1
|
Li S, Zhou X, Liu C, Wang Y, Zhou Q, Sun T. Causal association of circulating immune cells with nephrotic syndrome: evidence from a two-sample Mendelian randomization study. Int Urol Nephrol 2025; 57:1907-1917. [PMID: 39738855 DOI: 10.1007/s11255-024-04350-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 12/22/2024] [Indexed: 01/02/2025]
Abstract
OBJECTIVE Nephrotic syndrome, a debilitating manifestation of kidney disease, often arises from diverse glomerular disorders and is accompanied by notable comorbidities. Despite indications of an immunological etiology, the precise role of immune cells in its pathogenesis remains unclear. This study aimed to elucidate the causal relationships between circulating immune cell phenotypes and nephrotic syndrome using a rigorous bidirectional Mendelian randomization approach. METHODS We conducted a bidirectional Mendelian randomization analysis leveraging public genome-wide association studies (GWAS) datasets to investigate the causal links between 731 immune cell features and nephrotic syndrome. Our primary analysis employed inverse variance weighting (IVW), complemented by MR-Egger regression, simple model, weighted median method, and weighted model techniques to ensure robustness. Sensitivity analyses were performed to address potential biases arising from heterogeneity, horizontal pleiotropy, and single-nucleotide polymorphism (SNP) instability in nephrotic syndrome. RESULTS Among traits examined, 13 immune cell phenotypes were identified to have significant causal impacts on nephrotic syndrome (adjusted P > < 0.05). Among these phenotypes, CD25 on unswitched memory B cell, CD25 on memory B cell, CD25 on CD24 + CD27 + B cell, CD25 on IgD-CD38-B cell, CD33dim HLA DR-Absolute Count, and CD127 on granulocyte emerged as causal risk factors, while seven circulating immune cell phenotypes, predominantly monocyte subsets, exhibited protective effects. Furthermore, the reverse Mendelian randomization analysis demonstrated significant effects of nephrotic syndrome on 27 immune phenotypes (P < 0.05). CONCLUSION The genetic predictions indicate that multiple circulating immune cell phenotypes, particularly CD25 on specific B-cell subsets, serve as independent risk factors for the onset and progression of nephrotic syndrome. Conversely, monocytes expressing specific phenotypes may exert protective effects against the development of nephrotic syndrome. These findings offer a novel therapeutic approach for the prevention and treatment of nephrotic syndrome.
Collapse
Affiliation(s)
- Sheng Li
- Department of Nephrology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, China
| | - Xing Zhou
- Department of Urology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, China
| | - Chengmeng Liu
- Department of Nephrology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, China
| | - Yijie Wang
- Department of Urology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, China
| | - Qianhui Zhou
- Department of Emergency, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, China.
| | - Ting Sun
- Department of Emergency, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, China.
| |
Collapse
|
2
|
Horinouchi T, Nozu K, Iijima K. Genetic aspects of pediatric nephrotic syndrome and anti-nephrin antibodies. Clin Exp Nephrol 2025; 29:534-540. [PMID: 40085383 PMCID: PMC12049277 DOI: 10.1007/s10157-025-02645-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 02/14/2025] [Indexed: 03/16/2025]
Abstract
Nephrotic syndrome is the most common glomerular disease in children, and various hypotheses regarding its etiology have been proposed, primarily focusing on immune-related mechanisms. Nephrotic syndrome can manifest as a monogenic disease caused by deleterious variants in genes such as NPHS1, which encodes nephrin. In steroid-sensitive nephrotic syndrome, HLA class II and immune-related genes have been identified as susceptibility genes. Moreover, NPHS1 is a susceptibility gene for steroid-sensitive nephrotic syndrome in patients from East Asian populations. Anti-nephrin antibodies have been identified as a significant factor in the pathogenesis of nephrotic syndrome. These discoveries have substantially advanced our understanding of nephrotic syndrome. However, the mechanisms underlying the production of anti-nephrin antibodies and their association with genetic backgrounds have remained unclear and warrant further investigation.
Collapse
Affiliation(s)
- Tomoko Horinouchi
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe University, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe University, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kazumoto Iijima
- Hyogo Prefectural Kobe Children's Hospital, Hyogo, Japan
- Department of Advanced Pediatric Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
3
|
Alqudsi M, Alhumaid S, Almagooshi AS, Samman AM, Alqaraishi AM. Characteristics of Dyslipidemia in Primary Nephrotic Syndromes. Cureus 2025; 17:e84077. [PMID: 40519478 PMCID: PMC12163688 DOI: 10.7759/cureus.84077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2025] [Indexed: 06/18/2025] Open
Abstract
Background Although it is not a criterion for diagnosis, dyslipidemia is frequently found in nephrotic syndrome (NS). Cholesterol, triglyceride, and low-density lipoprotein (LDL) are usually elevated in NS, and high-density lipoprotein (HDL) can be normal or minimally decreased. Dyslipidemia in NS has been studied in isolation of the underlying glomerulopathy, and the comparison of lipid values between membranous nephropathy (MN), minimal change disease (MCD), and focal segmental glomerulosclerosis (FSGS), is not well recognized. Methods Retrospective chart review of patients with NS from 2010 to 2022. Patients with primary MN, MCD, and primary FSGS were included. Lipid profile was reported at the time of NS diagnosis and 12 months later. We compared lipid values between three primary NS using Kruskal-Wallis and Mann-Whitney U tests. Results There were 409 patients diagnosed with NS. 284 patients were excluded due to insufficient data or a diagnosis of secondary NS. One hundred and twenty-five patients with FSGS, MN, or MCD were included: FSGS (52, 41%), MCD (31, 25%), and MN (42, 34%). The average age was 32 years, with 55 females (44%), and 79 received statins (56%). After adjustment for serum albumin and proteinuria, initial cholesterol and triglyceride levels were similar in the three NS groups (P>0.05). Low-density lipid (LDL) was 216 mg/dL, 201 mg/dL, and 178 mg/dL in FSGS, MCD, and MN, respectively; the difference was only in FSGS vs MN group (p=0.04). Initial HDL was 58 mg/dL, 77 mg/dL, and 50 mg/dL in FSGS, MCD, and MN respectively (p=<0.001), differences were in MCD vs FSGS, and MCD vs MN groups (p=0.001, and p=<0.001 respectively). After 12 months of follow-up, lipid values were similar in the three NS groups regardless of statin use. Conclusion After adjustment for primary NS severity, cholesterol and triglyceride values were insignificantly different at the presentation of MN, MCD, and FSGS. HDL was significantly higher in MCD compared to MN and FSGS, and LDL was significantly higher in FSGS compared to MN. At the 12-month follow-up, statin use did not change lipid values.
Collapse
Affiliation(s)
| | - Sulaiman Alhumaid
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences College of Medicine, Riyadh, SAU
| | | | - Ahmad M Samman
- Nephrology, King Abdulaziz Medical City Riyadh, Riyadh, SAU
| | | |
Collapse
|
4
|
Robinson CH, Smoyer WE, Cara-Fuentes G. Unraveling the Immunogenetic Mechanisms of Childhood Idiopathic Nephrotic Syndrome. J Pediatr 2025; 282:114595. [PMID: 40252964 DOI: 10.1016/j.jpeds.2025.114595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/16/2025] [Accepted: 04/14/2025] [Indexed: 04/21/2025]
Affiliation(s)
- Cal H Robinson
- Division of Nephrology, The Hospital for Sick Children, Toronto, ON, Canada; Child Health Evaluative Sciences, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada; Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada.
| | - William E Smoyer
- The Center for Clinical and Translational Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH; Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH
| | - Gabriel Cara-Fuentes
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH
| |
Collapse
|
5
|
Liu H, Zhou C, Wang D, Meng H, Zhu S, Zhang J, Mao J, Ye Q. Autoantibodies Targeting Proteasome Subunit Alpha Type 1 in Autoimmune Podocytopathies. J Am Soc Nephrol 2025; 36:406-419. [PMID: 39382973 PMCID: PMC11888960 DOI: 10.1681/asn.0000000525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024] Open
Abstract
Key Points Serum anti-proteasome subunit alpha type 1 (PSMA1) autoantibodies were specifically elevated in the active phase of idiopathic nephrotic syndrome, which may assist in disease diagnosis and monitoring. Serum anti-PSMA1 antibodies could cause damage to the glomerular filtration barrier, which may be a pathogenic antibody of idiopathic nephrotic syndrome. PSMA1 played an important role in the maintenance of podocyte morphology and function. Background The antibody against proteasome subunit alpha type 1 (PSMA1) is a podocyte autoantibody in children with idiopathic nephrotic syndrome identified in our previous study. The aim of this study was to explore the characteristics of idiopathic nephrotic syndrome in children and the mechanism underlying its involvement in the development of idiopathic nephrotic syndrome. Methods The levels of serum anti-PSMA1 autoantibodies in children were detected through protein microarray and compared among different disease groups. The recombinant PSMA1 protein was injected subcutaneously and intraperitoneally into mice to observe glomerular morphology and function. The PSMA1-knockdown and PSMA1-overexpressing cell lines were constructed from mouse podocytes, and their cytoskeleton and function were analyzed. Homozygous zebrafish with psma1 knockout were observed. Results The levels of serum anti-PSMA1 autoantibodies were higher in children with idiopathic nephrotic syndrome and varied with urinary protein. In mice immunized with PSMA1, the presence of serum anti-PSMA1 autoantibody caused albuminuria and damage to the glomerular filtration membrane. Deficiency of PSMA1 impaired the podocyte cytoskeleton and physiological function. Complete deletion of psma1 caused edema, abnormal glomerular morphology, and effacement of foot processes in zebrafish. Conclusions PSMA1 played an important role in the maintenance of podocyte morphology and function.
Collapse
Affiliation(s)
- Huihui Liu
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Chao Zhou
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Dongjie Wang
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Hanyan Meng
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Shifan Zhu
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Jiayu Zhang
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Jianhua Mao
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Qing Ye
- Department of Laboratory Medicine, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| |
Collapse
|
6
|
Chen Q, Chen S, Ye Q, Lin W, Liao Y, Xiong Y, Xu J, Gao R, Li B, Liu L, Wei L. Anti-nephrin antibody: a potential biomarker of minimal change disease. Clin Kidney J 2025; 18:sfaf012. [PMID: 40046822 PMCID: PMC11879414 DOI: 10.1093/ckj/sfaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Indexed: 04/05/2025] Open
Abstract
Background Minimal change disease (MCD) is a common pathological type of nephrotic syndrome in children and adults, and the mechanisms remain obscure. The diagnosis of MCD still relies on renal biopsy, lacking effective biological markers. This study explores the diagnostic value of circulating anti-nephrin antibody in MCD patients and evaluates the correlation with disease activity indicators such as proteinuria. Methods The study included 108 adult patients with glomerular disease, including 36 with MCD, 16 with primary focal segmental glomerulosclerosis (FSGS), 20 with primary membranous nephropathy (MN), 17 with diabetic nephropathy (DN) and 19 with immunoglobulin A nephropathy (IgAN). Twenty healthy volunteers were included. Circulating anti-nephrin antibody was detected by indirect immunofluorescence method of cell-based assay. The receiver-operating characteristic (ROC) curve was used to evaluate the role of circulating anti-nephrin antibody in the diagnosis of MCD. The correlations between anti-nephrin antibody and clinical parameters were analyzed. Results The prevalence of circulating anti-nephrin antibody was 19.44% (7 of 36) in MCD and 26.92% (7 of 26) in MCD patients with nephrotic proteinuria, which was higher than in FSGS, PMN, DN, IgAN and healthy volunteers. The ROC curve showed that the sensitivity of anti-nephrin antibody used in the diagnosis of MCD was 19.4% and the specificity was 97.8%. The MCD patients with positive anti-nephrin antibody had heavier proteinuria and higher serum lipid levels, while having lower serum albumin and blood IgG levels. Anti-nephrin antibody might turn to negative when the MCD patient had a response to therapy. Conclusions Circulating anti-nephrin antibody may be a potential biomarker of MCD and may play a role in the MCD diagnosis.
Collapse
Affiliation(s)
- Qiaoling Chen
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Institute of Clinical Immunology, Fuzhou, China
| | - Sihui Chen
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Qiuping Ye
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Institute of Clinical Immunology, Fuzhou, China
| | - Wanjun Lin
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Institute of Clinical Immunology, Fuzhou, China
| | - Yonggen Liao
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Institute of Clinical Immunology, Fuzhou, China
| | - Yunfeng Xiong
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jiaming Xu
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ruiyu Gao
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Binbin Li
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Institute of Clinical Immunology, Fuzhou, China
| | - Lifang Liu
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Institute of Clinical Immunology, Fuzhou, China
| | - Lixin Wei
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Institute of Clinical Immunology, Fuzhou, China
| |
Collapse
|
7
|
May CJ, Ford NP, Welsh GI, Saleem MA. Biomarkers to predict or measure steroid resistance in idiopathic nephrotic syndrome: A systematic review. PLoS One 2025; 20:e0312232. [PMID: 39946431 PMCID: PMC11824968 DOI: 10.1371/journal.pone.0312232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 10/02/2024] [Indexed: 02/16/2025] Open
Abstract
In this systematic review we have sought to summarise the current knowledge concerning biomarkers that can distinguish between steroid-resistant nephrotic syndrome and steroid-sensitive nephrotic syndrome. Additionally, we aim to select biomarkers that have the best evidence-base and should be prioritised for further research. Pub med and web of science databases were searched using "steroid resistant nephrotic syndrome AND biomarker". Papers published between 01/01/2012 and 10/05/2022 were included. Papers that did not compare steroid resistant and steroid sensitive nephrotic syndrome, did not report sensitivity/specificity or area under curve and reviews/letters were excluded. The selected papers were then assessed for bias using the QUADAS-2 tool. The source of the biomarker, cut off, sensitivity/specificity, area under curve and sample size were all extracted. Quality assessment was performed using the BIOCROSS tool. 17 studies were included, comprising 15 case-control studies and 2 cross-sectional studies. Given the rarity of nephrotic syndrome and difficulty in recruiting large cohorts, case-control studies were accepted despite their limitations. We present a range of candidate biomarkers along with scores relating to the quality of the original publications and the risk of bias to inform future investigations. None of the selected papers stated whether the authors were blinded to the patient's disease when assessing the index test in the cohort. Highlighting a key problem in the field that needs to be addressed. These candidate biomarkers must now be tested with much larger sample sizes. Using new biobanks such as the one built by the NURTuRE-INS team will be very helpful in this regard.
Collapse
Affiliation(s)
- Carl J. May
- Bristol Renal, University of Bristol, Bristol, United Kingdom
| | | | - Gavin I. Welsh
- Bristol Renal, University of Bristol, Bristol, United Kingdom
| | - Moin A. Saleem
- Bristol Renal, University of Bristol, Bristol, United Kingdom
- Bristol Royal Hospital for Children, Bristol, United Kingdom
| |
Collapse
|
8
|
Kaneko K. Gut dysbiosis as a susceptibility factor in childhood idiopathic nephrotic syndrome. Pediatr Neonatol 2025; 66 Suppl 1:S2-S7. [PMID: 39521679 DOI: 10.1016/j.pedneo.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Idiopathic nephrotic syndrome (INS) is a relatively common renal disorder of childhood characterized by severe proteinuria and associated hypoproteinemia and edema. Although the pathogenesis of INS remains unknown, the prevailing theory of its pathogenesis is as follows. Antigenic stimulation, such as viral infections or vaccines, in children with susceptibility factors for INS triggers abnormal immune responses, resulting in production of pathogenic substances that injure podocytes (renal glomerular epithelial cells). The injured podocytes then change their function and morphology, resulting in increased permeability of plasma proteins. Consequently, plasma proteins, especially albumin, are leaked into urine and massive proteinuria ensues. Research on susceptibility factors for INS has focused on polymorphisms in several genes including human leukocyte antigen class II genes. However, we propose that dysbiosis of the intestinal microbiota could be a susceptibility factor for relapse. This proposal is based on our research group finding that children with INS and frequent relapses have gut dysbiosis characterized by a decreased proportion of beneficial bacteria such as short-chain fatty acid-producing bacteria. Dysbiosis from the neonatal period to infancy may result from environmental factors, such as cesarean section delivery and antibiotic administration, which prevent the establishment of a normal intestinal microbiota. Dysbiosis leads to aberrant gut immunity and is characterized by a decreased ratio of T helper 1 cells/T helper 2 cells and an increased ratio of T helper 17 cells/regulatory T-cells. Therefore, relapse occurs when immunologically pathogenic factors that injure podocytes are produced in response to trigger events in children with INS and gut dysbiosis. Our recent clinical trial suggested that long-term oral administration of butyric acid-producing bacterium as a probiotic is promising for suppressing relapse. Therefore, studying the causal relationship between dysbiosis and relapses in patients with INS in a larger number of patients is necessary.
Collapse
Affiliation(s)
- Kazunari Kaneko
- Department of Pediatrics, Kansai Medical University, 2-5-1, Shin-machi, Hirakata, Osaka, 573 1010, Japan.
| |
Collapse
|
9
|
Semenikhina M, Mathew RO, Barakat M, Van Beusecum JP, Ilatovskaya DV, Palygin O. Blood Pressure Management Strategies and Podocyte Health. Am J Hypertens 2025; 38:85-96. [PMID: 39269328 DOI: 10.1093/ajh/hpae120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 07/24/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024] Open
Abstract
Hypertension (HTN) is one of the key global cardiovascular risk factors, which is tightly linked to kidney health and disease development. Podocytes, glomerular epithelial cells that play a pivotal role in maintenance of the renal filtration barrier, are significantly affected by increased glomerular capillary pressure in HTN. Damage or loss of these cells causes proteinuria, which marks the initiation of the HTN-driven renal damage. It goes without saying that effective blood pressure (BP) management should not only mitigate cardiovascular risks but also preserve renal function by protecting podocyte integrity. This review offers a comprehensive examination of current BP management strategies and their implications for podocyte structure and function and emphasizes strategies for the reduction of proteinuria in HTN. We explore primary and secondary antihypertensive agents, including angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, calcium channel blockers, and diuretics, as well as newer therapies (sodium-glucose cotransporter-2 blocking and endothelin receptor antagonism), emphasizing their mechanistic roles in safeguarding podocytes and curtailing proteinuria.
Collapse
Affiliation(s)
- Marharyta Semenikhina
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Roy O Mathew
- Division of Nephrology, Department of Medicine, VA Loma Linda Healthcare System, Loma Linda, California, USA
| | - Munsef Barakat
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Justin P Van Beusecum
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Ralph H. Johnson VA Medical Center, Charleston, South Carolina, USA
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
10
|
Srivastava T, Sharma M. Emerging Role of SH3BP2 as Regulator of Immune and Nonimmune Cells in Nephrotic Syndrome. GLOMERULAR DISEASES 2025; 5:1-12. [PMID: 39991193 PMCID: PMC11842026 DOI: 10.1159/000542703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 11/17/2024] [Indexed: 02/25/2025]
Abstract
Background Minimal change disease (MCD) and focal segmental glomerulosclerosis (FSGS) are major forms of nephrotic syndrome that remain difficult to treat. MCD and FSGS have distinct but also overlapping clinical, histological, metabolic, and molecular features. Effective use of immunosuppressive drugs, activated immune cells, altered cytokine profiles, and upregulated signaling pathways suggest a link between immune dysfunction and nephrotic syndrome, but the exact mechanism of immunopathogenesis is unclear. Immune dysfunction is an area of ongoing research for identifying novel molecular targets for treating nephrotic syndrome. However, the available animal models do not directly address the role of immune dysfunction in nephrotic syndrome. Summary Genetic analysis indicates that heterogeneous genes related to the podocyte-specific proteins may indirectly cause damage to filtration barrier and influence the onset and progression of nephrotic syndrome. SH3BP2 protein regulates several pathways through its role as a scaffold for many signaling mediators and enzymes. SH3BP2 is expressed in immune as well as in nonimmune cells including podocytes. The role of SH3BP2 is discussed in the context of cells and molecules of adaptive and innate immune systems. Available information on the importance of SH3BP2 in diseases other than nephrotic syndrome and its role in the immunopathogenesis of human nephrotic syndrome are summarized. We outline the key features of a transgenic mouse strain with a gain-in-function mutation (Sh3bp2 KI/KI ) as a potential model to study immunopathogenesis of nephrotic syndrome. Key Messages Non-receptor, non-catalytic proteins such as SH3BP2 are a novel group of proteins that regulate the innate and adaptive immune responses in nephrotic syndrome. New evidence suggests a critical role of SH3BP2 in immunopathogenesis of nephrotic syndrome. Our recent results demonstrate that transgenic mice (Sh3bp2 KI/KI ) with a gain-in-function mutation will likely be a unique model to study immunopathogenesis of nephrotic syndrome.
Collapse
Affiliation(s)
- Tarak Srivastava
- Section of Nephrology, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO, USA
- Kansas City VA Medical Center, Kansas City, MO, USA
| | - Mukut Sharma
- Kansas City VA Medical Center, Kansas City, MO, USA
- Midwest Veterans’ Biomedical Research Foundation (MVBRF), Kansas City, MO, USA
| |
Collapse
|
11
|
Jefferson JA, Chen K, Hingorani S, Malik AB, Tykodi SS, Keller KH, Huang Y, Smith KD, Reed RC, Weins A, Akilesh S. Genetic and Iatrogenic Defects in Peripheral Tolerance Associated with Anti-Nephrin Antibody-Associated Minimal Change Disease. GLOMERULAR DISEASES 2025; 5:74-83. [PMID: 39991196 PMCID: PMC11845169 DOI: 10.1159/000543334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/19/2024] [Indexed: 02/25/2025]
Abstract
Introduction Minimal change disease (MCD) is a common cause of nephrotic syndrome in children and adults. Immune dysregulation is a contributor, but the relative roles of individual components of the immune system in MCD pathogenesis remain unclear. Case Presentation Here, we present 2 patients with defects in immune tolerance mechanisms that developed MCD associated with anti-nephrin antibodies. The first patient had a pathogenic deletion in FOXP3, leading to reduced regulatory T cells. Serum could not be obtained from this patient during the active phase of MCD to directly establish the presence of anti-nephrin antibodies. However, this patient demonstrated IgG dusting over podocyte cell bodies by immunofluorescence microscopy, as well as colocalization of IgG with nephrin in confocal microscopy. The second patient developed MCD in the context of immune checkpoint inhibitor treatment for metastatic carcinoma. Anti-nephrin antibodies were detected in this patient during active disease. The patient's kidney biopsy also showed evidence of binding of anti-nephrin antibodies within the glomeruli. Conclusion These cases demonstrate that genetic and iatrogenic mechanisms of breakdown in peripheral tolerance can lead to MCD.
Collapse
Affiliation(s)
- J. Ashley Jefferson
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Karin Chen
- Division of Immunology, Department of Pediatrics, University of Washington, Seattle, WA, USA
- Seattle Chilidren’s Research Institute, Seattle, WA, USA
| | - Sangeeta Hingorani
- Division of Immunology, Department of Pediatrics, University of Washington, Seattle, WA, USA
- Seattle Chilidren’s Research Institute, Seattle, WA, USA
| | - A. Bilal Malik
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Scott S. Tykodi
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Yuan Huang
- Robert J. Tomsich Department of Pathology and Laboratory Medicine, Diagnostics Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kelly D. Smith
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Robyn C. Reed
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, Seattle Children’s Hospital, Seattle, WA, USA
| | - Astrid Weins
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Shreeram Akilesh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Kidney Research Institute, Seattle, WA, USA
| |
Collapse
|
12
|
Zhu S, Zhang J, Gao L, Ye Q, Mao J. The Pathogenesis of Nephrotic Syndrome: A Perspective from B Cells. KIDNEY DISEASES (BASEL, SWITZERLAND) 2024; 10:531-544. [PMID: 39664337 PMCID: PMC11631018 DOI: 10.1159/000540511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/20/2024] [Indexed: 12/13/2024]
Abstract
Background Nephrotic syndrome is a special type of chronic kidney disease, the specific pathogenesis of which remains unclear. An increasing number of studies have suggested that B cells play an important role in the pathogenesis of nephrotic syndrome. Summary Idiopathic nephrotic syndrome is a common kidney disease in children. While previously believed to be primarily caused by T-cell disorders, recent research has shifted its focus to B cells. Studies have shown that B cells play a significant role in the pathogenesis of NS, potentially even more so than T cells. This article provides a comprehensive review of the involvement of B cells in the development of idiopathic nephrotic syndrome. Key Messages B cells are involved in the pathogenesis of nephrotic syndrome by producing autoantibodies and various cytokines.
Collapse
Affiliation(s)
- Shifan Zhu
- Department of Nephrology, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Jiayu Zhang
- Department of Nephrology, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Langping Gao
- Department of Nephrology, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Qing Ye
- Department of Clinical Laboratory, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Jianhua Mao
- Department of Nephrology, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| |
Collapse
|
13
|
Ichikawa Y, Sakakibara N, Nagano C, Inoki Y, Tanaka Y, Ueda C, Kitakado H, Kondo A, Ishimori S, Horinouchi T, Iijima K, Nozu K. In steroid-resistant nephrotic syndrome that meets the strict definition, monogenic variants are less common than expected. Pediatr Nephrol 2024; 39:3497-3503. [PMID: 39093455 PMCID: PMC11511720 DOI: 10.1007/s00467-024-06468-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND In patients with steroid-resistant nephrotic syndrome (SRNS), the presence of monogenic variants influences therapeutic strategies. Large cohort studies reported the detection of monogenic variants in approximately 30% of patients with SRNS. However, these cohorts included many patients, such as those with symptomatic proteinuria, who did not meet the strict diagnostic criteria for pediatric nephrotic syndrome (NS). Therefore, we investigated the proportion of causative monogenic variants detected in patients who strictly met the diagnostic criteria of SRNS and explored their clinical characteristics. METHODS We examined pediatric SRNS cases with genetic analysis conducted in our hospital. Cases satisfying all of the following criteria were included: (1) age at onset 1-18 years, (2) serum albumin at onset ≤ 2.5 g/dl, (3) persistent heavy proteinuria, and (4) no complete remission after 4 weeks of steroid monotherapy. RESULTS The proportion of detected monogenic variants was 12% (22/185) among all patients. The proportion was only 7% (9/129) in patients with edema at disease onset compared with 38% (9/24) in those without (p < 0.0001). Monogenic variants were rare in patients with acute kidney injury associated with NS (1% (1/11)) or a history of complete remission (4% (2/51)). CONCLUSIONS Our study revealed a monogenic cause in 12% of individuals with strictly defined SRNS, a much smaller proportion than previously reported. The presence or absence of edema at the onset was an important factor to distinguish SRNS with monogenic cause from SRNS without. Our results provide further evidence of the SRNS types attributable to monogenic causes.
Collapse
Affiliation(s)
- Yuta Ichikawa
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, 650-0017, Japan.
| | - Nana Sakakibara
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, 650-0017, Japan
| | - China Nagano
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, 650-0017, Japan
| | - Yuta Inoki
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, 650-0017, Japan
| | - Yu Tanaka
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, 650-0017, Japan
| | - Chika Ueda
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, 650-0017, Japan
| | - Hideaki Kitakado
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, 650-0017, Japan
| | - Atsushi Kondo
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, 650-0017, Japan
| | - Shingo Ishimori
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, 650-0017, Japan
| | - Tomoko Horinouchi
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, 650-0017, Japan
| | - Kazumoto Iijima
- Hyogo Prefectural Kobe Children's Hospital, Kobe, Japan
- Department of Advanced Pediatric Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, 650-0017, Japan
| |
Collapse
|
14
|
Liu J, Wang Y, Qu Z, Si J, Jiang Y. Aberrant frequency of circulating IL-21+ T follicular helper cells in patients with primary focal segmental glomerulosclerosis. Mol Immunol 2024; 176:30-36. [PMID: 39561490 DOI: 10.1016/j.molimm.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/03/2024] [Accepted: 11/12/2024] [Indexed: 11/21/2024]
Abstract
Follicular helper T (Tfh) cells have been implicated in the pathophysiology of numerous diseases. This study investigated the hypothetical function of peripheral blood IL-21+ Tfh cells in the etiology of focal segmental glomerulosclerosis (FSGS). Tfh cell subsets were identified via flow cytometry in PBMCs from 15 patients with FSGS and 9 healthy controls (HCs). Moreover, a cytometric bead array (CBA) was used to determine the level of IL-21 in the serum. The proportions of IL-21+ cTfh cells, IL-21+ PD-1+ cTfh cells and serum IL-21 were lower in FSGS patients than in HCs. In FSGS patients, the serum IL-21 concentration was positively correlated with the frequency of IL-21+ cTfh cells and IL-21+ PD-1+ cTfh cells. The frequencies of IL-21+ cTfh cells and IL-21+ PD-1+ cTfh cells were negatively associated with 24-h urine protein but positively correlated with eGFR, serum albumin and serum IgG. CONCLUSIONS: An aberrant frequency of IL-21+ Tfh cells was detected in FSGS patients, which may provide a better understanding of FSGS pathogenesis.
Collapse
Affiliation(s)
- Jing Liu
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun 130021, China; Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, 150 Ji Mo Road, Shanghai 200120, China.
| | - Yanbo Wang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun 130021, China.
| | - Zhihui Qu
- Department of Nephrology, the First Hospital of Jilin University, Changchun 130021, China.
| | - Junzhuo Si
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun 130021, China.
| | - Yanfang Jiang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
15
|
Kardani AK, Fitri LE, Samsu N, Subandiyah K. Forging the Future: B Cell Activating Factor's Impact on Nephrotic Syndrome. Malays J Med Sci 2024; 31:57-64. [PMID: 39830109 PMCID: PMC11740809 DOI: 10.21315/mjms2024.31.6.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 07/04/2024] [Indexed: 01/22/2025] Open
Abstract
Nephrotic syndrome is the most common glomerular disease in children. While the exact pathogenesis of nephrotic syndrome is not fully understood, recent research has shed light on some of the underlying mechanisms involved in it. Improvement by B cell depletion therapy using antiCD20 in nephrotic syndrome has led to a paradigm shift from immunoinflammatory disease influenced by T cell dysregulation to B cell involvement in the pathogenesis of nephrotic syndrome. The expression of the B cell activating factor (BAFF), an essential cytokine for the maturation and differentiation of B lymphocytes, in the podocytes of paediatric patients with nephrotic syndrome is known to be associated with worse renal outcomes. The pro-inflammatory cytokines and pathogenic antibodies produced by B cells allegedly cause podocyte injury leading to proteinuria due to effacement of foot processes. Considering the role of the BAFF in B cell proliferation and antibody production, BAFF signalling is a potential target for development as targeted therapy in nephrotic syndrome. Nevertheless, there is limited research regarding the role of BAFF in nephrotic syndrome, and the exact mechanism of BAFF involvement in the pathogenesis of nephrotic syndrome is still unknown. This review discusses the role of the BAFF in the pathogenesis of nephrotic syndrome and highlights the gap of knowledge for future research.
Collapse
Affiliation(s)
- Astrid Kristina Kardani
- Doctoral Program in Medical Sciences, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- Nephrology Division, Department of Paediatric, Faculty of Medicine, Universitas Brawijaya, Dr. Saiful Anwar General Hospital, Malang, Indonesia
| | - Loeki Enggar Fitri
- Department of Clinical Parasitology, Faculty of Medicine, Universitas Brawijaya, Dr. Saiful Anwar General Hospital, Malang, Indonesia
| | - Nur Samsu
- Nephrology Division, Department of Internal Medicine, Faculty of Medicine, Universitas Brawijaya, Dr. Saiful Anwar General Hospital, Malang, Indonesia
| | - Krisni Subandiyah
- Nephrology Division, Department of Paediatric, Faculty of Medicine, Universitas Brawijaya, Dr. Saiful Anwar General Hospital, Malang, Indonesia
| |
Collapse
|
16
|
Jia Y, Xiong S, Chen H, Liu D, Wu X. Exosomes secreted by podocytes regulate the differentiation of Th17/Treg cells in idiopathic nephrotic syndrome. Heliyon 2024; 10:e37866. [PMID: 39315171 PMCID: PMC11417541 DOI: 10.1016/j.heliyon.2024.e37866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/25/2024] Open
Abstract
Background Previous studies have demonstrated that immune cells release exosomes, which act as antigen-presenting vesicles to activate T cells. In our previous study, we discovered that podocytes, a type of kidney cell, can also exhibit antigen-presenting functions to naïve CD4+ T cells in idiopathic nephrotic syndrome (INS). Building upon these findings, the objective of this study was to investigate whether podocytes can regulate the balance between Th17 and Treg cells through the release of exosomes. Methods We co-cultured naïve CD4+ T cells with LPS-treated bone marrow dendritic cells (LPS-BMDC), LPS-treated mouse podocyte clone 5 (LPS-MPC-5), and exosomes derived from LPS-MPC-5 (LPS-EXO). As a control group, naïve CD4+ T cells were cultured with exosomes from untreated MPC-5 (EXO). After 48 h, we analyzed the percentages of Th17 and Treg cells using flow cytometry, measured the concentrations of IL-17A, IL-10, and IL-4 were using ELISA, and examined the expressions of IL-17a, IL-10, RORC, and FOXP3 using RT-qPCR. Results We confirmed the presence of exosomes derived from podocytes based on their morphology, size distribution, concentrations, and the levels of exosomes-specific markers. The percentage of Th17 and Treg cells in the LPS-EXO group was significantly higher than that in the control groups, but lower than in the LPS-MPC-5 group. Furthermore, the ratio of Th17/Treg was relatively higher in the LPS-EXO group compared to the LPS-MPC-5 group. Conclusion This study indicated further insights into the role of exosomes released from LPS-treated podocytes in regulating the balance between Th17 and Treg cells in INS.
Collapse
Affiliation(s)
- Yang Jia
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shiqiu Xiong
- Department of Gastroenterology, Xi'an Children's Hospital, Xi 'an, Shanxi, China
| | - Haixia Chen
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Donghai Liu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaochuan Wu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
17
|
Hengel FE, Dehde S, Lassé M, Zahner G, Seifert L, Schnarre A, Kretz O, Demir F, Pinnschmidt HO, Grahammer F, Lucas R, Mehner LM, Zimmermann T, Billing AM, Oh J, Mitrotti A, Pontrelli P, Debiec H, Dossier C, Colucci M, Emma F, Smoyer WE, Weins A, Schaefer F, Alachkar N, Diemert A, Hogan J, Hoxha E, Wiech T, Rinschen MM, Ronco P, Vivarelli M, Gesualdo L, Tomas NM, Huber TB. Autoantibodies Targeting Nephrin in Podocytopathies. N Engl J Med 2024; 391:422-433. [PMID: 38804512 DOI: 10.1056/nejmoa2314471] [Citation(s) in RCA: 81] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
BACKGROUND Minimal change disease and primary focal segmental glomerulosclerosis in adults, along with idiopathic nephrotic syndrome in children, are immune-mediated podocytopathies that lead to nephrotic syndrome. Autoantibodies targeting nephrin have been found in patients with minimal change disease, but their clinical and pathophysiological roles are unclear. METHODS We conducted a multicenter study to analyze antinephrin autoantibodies in adults with glomerular diseases, including minimal change disease, focal segmental glomerulosclerosis, membranous nephropathy, IgA nephropathy, antineutrophil cytoplasmic antibody-associated glomerulonephritis, and lupus nephritis, as well as in children with idiopathic nephrotic syndrome and in controls. We also created an experimental mouse model through active immunization with recombinant murine nephrin. RESULTS The study included 539 patients (357 adults and 182 children) and 117 controls. Among the adults, antinephrin autoantibodies were found in 46 of the 105 patients (44%) with minimal change disease, 7 of 74 (9%) with primary focal segmental glomerulosclerosis, and only in rare cases among the patients with other conditions. Of the 182 children with idiopathic nephrotic syndrome, 94 (52%) had detectable antinephrin autoantibodies. In the subgroup of patients with active minimal change disease or idiopathic nephrotic syndrome who were not receiving immunosuppressive treatment, the prevalence of antinephrin autoantibodies was as high as 69% and 90%, respectively. At study inclusion and during follow-up, antinephrin autoantibody levels were correlated with disease activity. Experimental immunization induced a nephrotic syndrome, a minimal change disease-like phenotype, IgG localization to the podocyte slit diaphragm, nephrin phosphorylation, and severe cytoskeletal changes in mice. CONCLUSIONS In this study, circulating antinephrin autoantibodies were common in patients with minimal change disease or idiopathic nephrotic syndrome and appeared to be markers of disease activity. Their binding at the slit diaphragm induced podocyte dysfunction and nephrotic syndrome, which highlights their pathophysiological significance. (Funded by Deutsche Forschungsgemeinschaft and others.).
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Child
- Child, Preschool
- Female
- Humans
- Male
- Mice
- Middle Aged
- Autoantibodies/blood
- Autoantibodies/immunology
- Biopsy
- Disease Models, Animal
- Glomerulonephritis, IGA/blood
- Glomerulonephritis, IGA/immunology
- Glomerulonephritis, IGA/pathology
- Glomerulosclerosis, Focal Segmental/blood
- Glomerulosclerosis, Focal Segmental/immunology
- Glomerulosclerosis, Focal Segmental/pathology
- Lupus Nephritis/blood
- Lupus Nephritis/immunology
- Lupus Nephritis/pathology
- Membrane Proteins/immunology
- Nephrosis, Lipoid/blood
- Nephrosis, Lipoid/immunology
- Nephrosis, Lipoid/pathology
- Nephrotic Syndrome/blood
- Nephrotic Syndrome/immunology
- Nephrotic Syndrome/pathology
- Podocytes/immunology
- Podocytes/pathology
Collapse
Affiliation(s)
- Felicitas E Hengel
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Silke Dehde
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Moritz Lassé
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Gunther Zahner
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Larissa Seifert
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Annabel Schnarre
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Oliver Kretz
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Fatih Demir
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Hans O Pinnschmidt
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Florian Grahammer
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Renke Lucas
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Lea Maxima Mehner
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Tom Zimmermann
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Anja M Billing
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Jun Oh
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Adele Mitrotti
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Paola Pontrelli
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Hanna Debiec
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Claire Dossier
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Manuela Colucci
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Francesco Emma
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - William E Smoyer
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Astrid Weins
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Franz Schaefer
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Nada Alachkar
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Anke Diemert
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Julien Hogan
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Elion Hoxha
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Thorsten Wiech
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Markus M Rinschen
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Pierre Ronco
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Marina Vivarelli
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Loreto Gesualdo
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Nicola M Tomas
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| | - Tobias B Huber
- From the III. Department of Medicine (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., E.H., M.M.R., N.M.T., T.B.H.) and the Departments of Medical Biometry and Epidemiology (H.O.P.), Pathology (T.Z., T.W.), Pediatric Nephrology (J.O.), and Obstetrics and Fetal Medicine (A.D.), Hamburg Center for Kidney Health (F.E.H., S.D., M.L., G.Z., L.S., A.S., O.K., F.G., R.L., L.M.M., J.O., E.H., M.M.R., N.M.T., T.B.H.), and the Hamburg Center for Translational Immunology (N.M.T., T.B.H.), University Medical Center Hamburg-Eppendorf, Hamburg, and the Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg (F.S.) - all in Germany; the Department of Biomedicine, Aarhus University, Aarhus, Denmark (F.D., A.M.B., M.M.R.); the Department of Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis, and Transplantation Unit, University of Bari Aldo Moro, Bari (A.M., P.P., L.G.), and the Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome (M.C., F.E., M.V.) - both in Italy; INSERM, Unité Mixte de Recherche S 1155, Sorbonne Université (H.D., P.R.), and the Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris (C.D., J.H.), Paris, and the Division of Nephrology, Centre Hospitalier du Mans, Le Mans (P.R.) - all in France; the Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, and the Department of Pediatrics, Ohio State University College of Medicine - both in Columbus (W.E.S.); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School - both in Boston (A.W.); and the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore (N.A.)
| |
Collapse
|
18
|
Wang C, Zhang MZ, Li LX, Yun XY, Chen CM, Wang FF, Li B. Ultra-low-dose rituximab is effective for the treatment of patients with minimal change disease - A retrospective study. J Formos Med Assoc 2024:S0929-6646(24)00289-4. [PMID: 38906733 DOI: 10.1016/j.jfma.2024.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 06/04/2024] [Accepted: 06/14/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND PURPOSE Rituximab (RTX),an anti-CD20 monoclonal antibody can effectively treat minimal change disease (MCD),with low toxicity and a reduced steroid dosage. The optimal dosage of RTX for treating MCD remains unclear. This study aimed to investigate the efficacy of an ultra-low-dose regimen of RTX (100 mg per week for 4 weeks) for treating MCD. METHODS We retrospectively analyzed clinical data from 31 patients with MCD who received RTX. Seventeen patients received ultra-low-dose RTX (ULD-RTX) therapy, and 14 patients received standard-dose RTX (SD-RTX) therapy (500 mg weekly for 4 weeks). All patients were followed up for at least 6 months. RESULTS Both groups showed significant increases in the serum albumin levels and notable decreases in the urinary protein levels in the 1st and 6th months after RTX therapy. There were no significant differences in the plasma albumin or urinary protein levels between the groups (p > 0.05). B-cell depletion was observed in all patients after 1 month of RTX administration. At 6 months after RTX treatment, the remission rate was 93% in the SD-RTX group and 88% in the ULD-RTX group (p > 0.05). The ULD-RTX therapy incurred lower costs than did the SD-RTX therapy. One patient in the SD-RTX group developed community-acquired pneumonia. CONCLUSION Ultra-low-dose RTX is effective at inducing remission in patients with MCD at a lower cost.
Collapse
Affiliation(s)
- Chang Wang
- Department of Nephrology, Institute of Nephrology, 2nd Affiliated Hospital of Hainan Medical University, Haikou, China; Hainan Clinical Research Center for Urinary System Disease, China; National Health Commission Key Laboratory of Tropical Diseases Prevention and Control, China; Department of Nephrology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Man-Zhu Zhang
- Department of Nephrology, Institute of Nephrology, 2nd Affiliated Hospital of Hainan Medical University, Haikou, China; Hainan Clinical Research Center for Urinary System Disease, China; National Health Commission Key Laboratory of Tropical Diseases Prevention and Control, China
| | - Ling-Xu Li
- Department of Nephrology, Institute of Nephrology, 2nd Affiliated Hospital of Hainan Medical University, Haikou, China; Hainan Clinical Research Center for Urinary System Disease, China; National Health Commission Key Laboratory of Tropical Diseases Prevention and Control, China
| | - Xiao-Ying Yun
- Department of Nephrology, Institute of Nephrology, 2nd Affiliated Hospital of Hainan Medical University, Haikou, China; Hainan Clinical Research Center for Urinary System Disease, China; National Health Commission Key Laboratory of Tropical Diseases Prevention and Control, China
| | - Chun-Miao Chen
- Department of Nephrology, Institute of Nephrology, 2nd Affiliated Hospital of Hainan Medical University, Haikou, China; Hainan Clinical Research Center for Urinary System Disease, China; National Health Commission Key Laboratory of Tropical Diseases Prevention and Control, China
| | - Fei-Fei Wang
- Department of Nephrology, Institute of Nephrology, 2nd Affiliated Hospital of Hainan Medical University, Haikou, China; Hainan Clinical Research Center for Urinary System Disease, China; National Health Commission Key Laboratory of Tropical Diseases Prevention and Control, China
| | - Bing Li
- Department of Nephrology, Institute of Nephrology, 2nd Affiliated Hospital of Hainan Medical University, Haikou, China; Hainan Clinical Research Center for Urinary System Disease, China; National Health Commission Key Laboratory of Tropical Diseases Prevention and Control, China; Department of Nephrology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
19
|
Iimori M, Sonomura K, Ueyama Y, Oobayashi Y, Adachi H, Nakayama M. Minimal Change Nephrotic Syndrome Secondary to Methotrexate-associated Hodgkin Lymphoma. Intern Med 2024; 63:1771-1776. [PMID: 37926543 PMCID: PMC11239266 DOI: 10.2169/internalmedicine.2572-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/20/2023] [Indexed: 11/07/2023] Open
Abstract
The patient was a 64-year-old man who had been receiving methotrexate (MTX) for rheumatoid arthritis for 8 years. Computed tomography (CT) obtained one month prior to admission revealed numerous enlarged lymph nodes. Lower leg edema appeared two weeks prior to admission. Severe proteinuria was confirmed, and the patient was admitted. A renal biopsy revealed minimal changes in glomeruli. CT on day 14 revealed shrinking lymph nodes, and the patient was diagnosed with Hodgkin lymphoma by a neck lymph node biopsy. This is the first report of secondary minimal change nephrotic syndrome caused by an MTX-associated lymphoproliferative disorder.
Collapse
Affiliation(s)
- Misa Iimori
- Department of Nephrology, Japanese Red Cross Kyoto Daiichi Hospital, Japan
| | | | - Yuichi Ueyama
- Department of Nephrology, Japanese Red Cross Kyoto Daiichi Hospital, Japan
| | - Yuki Oobayashi
- Department of Nephrology, Japanese Red Cross Kyoto Daiichi Hospital, Japan
| | - Hiroya Adachi
- Department of Nephrology, Matsushita Memorial Hospital, Japan
| | - Mayuka Nakayama
- Department of Nephrology, Japanese Red Cross Kyoto Daiichi Hospital, Japan
| |
Collapse
|
20
|
Sun Y, Li Z, Sun J, Zhang S, Wang R, Chen B. The efficacy and safety of rituximab with or without glucocorticoid in inducing remission of MCD with different clinical presentations in adults: a retrospective study. Clin Kidney J 2024; 17:sfae139. [PMID: 38854425 PMCID: PMC11161702 DOI: 10.1093/ckj/sfae139] [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: 10/19/2023] [Indexed: 06/11/2024] Open
Abstract
Background To investigate the efficacy and safety of rituximab (RTX) with or without glucocorticoid (GC) in inducing remission of minimal change disease (MCD) in adults. Methods Twenty-one adult MCD patients were included in the study. The patients were assigned to the following three groups according to their background before RTX treatment: an RTX single drug direct induction treatment group (Group A; n = 9), a short-term, low-dose GC combined with RTX induction treatment group (Group B; n = 4), and a short-term, adequate-dose GC-induced remission and RTX maintenance treatment group (Group C; n = 8). The primary endpoints were the time to induction of remission and the rate of clinical remission at 12 months. Results All patients achieved clinical remission, with 19 (90.48%) achieving complete remission (CR), and the median remission time was 4 (2.5, 12) weeks. Eight (88.89%) patients in Group A achieved CR, and the median remission time was 3 (2.25, 14) weeks. In Group B, three (75.00%) patients achieved CR, with a median remission time of 4 (4, 10) weeks. In Group C, eight (100.00%) patients achieved CR, and the median remission time was 3.5 (2, 4) weeks. Conclusions In MCD patients without acute kidney injury, adequate RTX alone or short-term combined treatment with low-dose GCs can effectively induce and maintain MCD remission. Adequate short-term GCs combined with RTX maintenance may be an effective alternative for MCD patients in context of acute kidney injury. There is a need to investigate different induction therapy regimens for the remission of MCD patients with different backgrounds.
Collapse
Affiliation(s)
- Yujiao Sun
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhuo Li
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jing Sun
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shasha Zhang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Rong Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Bing Chen
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
21
|
Bărar AA, Pralea IE, Maslyennikov Y, Munteanu R, Berindan-Neagoe I, Pîrlog R, Rusu I, Nuțu A, Rusu CC, Moldovan DT, Potra AR, Tirinescu D, Ticala M, Elec FI, Iuga CA, Kacso IM. Minimal Change Disease: Pathogenetic Insights from Glomerular Proteomics. Int J Mol Sci 2024; 25:5613. [PMID: 38891801 PMCID: PMC11171934 DOI: 10.3390/ijms25115613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024] Open
Abstract
The mechanism underlying podocyte dysfunction in minimal change disease (MCD) remains unknown. This study aimed to shed light on the potential pathophysiology of MCD using glomerular proteomic analysis. Shotgun proteomics using label-free quantitative mass spectrometry was performed on formalin-fixed, paraffin-embedded (FFPE) renal biopsies from two groups of samples: control (CTR) and MCD. Glomeruli were excised from FFPE renal biopsies using laser capture microdissection (LCM), and a single-pot solid-phase-enhanced sample preparation (SP3) digestion method was used to improve yield and protein identifications. Principal component analysis (PCA) revealed a distinct separation between the CTR and MCD groups. Forty-eight proteins with different abundance between the two groups (p-value ≤ 0.05 and |FC| ≥ 1.5) were identified. These may represent differences in podocyte structure, as well as changes in endothelial or mesangial cells and extracellular matrix, and some were indeed found in several of these structures. However, most differentially expressed proteins were linked to the podocyte cytoskeleton and its dynamics. Some of these proteins are known to be involved in focal adhesion (NID1 and ITGA3) or slit diaphragm signaling (ANXA2, TJP1 and MYO1C), while others are structural components of the actin and microtubule cytoskeleton of podocytes (ACTR3 and NES). This study suggests the potential of mass spectrometry-based shotgun proteomic analysis with LCM glomeruli to yield valuable insights into the pathogenesis of podocytopathies like MCD. The most significantly dysregulated proteins in MCD could be attributable to cytoskeleton dysfunction or may be a compensatory response to cytoskeleton malfunction caused by various triggers.
Collapse
Affiliation(s)
- Andrada Alina Bărar
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.A.B.); (Y.M.); (C.C.R.); (D.T.M.); (A.R.P.); (D.T.); (M.T.); (I.M.K.)
| | - Ioana-Ecaterina Pralea
- Department of Proteomics and Metabolomics, Research Center for Advanced Medicine–MedFuture, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, Louis Pasteur Street 4-6, 400349 Cluj-Napoca, Romania;
| | - Yuriy Maslyennikov
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.A.B.); (Y.M.); (C.C.R.); (D.T.M.); (A.R.P.); (D.T.); (M.T.); (I.M.K.)
| | - Raluca Munteanu
- Department of In Vivo Studies, Research Center for Advanced Medicine–MedFuture, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, Louis Pasteur Street 6, 400349 Cluj-Napoca, Romania;
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (I.B.-N.); (R.P.); (A.N.)
| | - Radu Pîrlog
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (I.B.-N.); (R.P.); (A.N.)
| | - Ioana Rusu
- Department of Pathology, Regional Institute of Gastroenterology and Hepatology, 400394 Cluj-Napoca, Romania;
| | - Andreea Nuțu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (I.B.-N.); (R.P.); (A.N.)
| | - Crina Claudia Rusu
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.A.B.); (Y.M.); (C.C.R.); (D.T.M.); (A.R.P.); (D.T.); (M.T.); (I.M.K.)
| | - Diana Tania Moldovan
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.A.B.); (Y.M.); (C.C.R.); (D.T.M.); (A.R.P.); (D.T.); (M.T.); (I.M.K.)
| | - Alina Ramona Potra
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.A.B.); (Y.M.); (C.C.R.); (D.T.M.); (A.R.P.); (D.T.); (M.T.); (I.M.K.)
| | - Dacian Tirinescu
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.A.B.); (Y.M.); (C.C.R.); (D.T.M.); (A.R.P.); (D.T.); (M.T.); (I.M.K.)
| | - Maria Ticala
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.A.B.); (Y.M.); (C.C.R.); (D.T.M.); (A.R.P.); (D.T.); (M.T.); (I.M.K.)
| | - Florin Ioan Elec
- Department of Urology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Cristina Adela Iuga
- Department of Proteomics and Metabolomics, Research Center for Advanced Medicine–MedFuture, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, Louis Pasteur Street 4-6, 400349 Cluj-Napoca, Romania;
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ina Maria Kacso
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.A.B.); (Y.M.); (C.C.R.); (D.T.M.); (A.R.P.); (D.T.); (M.T.); (I.M.K.)
| |
Collapse
|
22
|
Riganati M, Zotta F, Candino A, Conversano E, Gargiulo A, Scarsella M, Lo Russo A, Bettini C, Emma F, Vivarelli M, Colucci M. A novel flow cytometry panel to identify prognostic markers for steroid-sensitive forms of idiopathic nephrotic syndrome in childhood. Front Immunol 2024; 15:1379924. [PMID: 38629076 PMCID: PMC11018947 DOI: 10.3389/fimmu.2024.1379924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/14/2024] [Indexed: 04/19/2024] Open
Abstract
Introduction The clinical evolution of steroid-sensitive forms of pediatric idiopathic nephrotic syndrome (INS) is highly heterogeneous following the standard treatment with prednisone. To date, no prognostic marker has been identified to predict the severity of the disease course starting from the first episode. Methods In this monocentric prospective cohort study we set up a reproducible and standardized flow cytometry panel using two sample tubes (one for B-cell and one for T-cell subsets) to extensively characterized the lymphocyte repertoire of INS pediatric patients. A total of 44 children with INS at disease onset were enrolled, sampled before and 3 months after standard induction therapy with prednisone and followed for 12 months to correctly classify their disease based on relapses. Age-matched controls with non immune-mediated renal diseases or with urological disorders were also enrolled. Demographical, clinical, laboratory and immunosuppressive treatment data were registered. Results We found that children with INS at disease onset had significantly higher circulating levels of total CD19+ and specific B-cell subsets (transitional, mature-naïve, plasmablasts/plasmacells, CD19+CD27+, unswitched, switched and atypical memory B cells) and reduced circulating levels of Tregs, when compared to age-matched controls. Prednisone therapy restored most B- and T-cell alterations. When patients were subdivided based on disease relapse, relapsing patients had significantly more transitional, CD19+CD27+ memory and in particular unswitched memory B cells at disease onset, which were predictive of a higher risk of relapse in steroid-sensitive patients by logistic regression analysis, irrespective of age. In accordance, B-cell dysregulations resulted mainly associated with steroid-dependence when patients were stratified in different disease severity forms. Of note, Treg levels were reduced independently from the disease subgroup and were not completely normalized by prednisone treatment. Conclusion We have set up a novel, reproducible, disease-specific flow cytometry panel that allows a comprehensive characterization of circulating lymphocytes. We found that, at disease onset, relapsing patients had significantly more transitional, CD19+CD27+ memory and unswitched memory B cells and those who are at higher risk of relapse had increased circulating levels of unswitched memory B cells, independently of age. This approach can allow prediction of clinical evolution, monitoring of immunosuppression and tailored treatment in different forms of INS.
Collapse
Affiliation(s)
- Martina Riganati
- Laboratory of Nephrology, Bambino Gesù Children’s Hospital Scientific Institute for Research and Health Care (IRCCS), Rome, Italy
| | - Federica Zotta
- Division of Nephrology, Bambino Gesù Children’s Hospital Scientific Institute for Research and Health Care (IRCCS), Rome, Italy
| | - Annalisa Candino
- Division of Nephrology, Bambino Gesù Children’s Hospital Scientific Institute for Research and Health Care (IRCCS), Rome, Italy
| | - Ester Conversano
- Division of Nephrology, Bambino Gesù Children’s Hospital Scientific Institute for Research and Health Care (IRCCS), Rome, Italy
| | - Antonio Gargiulo
- Division of Nephrology, Bambino Gesù Children’s Hospital Scientific Institute for Research and Health Care (IRCCS), Rome, Italy
| | - Marco Scarsella
- Research Laboratories, Bambino Gesù Children’s Hospital Scientific Institute for Research and Health Care (IRCCS), Rome, Italy
| | - Anna Lo Russo
- Research Laboratories, Bambino Gesù Children’s Hospital Scientific Institute for Research and Health Care (IRCCS), Rome, Italy
| | - Chiara Bettini
- Division of Nephrology, Bambino Gesù Children’s Hospital Scientific Institute for Research and Health Care (IRCCS), Rome, Italy
| | - Francesco Emma
- Laboratory of Nephrology, Bambino Gesù Children’s Hospital Scientific Institute for Research and Health Care (IRCCS), Rome, Italy
- Division of Nephrology, Bambino Gesù Children’s Hospital Scientific Institute for Research and Health Care (IRCCS), Rome, Italy
| | - Marina Vivarelli
- Laboratory of Nephrology, Bambino Gesù Children’s Hospital Scientific Institute for Research and Health Care (IRCCS), Rome, Italy
- Division of Nephrology, Bambino Gesù Children’s Hospital Scientific Institute for Research and Health Care (IRCCS), Rome, Italy
| | - Manuela Colucci
- Laboratory of Nephrology, Bambino Gesù Children’s Hospital Scientific Institute for Research and Health Care (IRCCS), Rome, Italy
| |
Collapse
|
23
|
Zhang PL, Mahalingam VD, Metcalf BD, Al-Othman YA, Li W, Kanaan HD, Herrera GA. Punctate IgG staining particles localize in the budding ballooning clusters of reactive foot processes in minimal change disease. Ultrastruct Pathol 2024; 48:121-127. [PMID: 38098281 DOI: 10.1080/01913123.2023.2292590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/05/2023] [Indexed: 02/08/2024]
Abstract
The etiology of minimal change disease (MCD) remains a mystery as the only characteristic findings are the diffuse effacement of foot processes seen on electron microscopy (EM). Punctate IgG staining found floating outside glomerular capillary loops in MCD cases was recently identified as autoimmune antibodies against nephrin of podocytes. We hypothesized that the punctate IgG staining is located on budding ballooning clusters (BBC) of reactive foot processes in Bowman's space found on EM. We identified seven patients with MCD cases showing IgG staining that were subsequently evaluated for BBC on EM. We concurrently examined 12 negative controls, either unremarkable cases or tubulointerstitial diseases, by EM. Immunogold labeling was performed to confirm the presence of IgG and determine localization. In seven MCD cases, there were positive punctate IgG staining particles outside of the glomerular basement membranes (GBM) along with concurrent punctate staining for C3, kappa, and lambda. By EM, all seven (100%) MCD cases revealed BBC that was characterized by ballooning foot processes ranging from 1 to 6 µm and was either budding or detached from the GBM in 3-7 clusters; no electron-dense materials were seen in BBC. BBC was also seen in only 1 of 12 (8%) negative controls. Immunogold labeling identified IgG particles within BBC of MCD by EM, but not in the negative control. Our data suggest that BBC are EM structures of reactive foot processes that are most likely correlated with punctate IgG staining seen in cases of MCD, supported by immunogold labeling for IgG.
Collapse
Affiliation(s)
- Ping L Zhang
- Department of Pathology, Corewell Health East, Royal Oak, MI, USA
| | | | | | | | - Wei Li
- Department of Pathology, Corewell Health East, Royal Oak, MI, USA
| | - Hassan D Kanaan
- Department of Pathology, Corewell Health East, Royal Oak, MI, USA
| | | |
Collapse
|
24
|
Srivastava T, Garola RE, Zhou J, Boinpelly VC, Rezaiekhaligh MH, Joshi T, Jiang Y, Ebadi D, Sharma S, Sethna C, Staggs VS, Sharma R, Gipson DS, Hao W, Wang Y, Mariani LH, Hodgin JB, Rottapel R, Yoshitaka T, Ueki Y, Sharma M. Scaffold protein SH3BP2 signalosome is pivotal for immune activation in nephrotic syndrome. JCI Insight 2024; 9:e170055. [PMID: 38127456 PMCID: PMC10967477 DOI: 10.1172/jci.insight.170055] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 12/19/2023] [Indexed: 12/23/2023] Open
Abstract
Despite clinical use of immunosuppressive agents, the immunopathogenesis of minimal change disease (MCD) and focal segmental glomerulosclerosis (FSGS) remains unclear. Src homology 3-binding protein 2 (SH3BP2), a scaffold protein, forms an immune signaling complex (signalosome) with 17 other proteins, including phospholipase Cγ2 (PLCγ2) and Rho-guanine nucleotide exchange factor VAV2 (VAV2). Bioinformatic analysis of human glomerular transcriptome (Nephrotic Syndrome Study Network cohort) revealed upregulated SH3BP2 in MCD and FSGS. The SH3BP2 signalosome score and downstream MyD88, TRIF, and NFATc1 were significantly upregulated in MCD and FSGS. Immune pathway activation scores for Toll-like receptors, cytokine-cytokine receptor, and NOD-like receptors were increased in FSGS. Lower SH3BP2 signalosome score was associated with MCD, higher estimated glomerular filtration rate, and remission. Further work using Sh3bp2KI/KI transgenic mice with a gain-in-function mutation showed ~6-fold and ~25-fold increases in albuminuria at 4 and 12 weeks, respectively. Decreased serum albumin and unchanged serum creatinine were observed at 12 weeks. Sh3bp2KI/KI kidney morphology appeared normal except for increased mesangial cellularity and patchy foot process fusion without electron-dense deposits. SH3BP2 co-immunoprecipitated with PLCγ2 and VAV2 in human podocytes, underscoring the importance of SH3BP2 in immune activation. SH3BP2 and its binding partners may determine the immune activation pathways resulting in podocyte injury leading to loss of the glomerular filtration barrier.
Collapse
Affiliation(s)
- Tarak Srivastava
- Section of Nephrology, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
- Midwest Veterans’ Biomedical Research Foundation, Kansas City, Missouri, USA
- Department of Oral and Craniofacial Sciences, University of Missouri at Kansas City School of Dentistry, Kansas City, Missouri, USA
| | - Robert E. Garola
- Department of Pathology and Laboratory Medicine, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Jianping Zhou
- Midwest Veterans’ Biomedical Research Foundation, Kansas City, Missouri, USA
- Kansas City VA Medical Center, Kansas City, Missouri, USA
| | - Varun C. Boinpelly
- Midwest Veterans’ Biomedical Research Foundation, Kansas City, Missouri, USA
- Kansas City VA Medical Center, Kansas City, Missouri, USA
| | - Mohammad H. Rezaiekhaligh
- Section of Nephrology, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Trupti Joshi
- Department of Health Management and Informatics
- Department of Electrical Engineering and Computer Science
- Christopher S. Bond Life Sciences Center, and
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, Missouri, USA
| | - Yuexu Jiang
- Department of Electrical Engineering and Computer Science
- Christopher S. Bond Life Sciences Center, and
| | - Diba Ebadi
- The Ottawa Hospital Rehabilitation Centre, Ottawa, Ontario, Canada
| | - Siddarth Sharma
- Milken Institute School of Public Health, George Washington University, Washington, DC, USA
| | - Christine Sethna
- Cohen Children’s Medical Center of NY, New Hyde Park, New York, USA
| | - Vincent S. Staggs
- Biostatistics and Epidemiology Core, Children’s Mercy Research Institute and Department of Pediatrics, University of Missouri, Kansas City, Missouri, USA
| | - Ram Sharma
- Kansas City VA Medical Center, Kansas City, Missouri, USA
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Debbie S. Gipson
- Division of Nephrology, Department of Internal Medicine, School of Medicine, and
| | - Wei Hao
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Yujie Wang
- Division of Nephrology, Department of Internal Medicine, School of Medicine, and
| | - Laura H. Mariani
- Division of Nephrology, Department of Internal Medicine, School of Medicine, and
| | - Jeffrey B. Hodgin
- Division of Nephrology, Department of Internal Medicine, School of Medicine, and
| | - Robert Rottapel
- Princess Margaret Cancer Center, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Teruhito Yoshitaka
- Department of Orthopedic Surgery, Hiroshima City Rehabilitation Hospital, Hiroshima, Hiroshima, Japan
| | - Yasuyoshi Ueki
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, Indiana, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Mukut Sharma
- Midwest Veterans’ Biomedical Research Foundation, Kansas City, Missouri, USA
- Kansas City VA Medical Center, Kansas City, Missouri, USA
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
25
|
Teragaki M, Tanaka M, Yamamoto H, Watanabe T, Takeoka J, Fukumi A, Maeda K, Takami Y, Saita H, Iwanari S, Ikeda M, Takeoka H. Relapse of minimal change disease following the third mRNA COVID-19 vaccination: a case report and literature review. CEN Case Rep 2024; 13:53-58. [PMID: 37244881 PMCID: PMC10224756 DOI: 10.1007/s13730-023-00798-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 05/12/2023] [Indexed: 05/29/2023] Open
Abstract
Mass vaccination is the most important strategy to terminate the coronavirus disease 2019 (COVID-19) pandemic. Reports suggest the potential risk of the development of new-onset or relapse of minimal change disease (MCD) following COVID-19 vaccination; however, details on vaccine-associated MCD remain unclear. A 43-year-old man with MCD, who had been in remission for 29 years, developed nephrotic syndrome 4 days after receiving the third dose of the Pfizer-BioNTech vaccine. His kidney biopsy revealed relapsing MCD. Intravenous methylprednisolone pulse therapy followed by oral prednisolone therapy was administered, and his proteinuria resolved within 3 weeks. This report highlights the importance of careful monitoring of proteinuria after COVID-19 vaccination in patients with MCD, even if the disease is stable and no adverse events occurred during previous vaccinations. Our case report and literature review of COVID-19 vaccine-associated MCD indicated that MCD relapse tends to occur later after vaccination and slightly more often following the second and subsequent vaccine doses than new-onset MCD.
Collapse
Affiliation(s)
- Mariko Teragaki
- Department of Nephrology and Dialysis, Hyogo Prefectural Amagasaki General Medical Center, 2-17-77 Higashinaniwa-Cho, Amagasaki, Hyogo, 660-8550, Japan.
| | - Mari Tanaka
- Department of Nephrology and Dialysis, Hyogo Prefectural Amagasaki General Medical Center, 2-17-77 Higashinaniwa-Cho, Amagasaki, Hyogo, 660-8550, Japan
| | - Hiroko Yamamoto
- Department of Nephrology and Dialysis, Hyogo Prefectural Amagasaki General Medical Center, 2-17-77 Higashinaniwa-Cho, Amagasaki, Hyogo, 660-8550, Japan
| | - Tomoka Watanabe
- Department of Nephrology and Dialysis, Hyogo Prefectural Amagasaki General Medical Center, 2-17-77 Higashinaniwa-Cho, Amagasaki, Hyogo, 660-8550, Japan
| | - Jun Takeoka
- Department of Nephrology and Dialysis, Hyogo Prefectural Amagasaki General Medical Center, 2-17-77 Higashinaniwa-Cho, Amagasaki, Hyogo, 660-8550, Japan
| | - Awaisshafig Fukumi
- Department of Nephrology and Dialysis, Hyogo Prefectural Amagasaki General Medical Center, 2-17-77 Higashinaniwa-Cho, Amagasaki, Hyogo, 660-8550, Japan
| | - Kotaro Maeda
- Department of Nephrology and Dialysis, Hyogo Prefectural Amagasaki General Medical Center, 2-17-77 Higashinaniwa-Cho, Amagasaki, Hyogo, 660-8550, Japan
| | - Yohtaro Takami
- Department of Nephrology and Dialysis, Hyogo Prefectural Amagasaki General Medical Center, 2-17-77 Higashinaniwa-Cho, Amagasaki, Hyogo, 660-8550, Japan
| | - Hirona Saita
- Department of Nephrology and Dialysis, Hyogo Prefectural Amagasaki General Medical Center, 2-17-77 Higashinaniwa-Cho, Amagasaki, Hyogo, 660-8550, Japan
| | - Sachio Iwanari
- Department of Nephrology and Dialysis, Hyogo Prefectural Amagasaki General Medical Center, 2-17-77 Higashinaniwa-Cho, Amagasaki, Hyogo, 660-8550, Japan
| | - Masaki Ikeda
- Department of Nephrology and Dialysis, Hyogo Prefectural Amagasaki General Medical Center, 2-17-77 Higashinaniwa-Cho, Amagasaki, Hyogo, 660-8550, Japan
| | - Hiroya Takeoka
- Department of Nephrology and Dialysis, Hyogo Prefectural Amagasaki General Medical Center, 2-17-77 Higashinaniwa-Cho, Amagasaki, Hyogo, 660-8550, Japan
| |
Collapse
|
26
|
Jiang H, Shen Z, Zhuang J, Lu C, Qu Y, Xu C, Yang S, Tian X. Understanding the podocyte immune responses in proteinuric kidney diseases: from pathogenesis to therapy. Front Immunol 2024; 14:1335936. [PMID: 38288116 PMCID: PMC10822972 DOI: 10.3389/fimmu.2023.1335936] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 12/29/2023] [Indexed: 01/31/2024] Open
Abstract
The glomerular filtration barrier, comprising the inner layer of capillary fenestrated endothelial cells, outermost podocytes, and the glomerular basement membrane between them, plays a pivotal role in kidney function. Podocytes, terminally differentiated epithelial cells, are challenging to regenerate once injured. They are essential for maintaining the integrity of the glomerular filtration barrier. Damage to podocytes, resulting from intrinsic or extrinsic factors, leads to proteinuria in the early stages and eventually progresses to chronic kidney disease (CKD). Immune-mediated podocyte injury is a primary pathogenic mechanism in proteinuric glomerular diseases, including minimal change disease, focal segmental glomerulosclerosis, membranous nephropathy, and lupus nephritis with podocyte involvement. An extensive body of evidence indicates that podocytes not only contribute significantly to the maintenance of the glomerular filtration barrier and serve as targets of immune responses but also exhibit immune cell-like characteristics, participating in both innate and adaptive immunity. They play a pivotal role in mediating glomerular injury and represent potential therapeutic targets for CKD. This review aims to systematically elucidate the mechanisms of podocyte immune injury in various podocyte lesions and provide an overview of recent advances in podocyte immunotherapy. It offers valuable insights for a deeper understanding of the role of podocytes in proteinuric glomerular diseases, and the identification of new therapeutic targets, and has significant implications for the future clinical diagnosis and treatment of podocyte-related disorders.
Collapse
Affiliation(s)
- Hong Jiang
- Division of Nephrology, Department of Internal Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Zhirang Shen
- Division of Nephrology, Department of Internal Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Jing Zhuang
- Division of Nephrology, Department of Internal Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Chen Lu
- Division of Nephrology, Department of Internal Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yue Qu
- Division of Nephrology, Department of Internal Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Chengren Xu
- Division of Nephrology, Department of Internal Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Shufen Yang
- Division of Nephrology, Department of Internal Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Xuefei Tian
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
27
|
Chugh SS, Clement LC. "Idiopathic" minimal change nephrotic syndrome: a podocyte mystery nears the end. Am J Physiol Renal Physiol 2023; 325:F685-F694. [PMID: 37795536 PMCID: PMC10878723 DOI: 10.1152/ajprenal.00219.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/11/2023] [Accepted: 10/02/2023] [Indexed: 10/06/2023] Open
Abstract
The discovery of zinc fingers and homeoboxes (ZHX) transcriptional factors and the upregulation of hyposialylated angiopoietin-like 4 (ANGPTL4) in podocytes have been crucial in explaining the cardinal manifestations of human minimal change nephrotic syndrome (MCNS). Recently, uncovered genomic defects upstream of ZHX2 induce a ZHX2 hypomorph state that makes podocytes inherently susceptible to mild cytokine storms resulting from a common cold. In ZHX2 hypomorph podocytes, ZHX proteins are redistributed away from normal transmembrane partners like aminopeptidase A (APA) toward alternative binding partners like IL-4Rα. During disease relapse, high plasma soluble IL-4Rα (sIL-4Rα) associated with chronic atopy complements the cytokine milieu of a common cold to displace ZHX1 from podocyte transmembrane IL-4Rα toward the podocyte nucleus. Nuclear ZHX1 induces severe upregulation of ANGPTL4, resulting in incomplete sialylation of part of the ANGPTL4 protein, secretion of hyposialylated ANGPTL4, and hyposialylation-related injury in the glomerulus. This pattern of injury induces many of the classic manifestations of human minimal change disease (MCD), including massive and selective proteinuria, podocyte foot process effacement, and loss of glomerular basement membrane charge. Administration of glucocorticoids reduces ANGPTL4 upregulation, which reduces hyposialylation injury to improve the clinical phenotype. Improving sialylation of podocyte-secreted ANGPTL4 also reduces proteinuria and improves experimental MCD. Neutralizing circulating TNF-α, IL-6, or sIL-4Rα after the induction of the cytokine storm in Zhx2 hypomorph mice reduces albuminuria, suggesting potential new therapeutic targets for clinical trials to prevent MCD relapse. These studies collectively lay to rest prior suggestions of a role of single cytokines or soluble proteins in triggering MCD relapse.
Collapse
Affiliation(s)
- Sumant S Chugh
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, United States
| | - Lionel C Clement
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, United States
| |
Collapse
|
28
|
Veltkamp F, Thenot V, Mussies C, van Lieshout B, Peters-Sengers H, Kers J, Khan DH, Hogan J, Florquin S, Bouts AHM, Dossier C. Incidence of idiopathic nephrotic syndrome during the Covid-19 pandemic in the Paris area (France) and in the Netherlands. Pediatr Nephrol 2023; 38:3681-3692. [PMID: 37191940 PMCID: PMC10186275 DOI: 10.1007/s00467-023-06006-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 04/21/2023] [Accepted: 04/21/2023] [Indexed: 05/17/2023]
Abstract
BACKGROUND The aetiology of idiopathic nephrotic syndrome (INS) remains partially unknown. Viral infections have been associated with INS onset. Since we observed fewer first onset INS cases during the Covid-19 pandemic, we hypothesised that lower INS incidence was the result of lockdown measures. Therefore, the aim of this study was to evaluate the incidence of childhood INS before and during the COVID-19 pandemic in two independent European INS cohorts. METHODS Children with new INS in the Netherlands (2018-2021) and Paris area (2018-2021) were included. We estimated incidences using census data for each region. Incidences were compared using two proportion Z-tests. RESULTS A total of 128 and 324 cases of first onset INS were reported in the Netherlands and Paris area, respectively, corresponding to an annual incidence of 1.21 and 2.58 per 100,000 children/year. Boys and young children (< 7 years) were more frequently affected. Incidence before and during the pandemic did not differ. When schools were closed, incidence was lower in both regions: 0.53 vs. 1.31 (p = 0.017) in the Netherlands and 0.94 vs. 2.63 (p = 0.049) in the Paris area. During peaks of hospital admissions for Covid-19, no cases were reported in the Netherlands or Paris area. CONCLUSIONS Incidence of INS before and during the Covid-19 pandemic was not different, but when schools were closed during lockdown, incidence was significantly lower. Interestingly, incidences of other respiratory viral infections were also reduced as was air pollution. Together, these results argue for a link between INS onset and viral infections and/or environmental factors. A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
Affiliation(s)
- Floor Veltkamp
- Department of Pediatric Nephrology, Emma Children's Hospital, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam, 1109 AZ, The Netherlands.
| | - Victoire Thenot
- Department of Pediatric Nephrology, Robert-Debré Hospital, APHP, Paris, France
| | - Carlijn Mussies
- Department of Pediatric Nephrology, Emma Children's Hospital, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam, 1109 AZ, The Netherlands
| | - Bas van Lieshout
- Department of Pediatric Nephrology, Emma Children's Hospital, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam, 1109 AZ, The Netherlands
| | - Hessel Peters-Sengers
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Department of Epidemiology and Data Science, VU University Amsterdam, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Jesper Kers
- Department of Pathology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
- Van 't Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Djera H Khan
- Department of Epidemiology and Data Science, VU University Amsterdam, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Julien Hogan
- Department of Pediatric Nephrology, Robert-Debré Hospital, APHP, Paris, France
| | - Sandrine Florquin
- Department of Epidemiology and Data Science, VU University Amsterdam, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Antonia H M Bouts
- Department of Pediatric Nephrology, Emma Children's Hospital, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam, 1109 AZ, The Netherlands
| | - Claire Dossier
- Department of Pediatric Nephrology, Robert-Debré Hospital, APHP, Paris, France
| |
Collapse
|
29
|
Khan GH, Veltkamp F, Scheper M, Hoebe RA, Claessen N, Butter L, Bouts AHM, Florquin S, Guikema JEJ. Levamisole suppresses activation and proliferation of human T cells by the induction of a p53-dependent DNA damage response. Eur J Immunol 2023; 53:e2350562. [PMID: 37597325 DOI: 10.1002/eji.202350562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/17/2023] [Accepted: 08/18/2023] [Indexed: 08/21/2023]
Abstract
Levamisole (LMS) is a small molecule used in the treatment of idiopathic nephrotic syndrome (INS). The pathogenesis of INS remains unknown, but evidence points toward an immunological basis of the disease. Recently, LMS has been shown to increase the relapse-free survival in INS patients. While LMS has been hypothesized to exert an immunomodulatory effect, its mechanism of action remains unknown. Here, we show that LMS decreased activation and proliferation of human T cells. T-cell activation-associated cytokines such as IL-2, TNF-α, and IFN-γ were reduced upon LMS treatment, whereas IL-4 and IL-13 were increased. Gene expression profiling confirmed that the suppressive effects of LMS as genes involved in cell cycle progression were downregulated. Furthermore, genes associated with p53 activation were upregulated by LMS. In agreement, LMS treatment resulted in p53 phosphorylation and increased expression of the p53 target gene FAS. Accordingly, LMS sensitized activated T cells for Fas-mediated apoptosis. LMS treatment resulted in a mid-S phase cell cycle arrest accompanied by γH2AX-foci formation and phosphorylation of CHK1. Our findings indicate that LMS acts as an immunosuppressive drug that directly affects the activation and proliferation of human T cells by induction of DNA damage and the activation of a p53-dependent DNA damage response.
Collapse
Affiliation(s)
- Gerarda H Khan
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Floor Veltkamp
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Pediatric Nephrology, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Mirte Scheper
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ron A Hoebe
- Department of Medical Biology, Amsterdam UMC and Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Nike Claessen
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Loes Butter
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Antonia H M Bouts
- Department of Pediatric Nephrology, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Sandrine Florquin
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jeroen E J Guikema
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Hayward S, Parmesar K, Saleem MA. What is circulating factor disease and how is it currently explained? Pediatr Nephrol 2023; 38:3513-3518. [PMID: 36952039 PMCID: PMC10514121 DOI: 10.1007/s00467-023-05928-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/15/2023] [Accepted: 02/20/2023] [Indexed: 03/24/2023]
Abstract
Nephrotic syndrome (NS) consists of the clinical triad of hypoalbuminaemia, high levels of proteinuria and oedema, and describes a heterogeneous group of disease processes with different underlying drivers. The existence of circulating factor disease (CFD) as a driver of NS has been epitomised by a subset of patients who exhibit disease recurrence after transplantation, alongside laboratory work. Several circulating factors have been proposed and studied, broadly grouped into protease components such as soluble urokinase-type plasminogen activator (suPAR), hemopexin (Hx) and calcium/calmodulin-serine protease kinase (CASK), and other circulating proteases, and immune components such as TNF-α, CD40 and cardiotrophin-like cytokine-1 (CLC-1). While currently there is no definitive way of assessing risk of CFD pre-transplantation, promising work is emerging through the study of 'multi-omic' bioinformatic data from large national cohorts and biobanks.
Collapse
Affiliation(s)
- Samantha Hayward
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
| | - Kevon Parmesar
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Moin A Saleem
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
31
|
Vivarelli M, Colucci M, Algeri M, Zotta F, Emma F, L’Erario I, Busutti M, Rota S, Capelli C, Introna M, Todeschini M, Casiraghi F, Perna A, Peracchi T, De Salvo A, Rubis N, Locatelli F, Remuzzi G, Ruggenenti P. A phase I study of autologous mesenchymal stromal cells for severe steroid-dependent nephrotic syndrome. JCI Insight 2023; 8:e169424. [PMID: 37561590 PMCID: PMC10561718 DOI: 10.1172/jci.insight.169424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 08/08/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUNDSevere forms of idiopathic nephrotic syndrome (INS) require prolonged immunosuppressive therapies and repeated courses of high-dose glucocorticoids. Mesenchymal stromal cells (MSCs) have promising immunomodulatory properties that may be employed therapeutically to reduce patient exposure to medications and their side effects.METHODSWe performed a phase I open-label trial assessing safety and feasibility of autologous bone marrow-derived MSCs (BM-MSCs) in children and young adults with severe forms of steroid-dependent nephrotic syndrome. Following autologous BM-MSC preparation and infusion, oral immunosuppression was tapered. Safety, efficacy, and immunomodulatory effects in vivo were monitored for 12 months.RESULTSSixteen patients (10 children, 6 adults) were treated. Adverse events were limited and not related to BM-MSC infusions. All patients relapsed during follow-up, but in the 10 treated children, time to first relapse was delayed (P = 0.02) and number of relapses was reduced (P = 0.002) after BM-MSC infusion, compared with the previous 12 months. Cumulative prednisone dose was also reduced at 12 months compared with baseline (P < 0.05). No treatment benefit was observed in adults.In children, despite tapering of immunosuppression, clinical benefit was mirrored by a significant reduction in total CD19+, mature, and memory B cells and an increase in regulatory T cells in vivo up to 3-6 months following BM-MSC infusionCONCLUSIONTreatment with autologous BM-MSCs is feasible and safely reduces relapses and immunosuppression at 12 months in children with severe steroid-dependent INS. Immunomodulatory studies suggest that repeating MSC infusions at 3-6 months may sustain benefit.TRIAL REGISTRATIONEudraCT 2016-004804-77.FUNDINGAIFA Ricerca Indipendente 2016-02364623.
Collapse
Affiliation(s)
- Marina Vivarelli
- Division of Nephrology, and
- Laboratory of Nephrology, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Manuela Colucci
- Laboratory of Nephrology, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Mattia Algeri
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | | | | | | | | | - Stefano Rota
- Unit of Nephrology and Dialysis, Azienda Socio Sanitaria Territoriale (ASST), Bergamo, Italy
| | - Chiara Capelli
- Center of Cellular Therapy “G. Lanzani,” Haematology Department, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Martino Introna
- Center of Cellular Therapy “G. Lanzani,” Haematology Department, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Marta Todeschini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | | | - Annalisa Perna
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Tobia Peracchi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Andrea De Salvo
- Psychology Unit, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Nadia Rubis
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Piero Ruggenenti
- Unit of Nephrology and Dialysis, Azienda Socio Sanitaria Territoriale (ASST), Bergamo, Italy
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| |
Collapse
|
32
|
Salfi G, Casiraghi F, Remuzzi G. Current understanding of the molecular mechanisms of circulating permeability factor in focal segmental glomerulosclerosis. Front Immunol 2023; 14:1247606. [PMID: 37795085 PMCID: PMC10546017 DOI: 10.3389/fimmu.2023.1247606] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/05/2023] [Indexed: 10/06/2023] Open
Abstract
The pathogenetic mechanisms underlying the onset and the post-transplant recurrence of primary focal segmental glomerulosclerosis (FSGS) are complex and remain yet to be fully elucidated. However, a growing body of evidence emphasizes the pivotal role of the immune system in both initiating and perpetuating the disease. Extensive investigations, encompassing both experimental models and patient studies, have implicated T cells, B cells, and complement as crucial actors in the pathogenesis of primary FSGS, with various molecules being proposed as potential "circulating factors" contributing to the disease and its recurrence post kidney-transplantation. In this review, we critically assessed the existing literature to identify essential pathways for a comprehensive characterization of the pathogenesis of FSGS. Recent discoveries have shed further light on the intricate interplay between these mechanisms. We present an overview of the current understanding of the engagement of distinct molecules and immune cells in FSGS pathogenesis while highlighting critical knowledge gaps that require attention. A thorough characterization of these intricate immune mechanisms holds the potential to identify noninvasive biomarkers that can accurately identify patients at high risk of post-transplant recurrence. Such knowledge can pave the way for the development of targeted and personalized therapeutic approaches in the management of FSGS.
Collapse
Affiliation(s)
| | - Federica Casiraghi
- Istituto di Ricerche Farmacologiche Mario Negri Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Bergamo, Italy
| | | |
Collapse
|
33
|
Pokharel A, Anderson JD, Deebajah M, Blatt NB, Reddy G, Garlapaty V, Li W, Kanaan HD, Zhang PL. Podocytopathies related to either COVID-19 infection or its vaccination, our experience and literature review. Ultrastruct Pathol 2023; 47:373-381. [PMID: 37463165 DOI: 10.1080/01913123.2023.2237565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/13/2023] [Accepted: 07/13/2023] [Indexed: 07/20/2023]
Abstract
Coronavirus disease 2019 (COVID-19) affects several organs including the kidney resulting in acute kidney injury (AKI) and variants of podocytopathies. From the beginning to the middle period of the COVID-19 pandemic, we have collected eight renal biopsies with various renal diseases including 4 podocytopathies. In addition, from the middle period to the near end of the COVID-19 pandemic, we have seen two of the patients who developed nephrotic syndrome following COVID-19 vaccination. Three of 4 podocytopathies were collapsing glomerulopathy (also called collapsing focal segmental glomerulosclerosis) and the fourth was a minimal change disease (MCD). Two of three collapsing glomerulopathy were found in African American patients, one of who was tested positive for having the high-risk allele APOL-1 G1. In addition, the two renal biopsies showed either MCD or replaced MCD following COVID-19 vaccination. MCD can be a rare complication following COVID-19 infection and COVID-19 vaccination, raising the question if there are similar antigens induced by the infection or by the vaccination that trigger the MCD. This article reports our experience of diagnosing podocytopathies related to either COVID-19 infection or its vaccination and provides a literature review regarding the incidence and potential pathophysiology in the field.
Collapse
Affiliation(s)
- Ashbita Pokharel
- Department of Pathology, Corewell Health (East), Royal Oak, MI, USA
| | | | - Mustafa Deebajah
- Department of Pathology, Corewell Health (East), Royal Oak, MI, USA
| | - Neal B Blatt
- Division of Pediatric Nephrology, Corewell Health (East), Royal Oak, MI, USA
| | - Gampala Reddy
- Division of Nephrology, Corewell Health (East), Royal Oak, MI, USA
| | - Vamshi Garlapaty
- Division of Nephrology, Corewell Health (East), Royal Oak, MI, USA
| | - Wei Li
- Department of Pathology, Corewell Health (East), Royal Oak, MI, USA
| | - Hassan D Kanaan
- Department of Pathology, Corewell Health (East), Royal Oak, MI, USA
| | - Ping L Zhang
- Department of Pathology, Corewell Health (East), Royal Oak, MI, USA
| |
Collapse
|
34
|
Vivarelli M, Gibson K, Sinha A, Boyer O. Childhood nephrotic syndrome. Lancet 2023; 402:809-824. [PMID: 37659779 DOI: 10.1016/s0140-6736(23)01051-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 05/04/2023] [Accepted: 05/19/2023] [Indexed: 09/04/2023]
Abstract
Idiopathic nephrotic syndrome is the most common glomerular disease in children. Corticosteroids are the cornerstone of its treatment, and steroid response is the main prognostic factor. Most children respond to a cycle of oral steroids, and are defined as having steroid-sensitive nephrotic syndrome. Among the children who do not respond, defined as having steroid-resistant nephrotic syndrome, most respond to second-line immunosuppression, mainly with calcineurin inhibitors, and children in whom a response is not observed are described as multidrug resistant. The pathophysiology of nephrotic syndrome remains elusive. In cases of immune-mediated origin, dysregulation of immune cells and production of circulating factors that damage the glomerular filtration barrier have been described. Conversely, up to a third of cases of steroid-resistant nephrotic syndrome have a monogenic origin. Multidrug resistant nephrotic syndrome often leads to kidney failure and can cause relapse after kidney transplant. Although steroid-sensitive nephrotic syndrome does not affect renal function, most children with steroid-sensitive nephrotic syndrome have a relapsing course that requires repeated steroid cycles with significant side-effects. To minimise morbidity, some patients require steroid-sparing immunosuppressive agents, including levamisole, mycophenolate mofetil, calcineurin inhibitors, anti-CD20 monoclonal antibodies, and cyclophosphamide. Close monitoring and preventive measures are warranted at onset and during relapse to prevent acute complications (eg, hypovolaemia, acute kidney injury, infections, and thrombosis), whereas long-term management requires minimising treatment-related side-effects. A subset of patients have active disease into adulthood.
Collapse
Affiliation(s)
- Marina Vivarelli
- Division of Nephrology, Laboratory of Nephrology, Bambino Gesù Children's Hospital IRCCS, Rome, Italy.
| | - Keisha Gibson
- Division of Nephrology and Hypertension, University of North Carolina Kidney Center, University of North Carolina at Chapel Hill, NC, USA
| | - Aditi Sinha
- Division of Nephrology, Indian Council of Medical Research Center for Advanced Research in Nephrology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Olivia Boyer
- Néphrologie Pédiatrique, Centre de Référence Maladies Rénales Héréditaires de l'Enfant et de l'Adulte, Hôpital Necker - Enfants Malades, Assistance Publique Hôpitaux de Paris, Inserm U1163, Institut Imagine, Université Paris Cité, Paris, France
| |
Collapse
|
35
|
Xu X, Qu S, Zhang C, Zhang M, Qin W, Ren G, Bao H, Li L, Zen K, Liu Z. CD8 T Cell-Derived Exosomal miR-186-5p Elicits Renal Inflammation via Activating Tubular TLR7/8 Signal Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301492. [PMID: 37395441 PMCID: PMC10477851 DOI: 10.1002/advs.202301492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/27/2023] [Indexed: 07/04/2023]
Abstract
T cells play an important role in the development of focal segmental glomerulosclerosis (FSGS). The mechanism underlying such T cell-based kidney disease, however, remains elusive. Here the authors report that activated CD8 T cells elicit renal inflammation and tissue injury via releasing miR-186-5p-enriched exosomes. Continuing the cohort study identifying the correlation of plasma level of miR-186-5p with proteinuria in FSGS patients, it is demonstrated that circulating miR-186-5p is mainly derived from activated CD8 T cell exosomes. Renal miR-186-5p, which is markedly increased in FSGS patients and mice with adriamycin-induced renal injury, is mainly delivered by CD8 T cell exosomes. Depleting miR-186-5p strongly attenuates adriamycin-induced mouse renal injury. Supporting the function of exosomal miR-186-5p as a key circulating pathogenic factor, intravenous injection of miR-186-5p or miR-186-5p-containing T cell exosomes results in mouse renal inflammation and tissue injury. Tracing the injected T cell exosomes shows their preferential distribution in mouse renal tubules, not glomerulus. Mechanistically, miR-186-5p directly activates renal tubular TLR7/8 signal and initiates tubular cell apoptosis. Mutating the TLR7-binding sequence on miR-186-5p or deleting mouse TLR7 largely abolishes renal tubular injuries induced by miR-186-5p or adriamycin. These findings reveal a causative role of exosomal miR-186-5p in T cell-mediated renal dysfunction.
Collapse
Affiliation(s)
- Xiaodong Xu
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Shuang Qu
- School of Life Science and TechnologyChina Pharmaceutical University639 Longmian AvenueNanjingJiangsu211198China
| | - Changming Zhang
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Mingchao Zhang
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Weisong Qin
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Guisheng Ren
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Hao Bao
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Limin Li
- School of Life Science and TechnologyChina Pharmaceutical University639 Longmian AvenueNanjingJiangsu211198China
| | - Ke Zen
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing University School of Life SciencesNanjingJiangsu210093China
| | - Zhihong Liu
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| |
Collapse
|
36
|
Chen G, Zeng M, Liu Z, Zhou M, Zha J, Zhang L, Chen H, Liu H. The kinetics of mTORC1 activation associates with FOXP3 expression pattern of CD4+ T cells and outcome of steroid-sensitive minimal change disease. Int Immunopharmacol 2023; 122:110589. [PMID: 37418986 DOI: 10.1016/j.intimp.2023.110589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/09/2023]
Abstract
Minimal change disease (MCD) usually responds to glucocorticoids (GCs) but relapses in most cases. Relapse pathogenesis after complete remission (CR) remains unclear. We hypothesized that FOXP3+ T regulatory cell (Treg) dysregulation may drive early relapses (ER). In this study, a cohort of 23 MCD patients were treated with a conventional GC regimen for the initial onset of nephrotic syndrome. Upon GC withdrawal, seven patients suffered from ER, while 16 patients sustained remission (SR) during the 12-month follow-up. Patients with ER had reduced FOXP3+ Treg proportions compared with healthy controls. Treg reduction, accompanied by IL-10 impairment, was ascribed to a proportional decline of FOXP3medium rather than FOXP3high cells. GC-induced CR was marked by a rise in the proportions of FOXP3+ and FOXP3medium cells compared to baseline levels. These increases declined in patients with ER. The expression level of phosphorylated ribosomal protein S6 was used to track the dynamic shifts in mTORC1 activity within CD4+ T cells of MCD patients at various stages of treatment. Baseline mTORC1 activity was inversely correlated with FOXP3+ and FOXP3medium Treg proportion. The mTORC1 activity in CD4+ T cells served as a reliable indicator for ER and demonstrated improved performance when paired with FOXP3 expression. Mechanically, targeting mTORC1 intervention by siRNAs sufficiently altered the conversion pattern of CD4+ T cell to FOXP3+ Treg. Taken together, the activity of mTORC1 in CD4+ T cells can act as a credible predictor for ER in MCD, especially when combined with FOXP3 expression, and may offer a potential therapeutic avenue for the treatment of podocytopathies.
Collapse
Affiliation(s)
- Guochun Chen
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, China; Clinical Immunology Research Center of Central South University, Changsha, China.
| | - Mengru Zeng
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiwen Liu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Mi Zhou
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jie Zha
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Lei Zhang
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Huihui Chen
- Clinical Immunology Research Center of Central South University, Changsha, China; Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Hong Liu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
37
|
Al Riyami MS, Al Alawi I, Al Gaithi B, Al Maskari A, Al Kalbani N, Al Hashmi N, Al Balushi A, Al Shahi M, Al Saidi S, Al Bimani M, Al Hatali F, Mabillard H, Sayer JA. Genetic analysis and outcomes of Omani children with steroid-resistant nephrotic syndrome. Mol Genet Genomic Med 2023; 11:e2201. [PMID: 37204080 PMCID: PMC10496054 DOI: 10.1002/mgg3.2201] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Nephrotic syndrome (NS) is one of the most common kidney disorders seen by pediatric nephrologists and is defined by the presence of heavy proteinuria (>3.5 g/24 h), hypoalbuminemia (<3.5 g/dL), edema, and hyperlipidemia. Most children with NS are steroid-responsive and have a good prognosis following treatment with prednisolone. However, 10%-20% of them have steroid-resistant nephrotic syndrome (SRNS) and fail to respond to treatment. A significant proportion of these children progress to kidney failure. METHODS This retrospective study aimed to determine the underlying genetic causes of SRNS among Omani children below 13 years old, over a 15-year period and included 77 children from 50 different families. We used targeted Sanger sequencing combined with next-generation sequencing approaches to perform molecular diagnostics. RESULTS We found a high rate of underlying genetic causes of SRNS in 61 (79.2%) children with pathogenic variants in the associated genes. Most of these genetically solved SRNS patients were born to consanguineous parents and variants were in the homozygous state. Pathogenic variants in NPHS2 were the most common cause of SRNS in our study seen in 37 (48.05%) cases. Pathogenic variants in NPHS1 were also seen in 16 cases, especially in infants with congenital nephrotic syndrome (CNS). Other genetic causes identified included pathogenic variants in LAMB2, PLCE1, MYO1E, and NUP93. CONCLUSION NPHS2 and NPHS1 genetic variants were the most common inherited causes of SRNS in Omani children. However, patients with variants in several other SRNS causative genes were also identified. We recommend screening for all genes responsible for SRNS in all children who present with this phenotype, which will assist in clinical management decisions and genetic counseling for the affected families.
Collapse
Affiliation(s)
| | - Intisar Al Alawi
- Translational and Clinical Research Institute, Faculty of Medical ScienceNewcastle UniversityNewcastle upon TyneUK
- National Genetic Center, Ministry of HealthMuscatOman
| | - Badria Al Gaithi
- Pediatric Nephrology Unit, Department of Child HealthyRoyal HospitalMuscatOman
| | - Anisa Al Maskari
- Pediatric Nephrology Unit, Department of Child HealthyRoyal HospitalMuscatOman
| | - Naifain Al Kalbani
- Pediatric Nephrology Unit, Department of Child HealthyRoyal HospitalMuscatOman
| | - Nadia Al Hashmi
- Pediatric Metabolic and Genetic Disorder UnitRoyal HospitalMuscatOman
| | - Aisha Al Balushi
- Pediatric Metabolic and Genetic Disorder UnitRoyal HospitalMuscatOman
| | - Maryam Al Shahi
- Pediatric Clinical Genetic Unit, Royal Hospital, Department of Child HealthRoyal HospitalMuscatOman
| | - Suliman Al Saidi
- Pediatric Nephrology Unit, Department of Child HealthyRoyal HospitalMuscatOman
| | | | | | - Holly Mabillard
- Translational and Clinical Research Institute, Faculty of Medical ScienceNewcastle UniversityNewcastle upon TyneUK
- Renal ServicesThe Newcastle upon Tyne Hospitals NHS Foundation TrustNewcastle upon TyneUK
- Newcastle Biomedical Research Center, NIHRNewcastle upon TyneUK
| | - John A. Sayer
- Translational and Clinical Research Institute, Faculty of Medical ScienceNewcastle UniversityNewcastle upon TyneUK
- Renal ServicesThe Newcastle upon Tyne Hospitals NHS Foundation TrustNewcastle upon TyneUK
- Newcastle Biomedical Research Center, NIHRNewcastle upon TyneUK
| |
Collapse
|
38
|
Gauckler P, Regele H, Eller K, Säemann MD, Lhotta K, Zitt E, Neumann I, Rudnicki M, Odler B, Kronbichler A, Windpessl M. [Diagnosis and treatment of Minimal Change Disease in adults-2023]. Wien Klin Wochenschr 2023; 135:628-637. [PMID: 37728648 PMCID: PMC10511370 DOI: 10.1007/s00508-023-02258-5] [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] [Accepted: 07/24/2023] [Indexed: 09/21/2023]
Abstract
Minimal change disease is a glomerulopathy that clinically manifests as acute onset nephrotic syndrome. A diagnosis is made by renal biopsy, implying the absence of glomerular lesions on light microscopy but detection of extensive podocyte foot process effacement on electron miscroscopy. Considering the typically excellent response to immunosuppressive measures (especially to glucocorticoids), an autoimmune pathogenesis is assumed. Although general prognosis is overall beneficial, steroid-dependent, steroid-resistant and frequently-relapsing disease courses may complicate the management of these patients and necessitate the use of alternative immunosuppressive treatment strategies. Here, the Austrian Society of Nephrology (ÖGN) provides a consensus on how to best diagnose and manage adult patients with minimal change disease.
Collapse
Affiliation(s)
- Philipp Gauckler
- Department Innere Medizin IV (Nephrologie und Hypertensiologie), Medizinische Universität Innsbruck, Innsbruck, Österreich.
| | - Heinz Regele
- Klinisches Institut für Pathologie, Medizinische Universität Wien, Wien, Österreich
| | - Kathrin Eller
- Klinische Abteilung für Nephrologie, Abteilung für Innere Medizin III (Nephrologie, Dialyse und Hypertensiologie), Medizinische Universität Graz, Graz, Österreich
| | - Marcus D Säemann
- 6. Medizinische Abteilung für Nephrologie & Dialyse, Klinik Ottakring, Wien, Österreich
- Medizinische Fakultät, Sigmund-FreudUniversität, Wien, Österreich
| | - Karl Lhotta
- Abteilung für Innere Medizin III (Nephrologie, Dialyse und Hypertensiologie), Akademisches Lehrkrankenhaus Feldkirch, Feldkirch, Österreich
| | - Emanuel Zitt
- Abteilung für Innere Medizin III (Nephrologie, Dialyse und Hypertensiologie), Akademisches Lehrkrankenhaus Feldkirch, Feldkirch, Österreich
| | - Irmgard Neumann
- Vasculitis.at, Wien, Österreich
- Immunologiezentrum Zürich (IZZ), Zürich, Schweiz
| | - Michael Rudnicki
- Department Innere Medizin IV (Nephrologie und Hypertensiologie), Medizinische Universität Innsbruck, Innsbruck, Österreich
| | - Balazs Odler
- Klinische Abteilung für Nephrologie, Abteilung für Innere Medizin III (Nephrologie, Dialyse und Hypertensiologie), Medizinische Universität Graz, Graz, Österreich
| | - Andreas Kronbichler
- Department Innere Medizin 4 (Nephrologie und Hypertensiologie), Medizinische Universität Innsbruck, Innsbruck, Österreich
| | - Martin Windpessl
- Abteilung für Innere Medizin IV, Klinikum Wels-Grieskirchen, Wels, Österreich
| |
Collapse
|
39
|
Vincenti F, Angeletti A, Ghiggeri GM. State of the art in childhood nephrotic syndrome: concrete discoveries and unmet needs. Front Immunol 2023; 14:1167741. [PMID: 37503337 PMCID: PMC10368981 DOI: 10.3389/fimmu.2023.1167741] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 06/21/2023] [Indexed: 07/29/2023] Open
Abstract
Nephrotic syndrome (NS) is a clinical entity characterized by proteinuria, hypoalbuminemia, and peripheral edema. NS affects about 2-7 per 100,000 children aged below 18 years old yearly and is classified, based on the response to drugs, into steroid sensitive (SSNS), steroid dependent, (SDNS), multidrug dependent (MDNS), and multidrug resistant (MRNS). Forms of NS that are more difficult to treat are associated with a worse outcome with respect to renal function. In particular, MRNS commonly progresses to end stage renal failure requiring renal transplantation, with recurrence of the original disease in half of the cases. Histological presentations of NS may vary from minimal glomerular lesions (MCD) to focal segmental glomerulosclerosis (FSGS) and, of relevance, the histological patterns do not correlate with the response to treatments. Moreover, around half of MRNS cases are secondary to causative pathogenic variants in genes involved in maintaining the glomerular structure. The pathogenesis of NS is still poorly understood and therapeutic approaches are mostly based on clinical experience. Understanding of pathogenetic mechanisms of NS is one of the 'unmet needs' in nephrology and represents a significant challenge for the scientific community. The scope of the present review includes exploring relevant findings, identifying unmet needs, and reviewing therapeutic developments that characterize NS in the last decades. The main aim is to provide a basis for new perspectives and mechanistic studies in NS.
Collapse
Affiliation(s)
- Flavio Vincenti
- Division of Nephrology, Department of Medicine and Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Andrea Angeletti
- Nephrology Dialysis and Transplantation, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genoa, Italy
| | - Gian Marco Ghiggeri
- Nephrology Dialysis and Transplantation, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
40
|
Chen DP, Helmuth ME, Smith AR, Canetta PA, Ayoub I, Mucha K, Kallash M, Kopp JB, Gbadegesin R, Gillespie BW, Greenbaum LA, Parekh RS, Hunley TE, Sperati CJ, Selewski DT, Kidd J, Chishti A, Reidy K, Mottl AK, Gipson DS, Srivastava T, Twombley KE. Age of Onset and Disease Course in Biopsy-Proven Minimal Change Disease: An Analysis From the Cure Glomerulonephropathy Network. Am J Kidney Dis 2023; 81:695-706.e1. [PMID: 36608921 PMCID: PMC10200745 DOI: 10.1053/j.ajkd.2022.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 11/19/2022] [Indexed: 01/05/2023]
Abstract
RATIONALE & OBJECTIVE Adolescent- and adult-onset minimal change disease (MCD) may have a clinical course distinct from childhood-onset disease. We characterized the course of children and adults with MCD in the Cure Glomerulonephropathy Network (CureGN) and assessed predictors of rituximab response. STUDY DESIGN Prospective, multicenter, observational study. STUDY PARTICIPANTS CureGN participants with proven MCD on biopsy. EXPOSURE Age at disease onset, initiation of renin-angiotensin-aldosterone system (RAAS) blockade, and immunosuppression including rituximab during the study period. OUTCOME Relapse and remission, change in estimated glomerular filtration rate (eGFR), and kidney failure. ANALYTICAL APPROACH Remission and relapse probabilities were estimated using Kaplan-Meier curves and gap time recurrent event models. Linear regression models were used for the outcome of change in eGFR. Cox proportional hazards models were used to estimate the association between rituximab administration and remission. RESULTS The study included 304 childhood- (≤12 years old), 49 adolescent- (13-17 years old), and 201 adult- (≥18 years) onset participants with 2.7-3.2 years of follow-up after enrollment. Children had a longer time to biopsy (238 vs 23 and 36 days in adolescent- and adult-onset participants, respectively; P<0.001) and were more likely to have received therapy before biopsy. Children were more likely to be treated with immunosuppression but not RAAS blockade. The rate of relapse was higher in childhood- versus adult-onset participants (HR, 1.69 [95% CI, 1.29-2.21]). The probability of remission was also higher in childhood-onset disease (HR, 1.33 [95%CI, 1.02-1.72]). In all groups eGFR loss was minimal. Children were more likely to remit after rituximab than those with adolescent- or adult-onset disease (adjusted HR, 2.1; P=0.003). Across all groups, glucocorticoid sensitivity was associated with a greater likelihood of achieving complete remission after rituximab (adjusted HR, 2.62; P=0.002). LIMITATIONS CureGN was limited to biopsy-proven disease. Comparisons of childhood to nonchildhood cases of MCD may be subject to selection bias, given that childhood cases who undergo a biopsy may be limited to patients who are least responsive to initial therapy. CONCLUSIONS Among patients with MCD who underwent kidney biopsy, there were differences in the course (relapse and remission) of childhood-onset compared with adolescent- and adult-onset disease, as well as rituximab response. PLAIN-LANGUAGE SUMMARY Minimal change disease is a biopsy diagnosis for nephrotic syndrome. It is diagnosed in childhood, adolescence, or adulthood. Patients and clinicians often have questions about what to expect in the disease course and how to plan therapies. We analyzed a group of patients followed longitudinally as part of the Cure Glomerulonephropathy Network (CureGN) and describe the differences in disease (relapse and remission) based on the age of onset. We also analyzed rituximab response. We found that those with childhood-onset disease had a higher rate of relapse but also have a higher probability of reaching remission when compared with adolescent- or adult-onset disease. Children and all steroid-responsive patients are more likely to achieve remission after rituximab.
Collapse
Affiliation(s)
- Dhruti P Chen
- University of North Carolina, Chapel Hill, and Duke Children's Hospital Medical Center, Durham, North Carolina.
| | - Margaret E Helmuth
- Arbor Research Collaborative for Health, and University of Michigan, Ann Arbor, Michigan
| | - Abigail R Smith
- Arbor Research Collaborative for Health, and University of Michigan, Ann Arbor, Michigan
| | - Pietro A Canetta
- Division of Nephrology, Department of Medicine, and Irving Medical Center, Columbia University, New York, and Montefiore Medical Center, Bronx, New York
| | - Isabelle Ayoub
- Division of Nephrology, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Krzysztof Mucha
- Department of Immunology, Transplant Medicine and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Mahmoud Kallash
- Division of Nephrology, Wexner Medical Center, The Ohio State University, Columbus, Ohio; Division of Nephrology, Nationwide Children's Hospital, Columbus, Ohio
| | - Jeffrey B Kopp
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Baltimore, Maryland
| | - Rasheed Gbadegesin
- University of North Carolina, Chapel Hill, and Duke Children's Hospital Medical Center, Durham, North Carolina
| | - Brenda W Gillespie
- Arbor Research Collaborative for Health, and University of Michigan, Ann Arbor, Michigan
| | - Larry A Greenbaum
- Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Rulan S Parekh
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | - David T Selewski
- Medical University of South Carolina, Charleston, South Carolina
| | - Jason Kidd
- Virginia Commonwealth University, Richmond, Virginia
| | | | - Kimberly Reidy
- Division of Nephrology, Department of Medicine, and Irving Medical Center, Columbia University, New York, and Montefiore Medical Center, Bronx, New York
| | - Amy K Mottl
- University of North Carolina, Chapel Hill, and Duke Children's Hospital Medical Center, Durham, North Carolina
| | - Debbie S Gipson
- Arbor Research Collaborative for Health, and University of Michigan, Ann Arbor, Michigan
| | | | | |
Collapse
|
41
|
Del Nogal Avila M, Das R, Kharlyngdoh J, Molina-Jijon E, Donoro Blazquez H, Gambut S, Crowley M, Crossman DK, Gbadegesin RA, Chugh SS, Chugh SS, Avila-Casado C, Macé C, Clement LC, Chugh SS. Cytokine storm-based mechanisms for extrapulmonary manifestations of SARS-CoV-2 infection. JCI Insight 2023; 8:e166012. [PMID: 37040185 PMCID: PMC10322692 DOI: 10.1172/jci.insight.166012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 04/05/2023] [Indexed: 04/12/2023] Open
Abstract
Viral illnesses like SARS-CoV-2 have pathologic effects on nonrespiratory organs in the absence of direct viral infection. We injected mice with cocktails of rodent equivalents of human cytokine storms resulting from SARS-CoV-2/COVID-19 or rhinovirus common cold infection. At low doses, COVID-19 cocktails induced glomerular injury and albuminuria in zinc fingers and homeoboxes 2 (Zhx2) hypomorph and Zhx2+/+ mice to mimic COVID-19-related proteinuria. Common Cold cocktail induced albuminuria selectively in Zhx2 hypomorph mice to model relapse of minimal change disease, which improved after depletion of TNF-α, soluble IL-4Rα, or IL-6. The Zhx2 hypomorph state increased cell membrane to nuclear migration of podocyte ZHX proteins in vivo (both cocktails) and lowered phosphorylated STAT6 activation (COVID-19 cocktail) in vitro. At higher doses, COVID-19 cocktails induced acute heart injury, myocarditis, pericarditis, acute liver injury, acute kidney injury, and high mortality in Zhx2+/+ mice, whereas Zhx2 hypomorph mice were relatively protected, due in part to early, asynchronous activation of STAT5 and STAT6 pathways in these organs. Dual depletion of cytokine combinations of TNF-α with IL-2, IL-13, or IL-4 in Zhx2+/+ mice reduced multiorgan injury and eliminated mortality. Using genome sequencing and CRISPR/Cas9, an insertion upstream of ZHX2 was identified as a cause of the human ZHX2 hypomorph state.
Collapse
Affiliation(s)
- Maria Del Nogal Avila
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Ranjan Das
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Joubert Kharlyngdoh
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Eduardo Molina-Jijon
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Hector Donoro Blazquez
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Stéphanie Gambut
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Michael Crowley
- Genomics Core Lab, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David K. Crossman
- Genomics Core Lab, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rasheed A. Gbadegesin
- Division of Nephrology, Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
| | - Sunveer S. Chugh
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Sunjeet S. Chugh
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Carmen Avila-Casado
- Department of Anatomical Pathology, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
- Instituto Nacional de Cardiología, Mexico City, Mexico
| | - Camille Macé
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Lionel C. Clement
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Sumant S. Chugh
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
42
|
Barry A, McNulty MT, Jia X, Gupta Y, Debiec H, Luo Y, Nagano C, Horinouchi T, Jung S, Colucci M, Ahram DF, Mitrotti A, Sinha A, Teeninga N, Jin G, Shril S, Caridi G, Bodria M, Lim TY, Westland R, Zanoni F, Marasa M, Turudic D, Giordano M, Gesualdo L, Magistroni R, Pisani I, Fiaccadori E, Reiterova J, Maringhini S, Morello W, Montini G, Weng PL, Scolari F, Saraga M, Tasic V, Santoro D, van Wijk JAE, Milošević D, Kawai Y, Kiryluk K, Pollak MR, Gharavi A, Lin F, Simœs E Silva AC, Loos RJF, Kenny EE, Schreuder MF, Zurowska A, Dossier C, Ariceta G, Drozynska-Duklas M, Hogan J, Jankauskiene A, Hildebrandt F, Prikhodina L, Song K, Bagga A, Cheong H, Ghiggeri GM, Vachvanichsanong P, Nozu K, Lee D, Vivarelli M, Raychaudhuri S, Tokunaga K, Sanna-Cherchi S, Ronco P, Iijima K, Sampson MG. Multi-population genome-wide association study implicates immune and non-immune factors in pediatric steroid-sensitive nephrotic syndrome. Nat Commun 2023; 14:2481. [PMID: 37120605 PMCID: PMC10148875 DOI: 10.1038/s41467-023-37985-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 04/10/2023] [Indexed: 05/01/2023] Open
Abstract
Pediatric steroid-sensitive nephrotic syndrome (pSSNS) is the most common childhood glomerular disease. Previous genome-wide association studies (GWAS) identified a risk locus in the HLA Class II region and three additional independent risk loci. But the genetic architecture of pSSNS, and its genetically driven pathobiology, is largely unknown. Here, we conduct a multi-population GWAS meta-analysis in 38,463 participants (2440 cases). We then conduct conditional analyses and population specific GWAS. We discover twelve significant associations-eight from the multi-population meta-analysis (four novel), two from the multi-population conditional analysis (one novel), and two additional novel loci from the European meta-analysis. Fine-mapping implicates specific amino acid haplotypes in HLA-DQA1 and HLA-DQB1 driving the HLA Class II risk locus. Non-HLA loci colocalize with eQTLs of monocytes and numerous T-cell subsets in independent datasets. Colocalization with kidney eQTLs is lacking but overlap with kidney cell open chromatin suggests an uncharacterized disease mechanism in kidney cells. A polygenic risk score (PRS) associates with earlier disease onset. Altogether, these discoveries expand our knowledge of pSSNS genetic architecture across populations and provide cell-specific insights into its molecular drivers. Evaluating these associations in additional cohorts will refine our understanding of population specificity, heterogeneity, and clinical and molecular associations.
Collapse
Affiliation(s)
- Alexandra Barry
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA
- Kidney Disease Initiative & Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michelle T McNulty
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA
- Kidney Disease Initiative & Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Xiaoyuan Jia
- Genome Medical Science Project (Toyama), National Center for Global Health and Medicine (NCGM), Tokyo, Japan
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yask Gupta
- Division of Nephrology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Hanna Debiec
- Sorbonne Université, UPMC Paris 06, Institut National de la Santé et de la Recherde Médicale, Unité Mixte de Rechereche, S 1155, Paris, France
| | - Yang Luo
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, United Kingdom
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - China Nagano
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA
- Kidney Disease Initiative & Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoko Horinouchi
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Seulgi Jung
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Songpa-gu, Seoul, Korea
| | - Manuela Colucci
- Renal Diseases Research Unit, Genetics and Rare Diseases Research Division, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Dina F Ahram
- Division of Nephrology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Adele Mitrotti
- Division of Nephrology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Aditi Sinha
- Department of Pediatrics, AIIMS, New Delhi, India
| | - Nynke Teeninga
- Department of Pediatric Nephrology, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gina Jin
- Division of Nephrology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Shirlee Shril
- Department of Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Gianluca Caridi
- Laboratory on Molecular Nephrology, IRCCS Instituto Giannina Gaslini, Genoa, Italy
| | - Monica Bodria
- Department of Nephrology and Renal Transplantation, IRCCS Instituto Giannina Gaslini, Genoa, Italy
| | - Tze Y Lim
- Division of Nephrology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Rik Westland
- Department of Pediatric Nephrology, VU University Medical Center, Amsterdam, The Netherlands
| | - Francesca Zanoni
- Division of Nephrology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
- Division of Transplantation, Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Maddalena Marasa
- Division of Nephrology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Daniel Turudic
- Department of Pediatric Nephrology, Dialysis and Transplantation, Clinical Hospital Hospital Center Zagreb, University of Zagreb Medical School, Zagreb, Croatia
| | - Mario Giordano
- Division of Nephrology and Pediatric Dialysis, Bari Polyclinic Giovanni XXIII Children's Hospital, Bari, Italy
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Riccardo Magistroni
- Department of Nephrology, Dialysis and Transplant Unit, University Hospital of Modena, Modena, Italy
- Surgical, Medical and Dental Department of Morphological Sciences, Section of Nephrology, University of Modena and Reggio Emilia, Modena, Italy
| | - Isabella Pisani
- Unità Operativa Nefrologia, Azienda Ospedaliero-Universitaria di Parma, Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy
| | - Enrico Fiaccadori
- Unità Operativa Nefrologia, Azienda Ospedaliero-Universitaria di Parma, Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy
| | - Jana Reiterova
- Department of Nephrology, Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | | | - William Morello
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milano, Italy
| | - Giovanni Montini
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milano, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Patricia L Weng
- Department of Pediatric Nephrology, UCLA Medical Center and UCLA Medical Center-Santa Monica, Los Angeles, CA, USA
| | - Francesco Scolari
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, Division of Nephrology and Dialysis, University of Brescia and ASST Spedali Civili of Brescia, Brescia, Italy
| | - Marijan Saraga
- Department of Pediatrics, University of Split, Split, Croatia
| | - Velibor Tasic
- Department of Pediatric Nephrology, University Children's Hospital, Skopje, Macedonia
| | - Domenica Santoro
- Division of Nephrology and Dialysis Unit, University of Messina, Sicily, Italy
| | - Joanna A E van Wijk
- Department of Pediatric Nephrology, VU University Medical Center, Amsterdam, The Netherlands
| | - Danko Milošević
- Department of Pediatric Nephrology, Dialysis and Transplantation, Clinical Hospital Hospital Center Zagreb, University of Zagreb Medical School, Zagreb, Croatia
- Croatian Academy of Medical Sciences, Praska 2/III p.p. 27, 10000, Zagreb, Croatia
| | - Yosuke Kawai
- Genome Medical Science Project (Toyama), National Center for Global Health and Medicine (NCGM), Tokyo, Japan
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Martin R Pollak
- Division of Nephrology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Pediatric, Division of Pediatric Nephrology, Columbia University Irving Medical Center New York-Presbyterian Morgan Stanley Children's Hospital in New York, New York, NY, USA
| | - Ali Gharavi
- Division of Nephrology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Fangmin Lin
- Department of Pediatric, Division of Pediatric Nephrology, Columbia University Irving Medical Center New York-Presbyterian Morgan Stanley Children's Hospital in New York, New York, NY, USA
| | - Ana Cristina Simœs E Silva
- Department of Pediatrics, Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eimear E Kenny
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Genomic Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of General Internal Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michiel F Schreuder
- Department of Pediatric Nephrology, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Aleksandra Zurowska
- Department of Pediatrics, Nephrology and Hypertension, Medical University Gdansk, Gdansk, Poland
| | - Claire Dossier
- AP-HP, Pediatric Nephrology Department, Hôpital Robert-Debré, Paris, France
| | - Gema Ariceta
- Pediatric Nephrology, Hospital Universitari Vall d'Hebron, Universitat Autónoma de Barcelona, Barcelona, Spain
| | | | - Julien Hogan
- AP-HP, Pediatric Nephrology Department, Hôpital Robert-Debré, Paris, France
| | - Augustina Jankauskiene
- Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Friedhelm Hildebrandt
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Larisa Prikhodina
- Research and Clinical Institute for Pediatrics, Pirogov Russian National Research Medical University, Taldomskava St, 2, Moscow, Russia
| | - Kyuyoung Song
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Songpa-gu, Seoul, Korea
| | - Arvind Bagga
- Department of Pediatrics, AIIMS, New Delhi, India
| | - Hae Cheong
- Department of Pediatrics, Hallym University Sacred Heart Hospital, 22, Gwanpyeong-ro 170 beon-gil, Dongan-gu, Anyang-si, Gyeonggi-do, 14068, Korea
| | - Gian Marco Ghiggeri
- Department of Nephrology and Renal Transplantation, IRCCS Instituto Giannina Gaslini, Genoa, Italy
| | - Prayong Vachvanichsanong
- Department of Pediatrics, Faculty of Medicine, Prince of Songkla University, Hat-Yai, Songkhla, 90110, Thailand
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Dongwon Lee
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA
- Kidney Disease Initiative & Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Marina Vivarelli
- Division of Nephrology, and Dialysis, Department of Pediatric Subspecialities, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Centre for Genetics and Genomics Versus Arthritis, University of Manchester, Manchester, UK
| | - Katsushi Tokunaga
- Genome Medical Science Project (Toyama), National Center for Global Health and Medicine (NCGM), Tokyo, Japan
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Simone Sanna-Cherchi
- Division of Nephrology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Pierre Ronco
- Sorbonne Université, UPMC Paris 06, Institut National de la Santé et de la Recherde Médicale, Unité Mixte de Rechereche, S 1155, Paris, France
- Department of Nephrology, Centre Hospitalier du Mans, Le Mans, France
| | - Kazumoto Iijima
- Hyogo Prefectural Kobe Children's Hospital, Kobe, Japan
- Department of Advanced Pediatric Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Matthew G Sampson
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA.
- Kidney Disease Initiative & Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
43
|
Zhang J, Zhao H, Li X, Qian R, Gao P, Lu S, Ma Z. Efficacy of low-dose rituximab in minimal change disease and prevention of relapse. BMC Nephrol 2023; 24:112. [PMID: 37101300 PMCID: PMC10134665 DOI: 10.1186/s12882-023-03092-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 02/20/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND Minimal change disease (MCD) is a major cause of nephrotic syndrome (NS) in children and a minority of adults. The higher tendency to relapse put patients at risk for prolonged exposure to steroids and other immunosuppressive agents. B cell depletion with rituximab (RTX) may be beneficial to the treatment and prevention of frequently relapsing MCD. Therefore, this study aimed to verify the therapeutic/preventive effects of low-dose RTX on the relapse in adult with MCD. METHODS A total of 33 adult patients were selected for the study, including 22 patients with relapsing MCD in relapse treatment group who were treated with low-dose RTX (200 mg per week × 4 following by 200 mg every 6 months) and 11 patients in relapse prevention group with complete remission (CR) after steroid therapy were treated with RTX (200 mg ×1 every 6 months) for preventing the relapse of MCD. RESULTS Of the 22 patients with MCD in relapse treatment group, there were 21 cases (95.45%) of remission [2 (9.09%) partial remission (PR), 19 (86.36%) CR], 1 (4.56%) no remission (NR) and 20 (90.90%) relapse-free. The Median duration of sustained remission was 16.3 months (3, 23.5 months, inter quartile range (IQR)). 11 patients in the relapse prevention group during a follow-up of 12 months (9-31 months) had no relapse. The average dose of prednisone in two groups after RTX treatment was significantly lower than before treatment. CONCLUSION The results of this study suggested low-dose RTX can significantly reduce relapse rate and steroid dose in adults with MCD with fewer side effects. Low-dose RTX regimens may be beneficial for the treatment of relapsing MCD in adults and may be the preferred regimen for patients at high risk for the development of adverse events from corticosteroids.
Collapse
Affiliation(s)
- Jian Zhang
- Division of nephrology, Gansu Provincial Hospital, Lanzhou, 730001, China
| | - Hui Zhao
- Division of nephrology, Gansu Provincial Hospital, Lanzhou, 730001, China
| | - Xiaoli Li
- Division of nephrology, Gansu Provincial Hospital, Lanzhou, 730001, China
| | - Rui Qian
- Division of nephrology, Gansu Provincial Hospital, Lanzhou, 730001, China
| | - Peijuan Gao
- Division of nephrology, Gansu Provincial Hospital, Lanzhou, 730001, China
| | - Shouyan Lu
- Division of nephrology, Gansu Provincial Hospital, Lanzhou, 730001, China
| | - Zhigang Ma
- Department of nephrology, The Second Affiliated Hospital, School of Medcine, The Chinese University of Hong Kong, Longgang District People's Hospital of Shenzhen, Shenzhen, Guangdong, 518172, China.
| |
Collapse
|
44
|
Kanamori T, Kamei K, Sato M, Nishi K, Okutsu M, Ishiwa S, Ogura M, Sako M, Ishikura K, Ito S. CD4 + and CD8 + T-lymphocyte number as predictive marker of relapse after rituximab treatment in childhood-onset refractory nephrotic syndrome. Clin Exp Nephrol 2023:10.1007/s10157-023-02343-z. [PMID: 37095341 DOI: 10.1007/s10157-023-02343-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 03/23/2023] [Indexed: 04/26/2023]
Abstract
BACKGROUND Rituximab is a promising option for refractory idiopathic nephrotic syndrome. However, no simple predictive markers for relapse after rituximab have been established. To determine such markers, we investigated the relationship between CD4 + and CD8 + cell counts and relapse after rituximab administration. METHODS We retrospectively investigated patients with refractory nephrotic syndrome who received rituximab followed by immunosuppressive as maintenance therapy. Patients were divided into no relapse in 2 years after rituximab treatment or relapse group. After rituximab treatment, CD4 + /CD8 + cell counts were measured monthly, at prednisolone discontinuation, and at B-lymphocyte recovery. To predict relapse, these cell counts were analyzed using receiver operating characteristic (ROC). Additionally, relapse-free survival was reevaluated based on the result of ROC analysis for 2 years. RESULTS Forty-eight patients (18 in the relapse group) were enrolled. At prednisolone discontinuation (52 days after rituximab treatment), the relapse-free group showed significantly lower cell counts than the relapse group (median CD4 + cell count: 686 vs. 942 cells/µL, p = 0.006; CD8 + : 613 vs. 812 cells/µL, p = 0.005). In the ROC analysis, CD4 + cell count > 938 cell/µL and CD8 + cell count > 660 cells/µL could predict relapse in 2 years (sensitivity, 56% and 83%; specificity, 87% and 70%). The patient group with both lower CD4 + and CD8 + cell counts showed significantly longer 50% relapse-free survival (1379 vs. 615 days, p < 0.001 and 1379 vs. 640 days, p < 0.001). CONCLUSIONS Lower CD4 + and CD8 + cell counts in the early phase after rituximab administration may predict a lower risk of relapse.
Collapse
Affiliation(s)
- Toru Kanamori
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, Tokyo, Japan
- Department of Pediatrics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Koichi Kamei
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, Tokyo, Japan
| | - Mai Sato
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, Tokyo, Japan
| | - Kentaro Nishi
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, Tokyo, Japan
| | - Mika Okutsu
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, Tokyo, Japan
- Department of Pediatrics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sho Ishiwa
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, Tokyo, Japan
- Department of Pediatrics, Faculty of Medicine, Oita University, Oita, Japan
| | - Masao Ogura
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, Tokyo, Japan
| | - Mayumi Sako
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, Tokyo, Japan
| | - Kenji Ishikura
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, Tokyo, Japan
- Department of Pediatrics, Kitasato University School of Medicine, Kanagawa, Japan
| | - Shuichi Ito
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, Tokyo, Japan.
- Department of Pediatrics, Yokohama City University Hospital, 22-2 Seto, Kanazawa-Ku, Yokohama, Kanagawa, 236-0027, Japan.
| |
Collapse
|
45
|
Liu S, Bush WS, Miskimen K, Gonzalez-Vicente A, Bailey JNC, Konidari I, McCauley JL, Sedor JR, O'Toole JF, Crawford DC. T-cell receptor diversity in minimal change disease in the NEPTUNE study. Pediatr Nephrol 2023; 38:1115-1126. [PMID: 35943576 PMCID: PMC10037226 DOI: 10.1007/s00467-022-05696-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 07/15/2022] [Accepted: 07/15/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Minimal change disease (MCD) is the major cause of childhood idiopathic nephrotic syndrome, which is characterized by massive proteinuria and debilitating edema. Proteinuria in MCD is typically rapidly reversible with corticosteroid therapy, but relapses are common, and children often have many adverse events from the repeated courses of immunosuppressive therapy. The pathobiology of MCD remains poorly understood. Prior clinical observations suggest that abnormal T-cell function may play a central role in MCD pathogenesis. Based on these observations, we hypothesized that T-cell responses to specific exposures or antigens lead to a clonal expansion of T-cell subsets, a restriction in the T-cell repertoire, and an elaboration of specific circulating factors that trigger disease onset and relapses. METHODS To test these hypotheses, we sequenced T-cell receptors in fourteen MCD, four focal segmental glomerulosclerosis (FSGS), and four membranous nephropathy (MN) patients with clinical data and blood samples drawn during active disease and during remission collected by the Nephrotic Syndrome Study Network (NEPTUNE). We calculated several T-cell receptor diversity metrics to assess possible differences between active disease and remission states in paired samples. RESULTS Median productive clonality did not differ between MCD active disease (0.0083; range: 0.0042, 0.0397) and remission (0.0088; range: 0.0038, 0.0369). We did not identify dominant clonotypes in MCD active disease, and few clonotypes were shared with FSGS and MN patients. CONCLUSIONS While these data do not support an obvious role of the adaptive immune system T-cells in MCD pathogenesis, further study is warranted given the limited sample size. A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
Affiliation(s)
- Shiying Liu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - William S Bush
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Kristy Miskimen
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Agustin Gonzalez-Vicente
- Glickman Urological and Kidney Disease and Lerner Research Institutes, Cleveland Clinic, Cleveland, OH, USA
| | - Jessica N Cooke Bailey
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Ioanna Konidari
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Jacob L McCauley
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - John R Sedor
- Glickman Urological and Kidney Disease and Lerner Research Institutes, Cleveland Clinic, Cleveland, OH, USA
| | - John F O'Toole
- Glickman Urological and Kidney Disease and Lerner Research Institutes, Cleveland Clinic, Cleveland, OH, USA
| | - Dana C Crawford
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA.
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA.
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
46
|
Abo Zed SED, Hackl A, Bohl K, Ebert L, Kieckhöfer E, Müller C, Becker K, Fink G, Nüsken KD, Nüsken E, Müller RU, Schermer B, Weber LT. Mycophenolic acid directly protects podocytes by preserving the actin cytoskeleton and increasing cell survival. Sci Rep 2023; 13:4281. [PMID: 36922538 PMCID: PMC10017704 DOI: 10.1038/s41598-023-31326-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Mycophenolate Mofetil (MMF) has an established role as a therapeutic agent in childhood nephrotic syndrome. While other immunosuppressants have been shown to positively affect podocytes, direct effects of MMF on podocytes remain largely unknown. The present study examines the effects of MMF's active component Mycophenolic Acid (MPA) on the transcriptome of podocytes and investigates its biological significance. We performed transcriptomics in cultured murine podocytes exposed to MPA to generate hypotheses on podocyte-specific effects of MPA. Accordingly, we further analyzed biological MPA effects on actin cytoskeleton morphology after treatment with bovine serum albumin (BSA) by immunofluorescence staining, as well as on cell survival following exposure to TNF-α and cycloheximide by neutral red assay. MPA treatment significantly (adjusted p < 0.05) affected expression of 351 genes in podocytes. Gene Ontology term enrichment analysis particularly clustered terms related to actin and inflammation-related cell death. Indeed, quantification of the actin cytoskeleton of BSA treated podocytes revealed a significant increase of thickness and number of actin filaments after treatment with MPA. Further, MPA significantly reduced TNFα and cycloheximide induced cell death. MPA has a substantial effect on the transcriptome of podocytes in vitro, particularly including functional clusters related to non-immune cell dependent mechanisms. This may provide a molecular basis for direct beneficial effects of MPA on the structural integrity and survival of podocytes under pro-inflammatory conditions.
Collapse
Affiliation(s)
- Seif El Din Abo Zed
- Faculty of Medicine and University Hospital Cologne, Department of Pediatrics, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Agnes Hackl
- Faculty of Medicine and University Hospital Cologne, Department of Pediatrics, University of Cologne, Cologne, Germany.
- Faculty of Medicine and University Hospital Cologne, Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.
| | - Katrin Bohl
- Faculty of Medicine and University Hospital Cologne, Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Lena Ebert
- Faculty of Medicine and University Hospital Cologne, Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Emilia Kieckhöfer
- Faculty of Medicine and University Hospital Cologne, Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Carsten Müller
- Faculty of Medicine and University Hospital Cologne, Pharmacology at the Laboratory Center, Department of Therapeutic Drug Monitoring DE, University of Cologne, Cologne, Germany
| | - Kerstin Becker
- Faculty of Medicine and University Hospital Cologne, Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Gregor Fink
- Faculty of Medicine and University Hospital Cologne, Department of Pediatrics, University of Cologne, Cologne, Germany
| | - Kai-Dietrich Nüsken
- Faculty of Medicine and University Hospital Cologne, Department of Pediatrics, University of Cologne, Cologne, Germany
| | - Eva Nüsken
- Faculty of Medicine and University Hospital Cologne, Department of Pediatrics, University of Cologne, Cologne, Germany
| | - Roman-Ulrich Müller
- Faculty of Medicine and University Hospital Cologne, Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, Center for Rare Kidney Diseases Cologne, University of Cologne, Cologne, Germany
| | - Bernhard Schermer
- Faculty of Medicine and University Hospital Cologne, Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Lutz T Weber
- Faculty of Medicine and University Hospital Cologne, Department of Pediatrics, University of Cologne, Cologne, Germany
| |
Collapse
|
47
|
Ye Q, Wang DJ, Lan B, Mao JH. T-cell and B-cell repertoire diversity are selectively skewed in children with idiopathic nephrotic syndrome revealed by high-throughput sequencing. World J Pediatr 2023; 19:273-282. [PMID: 36449136 DOI: 10.1007/s12519-022-00640-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 10/12/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND Clinical studies suggest that the dysfunction of T cells and B cells may play an essential role in the pathogenesis of idiopathic nephrotic syndrome (INS), but laboratory evidence is lacking. Therefore, this study explored T-cell receptor (TCR) and B-cell receptor (BCR) profiling in children with idiopathic nephrotic syndrome. METHODS High-throughput sequencing technology was used to profile the TCR and BCR repertoires in children with INS. Peripheral blood was collected from ten INS patients, including five vinculin autoantibody-positive patients and five vinculin autoantibody-negative patients, before and after treatment. TCR and BCR libraries were constructed by 5'-RACE and sequenced by a DNBSEQ-T7 sequencer, and sequence analyses were performed using ReSeqTools, FastP, MiXCR, and VDJtools. RESULTS The TRA (T-cell receptor α), TRG (T-cell receptor γ), and IGH (immunoglobulin heavy chain) repertoires of the INS group were occupied by highly abundant clonotypes, whereas small clonotypes occupied the healthy group, especially TRA. A significant increase in the Shannon-Weaver index was observed for the TRA and TRG repertoires after treatment in vinculin autoantibody-negative patients, but a significant increase in the IGH repertoire after treatment was observed in vinculin autoantibody-positive patients. The frequency of some V-J pairs was significantly enriched in steroid-sensitive nephrotic syndrome patients. The usage frequency of the V and J genes was skewed in patients, which seemed not related to immunosuppressive therapy. However, after effective treatment, dynamic changes in the size of the individual clonotype were observed. CONCLUSION T-cell and B-cell immunity contribute to the pathogenesis of different INSs. Video: (MP4 99,786 KB).
Collapse
Affiliation(s)
- Qing Ye
- Department of Clinical Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, 310052, China
| | - Dong-Jie Wang
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, 310052, China
| | - Bing Lan
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, 310052, China
| | - Jian-Hua Mao
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, 310052, China.
| |
Collapse
|
48
|
Weerasooriya WALK, Abeyagunawardena AS, Thalgahagoda RS. Single vs split dose of prednisolone in the treatment of relapses of childhood nephrotic syndrome. Eur J Pediatr 2023; 182:1741-1747. [PMID: 36757496 DOI: 10.1007/s00431-023-04804-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 12/30/2022] [Accepted: 01/02/2023] [Indexed: 02/10/2023]
Abstract
Nephrotic syndrome is the commonest glomerular disease in childhood. It usually follows a relapsing and remitting course. Corticosteroids are the mainstay of treatment for both the first episode and subsequent relapses. This study was conducted at a single centre to compare the clinical response to a single dose vs. split dose of prednisolone in the treatment of relapses of childhood nephrotic syndrome. Children between the ages of 1 and 14 years admitted with a relapse of idiopathic steroid sensitive nephrotic syndrome from August 2019 to February 2020 were considered for recruitment. A block randomization method based on age was used for allocation. Patients randomised to group A received oral prednisolone at 60 mg/m2 as a single morning dose, while those randomised to group B received the same total dose as two divided doses, of which 2/3 was given in the morning and the rest in the evening. Treatment was continued until remission was achieved following which all patients were switched to alternate day prednisolone. An independent sample t test was used to compare the two groups. One hundred and four episodes of relapse occurring in 96 children were included of which 49 were treated with prednisolone as a split dose and 55 were treated with a single dose of prednisolone. The mean duration to achieve remission for the split-dose group was 8.02 days (SD 1.58) while it was 9.74 days (SD 3.72) for the single-dose group. This difference was statistically highly significant (t(102) = 3.004; p = 0.001; CI 0.58 to 2.86). There was no difference in the adverse events profile of the two groups. Conclusion: The use of prednisolone as a split dose results in a shorter duration to achieve remission when compared to a single morning dose, resulting in a lower cumulative dose of prednisolone to achieve remission. What is Known: • Corticosteroids are the mainstay of treatment for childhood nephrotic syndrome. • Corticosteroids are given as a single dose in the morning to minimise adrenocortical suppression. What is New: • A more rapid attainment of remission can be achieved with a split dose of corticosteroids.
Collapse
Affiliation(s)
- W A L K Weerasooriya
- Professorial Paediatric Unit, Teaching Hospital Peradeniya, Peradeniya, Sri Lanka
| | - A S Abeyagunawardena
- Department of Paediatrics, Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka
| | - R S Thalgahagoda
- Department of Paediatrics, Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka.
| |
Collapse
|
49
|
Network-Based Assessment of Minimal Change Disease Identifies Glomerular Response to IL-7 and IL-12 Pathways Activation as Innovative Treatment Target. Biomedicines 2023; 11:biomedicines11010226. [PMID: 36672735 PMCID: PMC9856051 DOI: 10.3390/biomedicines11010226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/20/2022] [Accepted: 01/07/2023] [Indexed: 01/18/2023] Open
Abstract
Background: Minimal change disease (MCD), a major cause of nephrotic syndrome, is usually treated by corticosteroid administration. MCD unresponsiveness to therapy and recurrences are nonetheless frequently observed, particularly in adults. To explore MCD-related pathogenetic mechanisms and to identify novel drug targets ultimately contributing to novel therapeutic avenues with a certain specificity for MCD, we compared glomerular transcriptomes from MCD with membranous nephropathy (MN) patients and healthy controls. Methods: Renal biopsies from adult patients with MCD (n = 14) or MN (n = 12), and non-diseased controls (n = 8) were selected from the Norwegian Kidney Biopsy Registry. RNA for 75 base-pair paired-end RNASeq were obtained from laser capture micro-dissected (LCM) glomeruli from FFPE sections. Transcriptional landscapes were computed by combining pathway-centered analyses and network science methodologies that integrate multiple bioinformatics resources. Results: Compared to normal glomeruli, cells from MCD displayed an inflammatory signature apparently governed by the IL1 and IL7 systems. While enrichment of IL1 production and secretion was a shared feature of MCD and MN compared to normal tissue, responses involving IL7 pathway activation were unique to MCD. Indeed, IL7R expressed by glomeruli was the most upregulated gene of the interleukin family in MCD versus normal controls. IL7 pathway activation was paralleled by significant enrichment in adaptive immune system processes and transcriptional regulation and depletion in pathways related to energy metabolism and transcription. Downregulation of these organ function-related themes again occurred predominately in MCD and was significantly less pronounced in MN. Immunofluorescence and immunohistochemistry, respectively, confirmed the expression of phosphorylated IL-7 receptor alpha (IL7RA, CD127) and IL12 receptor beta 1 (IL12RB1) proteins. Conclusions: Gene expression profiling of archival FFPE-biopsies identifies MCD-specific signatures with IL7RA and IL12RB1 as novel targets for MCD treatment.
Collapse
|
50
|
Zen RDC, Dominguez WV, Braga I, dos Reis LM, Jorge LB, Yu L, Woronik V, Dias CB. Urinary CD80 and Serum suPAR as Biomarkers of Glomerular Disease among Adults in Brazil. Diagnostics (Basel) 2023; 13:diagnostics13020203. [PMID: 36673014 PMCID: PMC9857681 DOI: 10.3390/diagnostics13020203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION Urinary CD80 has been shown to have good specificity for minimal change disease (MCD) in children. However, the investigation of circulating factors such as soluble urokinase plasminogen activator receptor (suPAR) as biomarkers of focal segmental glomerulosclerosis (FSGS) is quite controversial. The objective of this study was to determine whether urinary CD80 and serum suPAR can be used for the diagnosis of MCD and FSGS, respectively, in the adult population of Brazil. We also attempted to determine whether those biomarkers assess the response to immunosuppressive treatment. METHODS This was a prospective study in which urine and blood samples were collected for analysis of CD80 and suPAR, respectively, only in the moment of renal biopsy, from patients undergoing to diagnostic renal biopsy. At and six months after biopsy, we analyzed serum creatinine, serum albumin, and proteinuria in order to evaluate the use of the CD80 and suPAR collected in diagnosis as markers of response to immunosuppressive treatment. In healthy controls were collected urinary CD80 and proteinuria, serum suPAR, and creatinine. RESULTS The results of 70 renal biopsies were grouped, by diagnosis, as follows: FSGS (n = 18); membranous nephropathy (n = 14); MCD (n = 5); and other glomerulopathies (n = 33). There was no significant difference among the groups in terms of the urinary CD80 levels, and serum suPAR was not significantly higher in the FSGS group, as would have been expected. Urinary CD80 correlated positively with nephrotic syndrome, regardless of the type of glomerular disease. Neither biomarker correlated with proteinuria at six months after biopsy. CONCLUSION In adults, urinary CD80 can serve as a marker of nephrotic syndrome but is not specific for MCD, whereas serum suPAR does not appear to be useful as a diagnostic or treatment response marker.
Collapse
Affiliation(s)
- Renata de Cássia Zen
- Nephrology Department, Hospital das Clínicas, Faculty of Medicine, University of São Paulo, São Paulo 01246-903, SP, Brazil
- Correspondence: ; Tel.: +55-11-981273865; Fax: +55-11-26617261
| | - Wagner Vasques Dominguez
- Laboratory of Renal Pathophysiology, Hospital das Clínicas, Faculty of Medicine, University of São Paulo, São Paulo 01246-903, SP, Brazil
| | - Ivone Braga
- Laboratory of Renal Pathophysiology, Hospital das Clínicas, Faculty of Medicine, University of São Paulo, São Paulo 01246-903, SP, Brazil
| | - Luciene Machado dos Reis
- Laboratory of Renal Pathophysiology, Hospital das Clínicas, Faculty of Medicine, University of São Paulo, São Paulo 01246-903, SP, Brazil
| | - Lectícia Barbosa Jorge
- Nephrology Department, Hospital das Clínicas, Faculty of Medicine, University of São Paulo, São Paulo 01246-903, SP, Brazil
| | - Luis Yu
- Nephrology Department, Hospital das Clínicas, Faculty of Medicine, University of São Paulo, São Paulo 01246-903, SP, Brazil
| | - Viktoria Woronik
- Laboratory of Renal Pathophysiology, Hospital das Clínicas, Faculty of Medicine, University of São Paulo, São Paulo 01246-903, SP, Brazil
| | - Cristiane Bitencourt Dias
- Laboratory of Renal Pathophysiology, Hospital das Clínicas, Faculty of Medicine, University of São Paulo, São Paulo 01246-903, SP, Brazil
| |
Collapse
|