pathways to the development of kidney disease in SLE has created an opportunity to target the complement system with novel therapies to improve outcomes in lupus nephritis. In this review, we explore the interactions between complement and the kidney in SLE and their implications for the treatment of lupus nephritis.
Collapse
|
27
|
Koopman JJE, Rennke HG, Leatherwood C, Speyer CB, D'Silva K, McMahon GM, Waikar SS, Costenbader KH. Renal deposits of complement factors as predictors of end-stage renal disease and death in patients with lupus nephritis. Rheumatology (Oxford) 2021; 59:3751-3758. [PMID: 32413140 PMCID: PMC7733722 DOI: 10.1093/rheumatology/keaa174] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 03/17/2020] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE Lupus nephritis (LN) increases the risks of end-stage renal disease (ESRD) and death, but these risks are difficult to estimate. Since complement factors play an essential role in the pathogenesis and are deposited in the kidneys as C1q and C3, we studied whether these deposits predict ESRD and death in patients with LN. METHODS We collected demographic, clinical and pathological data from 183 adult patients with LN classes II-V diagnosed with a first native kidney biopsy. Pathological data included the localization and intensity of immunofluorescence staining of C1q and C3. We obtained dates of incident ESRD and death from the United States Renal Data System and National Death Index, respectively, and evaluated survival curves and hazard ratios for ESRD and death as a composite outcome and as separate outcomes. RESULTS The presence and intensity of deposits of C1q and C3 in glomeruli, tubular walls and vascular walls differed between classes and were associated with known unfavourable prognostic factors, such as hypertension, hypoalbuminemia and hypocomplementemia. However, over a median follow-up of 7.5 years, their presence and intensity were associated with neither survival free of ESRD and death nor hazard ratios for ESRD and death. CONCLUSION Renal deposits of complement factors did not predict ESRD and death in patients with LN.
Collapse
Affiliation(s)
- Jacob J E Koopman
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Helmut G Rennke
- Renal Pathology Service, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Cianna Leatherwood
- Department of Rheumatology, Richmond Medical Center, Kaiser Permanente Medical Group, Richmond, CA, USA
| | - Cameron B Speyer
- Division of Rheumatology, Immunology, and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kristin D'Silva
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Gearoid M McMahon
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sushrut S Waikar
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Karen H Costenbader
- Division of Rheumatology, Immunology, and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
28
|
Fakhouri F, Sadallah S, Frémeaux-Bacchi V. Malignant hypertension and thrombotic microangiopathy: complement as a usual suspect. Nephrol Dial Transplant 2020; 36:gfaa362. [PMID: 33301026 DOI: 10.1093/ndt/gfaa362] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 02/24/2024] Open
Affiliation(s)
- Fadi Fakhouri
- Service of nephrology and hypertension, Department of Medicine, Lausanne University Hospital
| | - Salima Sadallah
- Service of Immunology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Véronique Frémeaux-Bacchi
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service d'Immunologie and Paris University, Paris, France
| |
Collapse
|
29
|
Boudhabhay I, Poillerat V, Grunenwald A, Torset C, Leon J, Daugan MV, Lucibello F, El Karoui K, Ydee A, Chauvet S, Girardie P, Sacks S, Farrar CA, Garred P, Berthaud R, Le Quintrec M, Rabant M, de Lonlay P, Rambaud C, Gnemmi V, Fremeaux-Bacchi V, Frimat M, Roumenina LT. Complement activation is a crucial driver of acute kidney injury in rhabdomyolysis. Kidney Int 2020; 99:581-597. [PMID: 33137339 DOI: 10.1016/j.kint.2020.09.033] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 09/01/2020] [Accepted: 09/25/2020] [Indexed: 12/17/2022]
Abstract
Rhabdomyolysis is a life-threatening condition caused by skeletal muscle damage with acute kidney injury being the main complication dramatically worsening the prognosis. Specific treatment for rhabdomyolysis-induced acute kidney injury is lacking and the mechanisms of the injury are unclear. To clarify this, we studied intra-kidney complement activation (C3d and C5b-9 deposits) in tubules and vessels of patients and mice with rhabdomyolysis-induced acute kidney injury. The lectin complement pathway was found to be activated in the kidney, likely via an abnormal pattern of Fut2-dependent cell fucosylation, recognized by the pattern recognition molecule collectin-11 and this proceeded in a C4-independent, bypass manner. Concomitantly, myoglobin-derived heme activated the alternative pathway. Complement deposition and acute kidney injury were attenuated by pre-treatment with the heme scavenger hemopexin. This indicates that complement was activated in a unique double-trigger mechanism, via the alternative and lectin pathways. The direct pathological role of complement was demonstrated by the preservation of kidney function in C3 knockout mice after the induction of rhabdomyolysis. The transcriptomic signature for rhabdomyolysis-induced acute kidney injury included a strong inflammatory and apoptotic component, which were C3/complement-dependent, as they were normalized in C3 knockout mice. The intra-kidney macrophage population expressed a complement-sensitive phenotype, overexpressing CD11b and C5aR1. Thus, our results demonstrate a direct pathological role of heme and complement in rhabdomyolysis-induced acute kidney injury. Hence, heme scavenging and complement inhibition represent promising therapeutic strategies.
Collapse
Affiliation(s)
- Idris Boudhabhay
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
| | - Victoria Poillerat
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
| | - Anne Grunenwald
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
| | - Carine Torset
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
| | - Juliette Leon
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
| | - Marie V Daugan
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
| | - Francesca Lucibello
- Institut National de la Santé et de la Recherche Médicale U932, Paris Sciences et Lettres University, Institut Curie, Paris, France
| | - Khalil El Karoui
- Service de Néphrologie et Transplantation Rénale, Hôpital Henri Mondor, Assistance Publique - Hôpitaux de Paris, Créteil, Paris, France
| | - Amandine Ydee
- Pathology Department, Lille University Hospital (Centre Hospitalier Universitaire), Pathology Institute, Institut National de la Santé et de la Recherche Médicale UMR-S1172 Lille, JPARC-Jean-Pierre Aubert Research Center, Team "Mucins, Epithelial Differentiation and Carcinogenesis," Lille University, Centre Hospitalier Universitaire Lille, Lille, France
| | - Sophie Chauvet
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France; Department of Nephrology, Georges Pompidou European Hospital, Paris, France
| | - Patrick Girardie
- Intensive Care Department, Université de Lille, Centre Hospitalier Universitaire Lille, Lille, France
| | - Steven Sacks
- Medical Research Council Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Conrad A Farrar
- Medical Research Council Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, University of Copenhagen, Copenhagen, Denmark
| | - Romain Berthaud
- Department of Pediatric Nephrology, Necker Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Moglie Le Quintrec
- Department of Nephrology and Kidney Transplantation, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Marion Rabant
- Department of Pathology, Necker Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | - Pascale de Lonlay
- Reference Centre for Metabolic Diseases, Necker-Enfants Malades Hospital, Imagine Institute, Université Paris-Descartes, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Caroline Rambaud
- Service Médecine Légale, Hôpital Raymond Poincaré, Assistance Publique - Hôpitaux de Paris, Garches, France
| | - Viviane Gnemmi
- Pathology Department, Lille University Hospital (Centre Hospitalier Universitaire), Pathology Institute, Institut National de la Santé et de la Recherche Médicale UMR-S1172 Lille, JPARC-Jean-Pierre Aubert Research Center, Team "Mucins, Epithelial Differentiation and Carcinogenesis," Lille University, Centre Hospitalier Universitaire Lille, Lille, France
| | - Veronique Fremeaux-Bacchi
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France; Laboratory of Immunology, Hôpital Européen Georges Pompidou, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Marie Frimat
- University of Lille, U995-LIRIC-Lille Inflammation Research International Center, Lille, France; Department of Nephrology, Lille University Hospital, Centre Hospitalier Universitaire, Lille, France
| | - Lubka T Roumenina
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France.
| |
Collapse
|
30
|
Complement activity is regulated in C3 glomerulopathy by IgG-factor H fusion proteins with and without properdin targeting domains. Kidney Int 2020; 99:396-404. [PMID: 33129896 PMCID: PMC7863913 DOI: 10.1016/j.kint.2020.09.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 09/11/2020] [Accepted: 09/17/2020] [Indexed: 11/29/2022]
Abstract
C3 glomerulopathy is characterized by accumulation of complement C3 within glomeruli. Causes include, but are not limited to, abnormalities in factor H, the major negative regulator of the complement alternative pathway. Factor H-deficient (Cfh-/-) mice develop C3 glomerulopathy together with a reduction in plasma C3 levels. Using this model, we assessed the efficacy of two fusion proteins containing the factor H alternative pathway regulatory domains (FH1-5) linked to either a non-targeting mouse immunoglobulin (IgG-FH1-5) or to an anti-mouse properdin antibody (Anti-P-FH1-5). Both proteins increased plasma C3 and reduced glomerular C3 deposition to an equivalent extent, suggesting that properdin-targeting was not required for FH1-5 to alter C3 activation in either plasma or glomeruli. Following IgG-FH1-5 administration, plasma C3 levels temporally correlated with changes in factor B levels whereas plasma C5 levels correlated with changes in plasma properdin levels. Notably, the increases in plasma C5 and properdin levels persisted for longer than the increases in C3 and factor B. In Cfh-/- mice IgG-FH1-5 reduced kidney injury during accelerated serum nephrotoxic nephritis. Thus, our data demonstrate that IgG-FH1-5 restored circulating alternative pathway activity and reduced glomerular C3 deposition in Cfh-/- mice and that plasma properdin levels are a sensitive marker of C5 convertase activity in factor H deficiency. The immunoglobulin conjugated FH1-5 protein, through its comparatively long plasma half-life, may be a potential therapy for C3 glomerulopathy.
Collapse
|
31
|
Hong S, Healy H, Kassianos AJ. The Emerging Role of Renal Tubular Epithelial Cells in the Immunological Pathophysiology of Lupus Nephritis. Front Immunol 2020; 11:578952. [PMID: 33072122 PMCID: PMC7538705 DOI: 10.3389/fimmu.2020.578952] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/18/2020] [Indexed: 12/17/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic, autoimmune disease that can involve virtually any organ of the body. Lupus nephritis (LN), the clinical manifestation of this disease in the kidney, is one of the most common and severe outcomes of SLE. Although a key pathological hallmark of LN is glomerular inflammation and damage, tubulointerstitial lesions have been recognized as an important component in the pathology of LN. Renal tubular epithelial cells are resident cells in the tubulointerstitium that have been shown to play crucial roles in various acute and chronic kidney diseases. In this context, recent progress has been made in examining the functional role of tubular epithelial cells in LN pathogenesis. This review summarizes recent advances in our understanding of renal tubular epithelial cells in LN, the potential role of tubular epithelial cells as biomarkers in the diagnosis, prognosis, and treatment of LN, and the future therapeutic potential of targeting the tubulointerstitium for the treatment of patients with LN.
Collapse
Affiliation(s)
- Seokchan Hong
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia.,Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Helen Healy
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia.,Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, QLD, Australia.,Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Andrew J Kassianos
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia.,Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, QLD, Australia.,Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
32
|
Fakhouri F, Le Quintrec M, Frémeaux-Bacchi V. Practical management of C3 glomerulopathy and Ig-mediated MPGN: facts and uncertainties. Kidney Int 2020; 98:1135-1148. [PMID: 32622830 DOI: 10.1016/j.kint.2020.05.053] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/06/2020] [Accepted: 05/27/2020] [Indexed: 01/11/2023]
Abstract
In recent years, a substantial body of experimental and clinical work has been devoted to C3 glomerulopathy and Ig-mediated membranoproliferative glomerulonephritis. Despite the rapid accumulation of data, several uncertainties about these 2 rare forms of nephropathies persist. They concern their pathophysiology, classification, clinical course, relevance of biomarkers and of pathology findings, and assessment of the efficacy of the available therapies. The present review discusses the impact of these uncertainties on the clinical management of patients.
Collapse
Affiliation(s)
- Fadi Fakhouri
- Service of Nephrology and Hypertension, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| | - Moglie Le Quintrec
- Department of nephrology, Université de Montpellier, CHU de Montpellier, Montpellier, France
| | - Véronique Frémeaux-Bacchi
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service d'Immunologie and Paris University, Paris, France
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW The management of lupus nephritis remains unsatisfactory due to insufficiently effective treatment regimens and the dearth of reliable predictors of disease onset or progression to guide individualized therapeutic decisions. This review summarizes new findings related to lupus nephritis over the last 18 months and discusses clinical needs that should be considered to advance trials of mechanism-based therapeutic strategies. RECENT FINDINGS Collaborative teams are addressing how to improve disease definitions and are developing predictive models for disease onset, disease response and risk of flare in individual patients. More attention is being paid to clinical trial design. Advanced technologic approaches are allowing the analysis of small amounts of human tissue and urine in unprecedented detail so as to discover new pathogenic mechanisms and identify disease biomarkers. Novel therapies continue to be tested in disease models and include new strategies to protect renal tissue from cell damage and fibrosis. SUMMARY The collaborative efforts of patients, clinical and translational researchers, the pharmaceutical industry and funding sources are needed to advance therapies for lupus nephritis. Specialized clinical centers can then deliver optimal and more personalized patient care that will improve patient outcomes.
Collapse
Affiliation(s)
- Anne Davidson
- Center for Autoimmunity, Musculoskeletal and Hematologic Diseases, Feinstein Institute for Medical Research, New York, New York, USA
| | | | | |
Collapse
|
34
|
Cassol CA, Brodsky SV, Satoskar AA, Blissett AR, Cataland S, Nadasdy T. Eculizumab deposits in vessel walls in thrombotic microangiopathy. Kidney Int 2019; 96:761-768. [DOI: 10.1016/j.kint.2019.05.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 01/05/2023]
|
35
|
Clinical promise of next-generation complement therapeutics. Nat Rev Drug Discov 2019; 18:707-729. [PMID: 31324874 DOI: 10.1038/s41573-019-0031-6] [Citation(s) in RCA: 243] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2019] [Indexed: 02/07/2023]
Abstract
The complement system plays a key role in pathogen immunosurveillance and tissue homeostasis. However, subversion of its tight regulatory control can fuel a vicious cycle of inflammatory damage that exacerbates pathology. The clinical merit of targeting the complement system has been established for rare clinical disorders such as paroxysmal nocturnal haemoglobinuria and atypical haemolytic uraemic syndrome. Evidence from preclinical studies and human genome-wide analyses, supported by new molecular and structural insights, has revealed new pathomechanisms and unmet clinical needs that have thrust a new generation of complement inhibitors into clinical development for a variety of indications. This review critically discusses recent clinical milestones in complement drug discovery, providing an updated translational perspective that may guide optimal target selection and disease-tailored complement intervention.
Collapse
|
36
|
Medjeral-Thomas NR, Moffitt H, Lomax-Browne HJ, Constantinou N, Cairns T, Cook HT, Pickering MC. Glomerular Complement Factor H-Related Protein 5 (FHR5) Is Highly Prevalent in C3 Glomerulopathy and Associated With Renal Impairment. Kidney Int Rep 2019; 4:1387-1400. [PMID: 31701048 PMCID: PMC6829196 DOI: 10.1016/j.ekir.2019.06.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/30/2019] [Accepted: 06/10/2019] [Indexed: 12/19/2022] Open
Abstract
Introduction Therapeutic agents that target complement are increasingly available for glomerular diseases. However, the mechanisms linking glomerular complement deposition with inflammation and damage are incompletely understood. Complement factor H-related protein 5 (FHR5) interacts with complement C3 and is considered to promote activation. Circulating and glomerular FHR5 associates with IgA nephropathy and abnormal FHR5 associates with familial C3 glomerulopathy (C3G). We characterized glomerular FHR5 staining in C3G and assessed its relationships with histological features of glomerular injury and clinical outcome. Methods We developed FHR5 staining protocols for formalin-fixed paraffin-embedded (FFPE) renal tissue and applied them to surplus biopsy sections from a C3G cohort. Results Glomerular FHR5 was highly prevalent in native and transplant C3G and correlated with glomerular C3 and C5b-9 staining. Glomerular FHR5 staining correlated negatively with estimated glomerular filtration rate (eGFR) (P = 0.04, difference of medians 19.7 ml/min per 1.73 m2; 95% confidence interval [CI] 1.1-43.0) and positively with a membranoproliferative glomerulonephritis pattern at diagnostic biopsy (odds ratio 18; 95% CI 1.6-201; P = 0.049). Glomerular FHR5 staining intensity positively correlated with glomerular complement C3b/iC3b/C3c (Pearson's correlation coefficient [R] = 0.59; P = 0.0008), C3dg (R = 0.47; P = 0.02) and C5b9 (R = 0.44, P = 0.02). Conclusions Glomerular FHR5 is highly prevalent in C3G, interacts with glomerular C3, and is associated with markers of disease severity. Glomerular FHR5 likely exacerbates complement-mediated glomerular damage in C3G and its interaction with glomerular complement might be exploited to target complement therapeutic agents.
Collapse
Affiliation(s)
- Nicholas R Medjeral-Thomas
- Centre for Inflammatory Disease, Division of Immunology and Inflammation, Department of Medicine, Imperial College London, UK
| | - Hilary Moffitt
- Centre for Inflammatory Disease, Division of Immunology and Inflammation, Department of Medicine, Imperial College London, UK
| | - Hannah J Lomax-Browne
- Centre for Inflammatory Disease, Division of Immunology and Inflammation, Department of Medicine, Imperial College London, UK
| | - Nicholas Constantinou
- Centre for Inflammatory Disease, Division of Immunology and Inflammation, Department of Medicine, Imperial College London, UK
| | - Tom Cairns
- Renal and Transplant Centre, Imperial College Healthcare NHS Trust, UK
| | - H Terence Cook
- Centre for Inflammatory Disease, Division of Immunology and Inflammation, Department of Medicine, Imperial College London, UK
| | - Matthew C Pickering
- Centre for Inflammatory Disease, Division of Immunology and Inflammation, Department of Medicine, Imperial College London, UK
| |
Collapse
|