1
|
Prakash S, Steers NJ, Li Y, Sanchez-Rodriguez E, Verbitsky M, Robbins I, Simpson J, Pathak S, Raska M, Reily C, Ng A, Liang J, DeMaria N, Katiraei A, Stevens KO, Fischman C, Shapiro S, Kodali S, McCutchan J, Park H, Eliby D, Delsante M, Allegri L, Fiaccadori E, Bodria M, Marasa M, Raveche E, Julian BA, Uhlemann AC, Kiryluk K, Zhang H, D’Agati VD, Sanna-Cherchi S, Novak J, Gharavi AG. Loss of GalNAc-T14 links O-glycosylation defects to alterations in B cell homing in IgA nephropathy. J Clin Invest 2025; 135:e181164. [PMID: 40153534 PMCID: PMC12077892 DOI: 10.1172/jci181164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 03/12/2025] [Indexed: 03/30/2025] Open
Abstract
Aberrant O-glycosylation of the IgA1 hinge region is a characteristic finding in patients with IgA nephropathy (IgAN) and is thought to contribute to immune-complex formation and kidney injury. Other studies have suggested that abnormalities in mucosal immunity and lymphocyte homing are major contributors to disease. We identified a family with IgAN segregating a heterozygous predicted loss-of-function (LOF) variant in GALNT14, the gene encoding N-acetylgalactosaminyltransferase 14, one of the enzymes involved in mucin-type protein O-glycosylation. While GALNT14 is expressed in IgA1-producing cells, carriers of the LOF variant did not have altered levels of poorly glycosylated IgA1, suggesting other disease mechanisms. Investigation of Galnt14-null mice revealed elevated serum IgA levels and ex vivo IgA production by B cells. These mice developed glomerular IgA deposition with aging and after induction of sterile colitis. Galnt14-null mice also displayed an attenuated mucin layer in the colon and redistribution of IgA-producing cells from mucosal to systemic sites. Adoptive-transfer experiments indicated impaired homing of spleen-derived Galnt14-deficient B lymphocytes, resulting in increased retention in peripheral blood. These findings suggest that abnormalities in O-glycosylation alter mucosal immunity and B lymphocyte homing, pointing to an expanded role of aberrant O-glycosylation in the pathogenesis of IgAN.
Collapse
Affiliation(s)
- Sindhuri Prakash
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Nicholas J. Steers
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Yifu Li
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Elena Sanchez-Rodriguez
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Miguel Verbitsky
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Isabel Robbins
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Jenna Simpson
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Sharvari Pathak
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | | | - Colin Reily
- Department of Microbiology, and
- Department of Medicine, Division of Nephrology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Anna Ng
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Judy Liang
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Natalia DeMaria
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Amanda Katiraei
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Kelsey O. Stevens
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Clara Fischman
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Samantha Shapiro
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Swetha Kodali
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Jason McCutchan
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Heekuk Park
- Department of Medicine, Division of Infectious Diseases, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Djamila Eliby
- Department of Medicine, Division of Infectious Diseases, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Marco Delsante
- Departments of Medicine and Surgery, University of Parma, Parma, Italy
| | - Landino Allegri
- Departments of Medicine and Surgery, University of Parma, Parma, Italy
| | - Enrico Fiaccadori
- Departments of Medicine and Surgery, University of Parma, Parma, Italy
| | - Monica Bodria
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Maddalena Marasa
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Elizabeth Raveche
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
| | - Bruce A. Julian
- Department of Medicine, Division of Nephrology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Anne-Catrin Uhlemann
- Department of Medicine, Division of Infectious Diseases, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Krzysztof Kiryluk
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | - Hong Zhang
- The Renal Division, Peking University First Hospital, Beijing, China
| | - Vivette D. D’Agati
- Department of Pathology and Cell Biology, Division of Renal Pathology, Columbia University Irving Medical Center, New York, New York, USA
| | - Simone Sanna-Cherchi
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| | | | - Ali G. Gharavi
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
2
|
De Jager P, Zeng L, Khan A, Lama T, Chitnis T, Weiner H, Wang G, Fujita M, Zipp F, Taga M, Kiryluk K. GWAS highlights the neuronal contribution to multiple sclerosis susceptibility. RESEARCH SQUARE 2025:rs.3.rs-5644532. [PMID: 39866869 PMCID: PMC11760239 DOI: 10.21203/rs.3.rs-5644532/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Multiple Sclerosis (MS) is a chronic inflammatory and neurodegenerative disease affecting the brain and spinal cord. Genetic studies have identified many risk loci, that were thought to primarily impact immune cells and microglia. Here, we performed a multi-ancestry genome-wide association study with 20,831 MS and 729,220 control participants, identifying 236 susceptibility variants outside the Major Histocompatibility Complex, including four novel loci. We derived a polygenic score for MS and, optimized for European ancestry, it is informative for African-American and Latino participants. Integrating single-cell data from blood and brain tissue, we identified 76 genes affected by MS risk variants. Notably, while T cells showed the strongest enrichment, inhibitory neurons emerged as a key cell type. The expression of IL7 and STAT3 are affected only in inhibitory neurons, highlighting the importance of neuronal and glial dysfunction in MS susceptibility.
Collapse
Affiliation(s)
| | - Lu Zeng
- Columbia University Irving Medical Center
| | | | | | | | | | | | | | - Frauke Zipp
- University Medical Center of the Johannes Gutenberg University Mainz
| | - Mariko Taga
- Center for Translational & Computational Neuroimmunology
| | | |
Collapse
|
3
|
Cheung CK, Alexander S, Reich HN, Selvaskandan H, Zhang H, Barratt J. The pathogenesis of IgA nephropathy and implications for treatment. Nat Rev Nephrol 2025; 21:9-23. [PMID: 39232245 PMCID: PMC7616674 DOI: 10.1038/s41581-024-00885-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2024] [Indexed: 09/06/2024]
Abstract
IgA nephropathy (IgAN) is a common form of primary glomerulonephritis and represents an important cause of chronic kidney disease globally, with observational studies indicating that most patients are at risk of developing kidney failure within their lifetime. Several research advances have provided insights into the underlying disease pathogenesis, framed by a multi-hit model whereby an increase in circulating IgA1 that lacks galactose from its hinge region - probably derived from the mucosal immune system - is followed by binding of specific IgG and IgA antibodies, generating immune complexes that deposit within the glomeruli, which triggers inflammation, complement activation and kidney damage. Although treatment options are currently limited, new therapies are rapidly emerging that target different pathways, cells and mediators involved in the disease pathogenesis, including B cell priming in the gut mucosa, the cytokines APRIL and BAFF, plasma cells, complement activation and endothelin pathway activation. As more treatments become available, there is a realistic possibility of transforming the long-term outlook for many individuals with IgAN.
Collapse
Affiliation(s)
- Chee Kay Cheung
- Mayer IgA Nephropathy Laboratories, Department of Cardiovascular Sciences, University of Leicester, Leicester, UK.
- John Walls Renal Unit, University Hospitals of Leicester NHS Trust, Leicester, UK.
| | | | - Heather N Reich
- Department of Medicine, Division of Nephrology, University of Toronto, University Health Network, Toronto, ON, Canada
| | - Haresh Selvaskandan
- Mayer IgA Nephropathy Laboratories, Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- John Walls Renal Unit, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, P. R. China
| | - Jonathan Barratt
- Mayer IgA Nephropathy Laboratories, Department of Cardiovascular Sciences, University of Leicester, Leicester, UK.
- John Walls Renal Unit, University Hospitals of Leicester NHS Trust, Leicester, UK.
| |
Collapse
|
4
|
Zhang S, Song Q, Zhang P, Wang X, Guo R, Li Y, Liu S, Yan X, Zhang J, Niu Y, Shi Y, Song T, Xu T, He S. Genome-wide investigation of VNTR motif polymorphisms in 8,222 genomes: Implications for biological regulation and human traits. CELL GENOMICS 2024; 4:100699. [PMID: 39609246 DOI: 10.1016/j.xgen.2024.100699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/31/2024] [Accepted: 11/01/2024] [Indexed: 11/30/2024]
Abstract
Variable number tandem repeat (VNTR) is a pervasive and highly mutable genetic feature that varies in both length and repeat sequence. Despite the well-studied copy-number variants, the functional impacts of repeat motif polymorphisms remain unknown. Here, we present the largest genome-wide VNTR polymorphism map to date, with over 2.5 million VNTR length polymorphisms (VNTR-LPs) and over 11 million VNTR motif polymorphisms (VNTR-MPs) detected in 8,222 high-coverage genomes. Leveraging the large-scale NyuWa cohort, we identified 2,982,456 (31.8%) NyuWa-specific VNTR-MPs, of which 95.3% were rare. Moreover, we found 1,937 out of 38,685 VNTRs that were associated with gene expression through VNTR-MPs in lymphoblastoid cell lines. Specifically, we clarified that the expansion of a likely causal motif could upregulate gene expression by improving the binding concentration of PU.1. We also explored the potential impacts of VNTR polymorphisms on phenotypic differentiation and disease susceptibility. This study expands our knowledge of VNTR-MPs and their functional implications.
Collapse
Affiliation(s)
- Sijia Zhang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Department of Scientific Research, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Qiao Song
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Peng Zhang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaona Wang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Rong Guo
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yanyan Li
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Shuai Liu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaoyu Yan
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jingjing Zhang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yiwei Niu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yirong Shi
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Tingrui Song
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Tao Xu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China.
| | - Shunmin He
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
5
|
Zeng L, Atlas K, Lama T, Chitnis T, Weiner H, Wang G, Fujita M, Zipp F, Taga M, Kiryluk K, De Jager PL. GWAS highlights the neuronal contribution to multiple sclerosis susceptibility. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.04.24318500. [PMID: 39677438 PMCID: PMC11643295 DOI: 10.1101/2024.12.04.24318500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Multiple Sclerosis (MS) is a chronic inflammatory and neurodegenerative disease affecting the brain and spinal cord. Genetic studies have identified many risk loci, that were thought to primarily impact immune cells and microglia. Here, we performed a multi-ancestry genome-wide association study with 20,831 MS and 729,220 control participants, identifying 236 susceptibility variants outside the Major Histocompatibility Complex, including four novel loci. We derived a polygenic score for MS and, optimized for European ancestry, it is informative for African-American and Latino participants. Integrating single-cell data from blood and brain tissue, we identified 76 genes affected by MS risk variants. Notably, while T cells showed the strongest enrichment, inhibitory neurons emerged as a key cell type, highlighting the importance of neuronal and glial dysfunction in MS susceptibility.
Collapse
Affiliation(s)
- Lu Zeng
- Center for Translational and Computational Neuroimmunology & Columbia Multiple Sclerosis Center, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Khan Atlas
- Division of Nephrology, Dept of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Tsering Lama
- Center for Translational and Computational Neuroimmunology & Columbia Multiple Sclerosis Center, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Tanuja Chitnis
- Anne Romney Center for Neurologic Diseases and Brigham Multiple Sclerosis Center, Department of Neurology, Brigham & Women’s Hospital, Boston MA
| | - Howard Weiner
- Anne Romney Center for Neurologic Diseases and Brigham Multiple Sclerosis Center, Department of Neurology, Brigham & Women’s Hospital, Boston MA
| | - Gao Wang
- The Gertrude H. Sergievsky Center and the Department of Neurology, Columbia University, New York, NY, USA
| | - Masashi Fujita
- Center for Translational and Computational Neuroimmunology & Columbia Multiple Sclerosis Center, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Frauke Zipp
- Department of Neurology and Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Mariko Taga
- Center for Translational and Computational Neuroimmunology & Columbia Multiple Sclerosis Center, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Krzysztof Kiryluk
- Division of Nephrology, Dept of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Philip L. De Jager
- Center for Translational and Computational Neuroimmunology & Columbia Multiple Sclerosis Center, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
6
|
Regalia A, Abinti M, Alfieri CM, Campise M, Verdesca S, Zanoni F, Castellano G. Post-transplant glomerular diseases: update on pathophysiology, risk factors and management strategies. Clin Kidney J 2024; 17:sfae320. [PMID: 39664990 PMCID: PMC11630810 DOI: 10.1093/ckj/sfae320] [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: 06/27/2024] [Indexed: 12/13/2024] Open
Abstract
In recent years, advancements in immunosuppressive medications and post-transplant management have led to a significant decrease in acute rejection rates in renal allografts and consequent improvement in short-term graft survival. In contrast, recent data have shown an increased incidence of post-transplant glomerular diseases, which currently represent a leading cause of allograft loss. Although pathogenesis is not fully understood, growing evidence supports the role of inherited and immunological factors and has identified potential pre- and post-transplant predictors. In this review, we illustrate recent advancements in the pathogenesis of post-transplant glomerular disease and the role of risk factors and immunological triggers. In addition, we discuss potential prevention and management strategies.
Collapse
Affiliation(s)
- Anna Regalia
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Matteo Abinti
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Post-Graduate School of Specialization in Nephrology, University of Milan, Milan, Italy
| | - Carlo Maria Alfieri
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Mariarosaria Campise
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Simona Verdesca
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesca Zanoni
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giuseppe Castellano
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
7
|
Chen Z, Zheng Z, Jiang B, Xu Y. Genetic association between celiac disease and chronic kidney disease: a two-sample Mendelian randomization study. Ren Fail 2024; 46:2357246. [PMID: 38832490 DOI: 10.1080/0886022x.2024.2357246] [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: 10/17/2023] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
OBJECTIVE A two-sample Mendelian randomization (MR) analysis was performed to elucidate the causal impact of celiac disease on the risk of chronic kidney disease (CKD). METHODS The study comprised data from three genome-wide association studies involving individuals of European ancestry. The study groups included participants with celiac disease (n = 24,269), CKD (n = 117,165), and estimated glomerular filtration rate levels based on serum creatinine (eGFRcrea, n = 133,413). We employed four widely recognized causal inference algorithms: MR-Egger, inverse variance weighted (IVW), weighted median, and weighted mode. To address potential issues related to pleiotropy and overall effects, MR-Egger regression and the MR-PRESSO global test were performed. Heterogeneity was assessed using Cochran's Q test. RESULTS We identified 14 genetic variants with genome-wide significance. The MR analysis provided consistent evidence across the various methodologies, supporting a causal relationship between celiac disease and an elevated risk of CKD (odds ratio (OR)IVW = 1.027, p = 0.025; ORweighted median = 1.028, P = 0.049; ORweighted mode = 1.030, p = 0.044). Furthermore, we observed a causal link between celiac disease and a decreased eGFRcrea (ORIVW = 0.997, P = 2.94E-06; ORweighted median = 0.996, P = 1.68E-05; ORweighted mode = 0.996, P = 3.11E-04; ORMR Egger = 0.996, P = 5.00E-03). We found no significant evidence of horizontal pleiotropy, heterogeneity, or bias based on MR-Egger regression, MR-PRESSO, and Cochran's Q test. CONCLUSION The results of this study indicate a causal relationship between celiac disease and an increased risk of CKD.
Collapse
Affiliation(s)
- Zhimin Chen
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Zigui Zheng
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Bingjing Jiang
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yanfang Xu
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
8
|
Zhang J, Dong L, Liu W, Sun Y, Lu Y, Lv H, Zhang Y, Wang S. Short-Term Exposure to 2-Amino-1-methyl-6-phenylimidazo[4,5-b]pyridine Induces Colonic Energy Metabolism Disorders in Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:25931-25943. [PMID: 39500793 DOI: 10.1021/acs.jafc.4c08494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) is one of the most abundant heterocyclic aromatic amines generated in thermally processed meat products, and the toxicities of its short-term exposure in the intestines remain unclear. This study aimed to elucidate the short-term PhIP toxicity in colons through administering PhIP orally to rats for 4 weeks. The results indicated that short-term PhIP exposure induced colonic oxidative stress, a significant decrease of serum triglyceride, and a disrupted colonic gene expression pattern associated with mitochondrial electron transport chain and energy metabolism. Thirteen energy metabolites, including lactate and d-erythrose-4-phosphate, showed significant changes under short-term PhIP effects. Energy metabolism pathway analysis revealed that PhIP-induced colonic energy metabolism disorders are characterized by inhibited glycolysis and enhanced tricarboxylic acid cycle. Further investigation found that PhIP altered the energy metabolic phenotype of colon epithelial cells to increase aerobic respiration. In summary, our study provides new insights into the colon toxicity induced by a short-term PhIP exposure.
Collapse
Affiliation(s)
- Jinhui Zhang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Lu Dong
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Quality and Health of Tianjin, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Weiye Liu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Yi Sun
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Yingshuang Lu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Huan Lv
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Yan Zhang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
9
|
Willis TW, Gkrania-Klotsas E, Wareham NJ, McKinney EF, Lyons PA, Smith KGC, Wallace C. Leveraging pleiotropy identifies common-variant associations with selective IgA deficiency. Clin Immunol 2024; 268:110356. [PMID: 39241920 DOI: 10.1016/j.clim.2024.110356] [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: 06/27/2024] [Revised: 08/22/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
Selective IgA deficiency (SIgAD) is the most common inborn error of immunity (IEI). Unlike many IEIs, evidence of a role for highly penetrant rare variants in SIgAD is lacking. Previous SIgAD studies have had limited power to identify common variants due to their small sample size. We overcame this problem first through meta-analysis of two existing GWAS. This identified four novel common-variant associations and enrichment of SIgAD-associated variants in genes linked to Mendelian IEIs. SIgAD showed evidence of shared genetic architecture with serum IgA and a number of immune-mediated diseases. We leveraged this pleiotropy through the conditional false discovery rate procedure, conditioning our SIgAD meta-analysis on large GWAS of asthma and rheumatoid arthritis, and our own meta-analysis of serum IgA. This identified an additional 18 variants, increasing the number of known SIgAD-associated variants to 27 and strengthening the evidence for a polygenic, common-variant aetiology for SIgAD.
Collapse
Affiliation(s)
- Thomas W Willis
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK; Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK.
| | - Effrossyni Gkrania-Klotsas
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, UK; Department of Infectious Diseases, Cambridge University Hospital NHS Foundation Trust, Cambridge, UK
| | - Nicholas J Wareham
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Eoin F McKinney
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Paul A Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Kenneth G C Smith
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Chris Wallace
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK; Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
10
|
Liu L, Zhu L, Monteiro-Martins S, Griffin A, Vlahos LJ, Fujita M, Berrouet C, Zanoni F, Marasa M, Zhang JY, Zhou XJ, Caliskan Y, Akchurin O, Al-Akash S, Jankauskiene A, Bodria M, Chishti A, Esposito C, Esposito V, Claes D, Tesar V, Davis TK, Samsonov D, Kaminska D, Hryszko T, Zaza G, Flynn JT, Iorember F, Lugani F, Rizk D, Julian BA, Hidalgo G, Kallash M, Biancone L, Amoroso A, Bono L, Mani LY, Vogt B, Lin F, Sreedharan R, Weng P, Ranch D, Xiao N, Quiroga A, Matar RB, Rheault MN, Wenderfer S, Selewski D, Lundberg S, Silva C, Mason S, Mahan JD, Vasylyeva TL, Mucha K, Foroncewicz B, Pączek L, Florczak M, Olszewska M, Gradzińska A, Szczepańska M, Machura E, Badeński A, Krakowczyk H, Sikora P, Kwella N, Miklaszewska M, Drożdż D, Zaniew M, Pawlaczyk K, SiniewiczLuzeńczyk K, Bomback AS, Appel GB, Izzi C, Scolari F, Materna-Kiryluk A, Mizerska-Wasiak M, Berthelot L, Pillebout E, Monteiro RC, Novak J, Green TJ, Smoyer WE, Hastings MC, Wyatt RJ, Nelson R, Martin J, González-Gay MA, De Jager PL, Köttgen A, Califano A, Gharavi AG, Zhang H, Kiryluk K. Genome-wide studies define new genetic mechanisms of IgA vasculitis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.10.24315041. [PMID: 39417133 PMCID: PMC11482997 DOI: 10.1101/2024.10.10.24315041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
IgA vasculitis (IgAV) is a pediatric disease with skin and systemic manifestations. Here, we conducted genome, transcriptome, and proteome-wide association studies in 2,170 IgAV cases and 5,928 controls, generated IgAV-specific maps of gene expression and splicing from blood of 255 pediatric cases, and reconstructed myeloid-specific regulatory networks to define disease master regulators modulated by the newly identified disease driver genes. We observed significant association at the HLA-DRB1 (OR=1.55, P=1.1×10-25) and fine-mapped specific amino-acid risk substitutions in DRβ1. We discovered two novel non-HLA loci: FCAR (OR=1.51, P=1.0×10-20) encoding a myeloid IgA receptor FcαR, and INPP5D (OR=1.34, P=2.2×10-9) encoding a known inhibitor of FcαR signaling. The FCAR risk locus co-localized with a cis-eQTL increasing FCAR expression; the risk alleles disrupted a PRDM1 binding motif within a myeloid enhancer of FCAR. Another risk locus was associated with a higher genetically predicted levels of plasma IL6R. The IL6R risk haplotype carried a missense variant contributing to accelerated cleavage of IL6R into a soluble form. Using systems biology approaches, we prioritized IgAV master regulators co-modulated by FCAR, INPP5D and IL6R in myeloid cells. We additionally identified 21 shared loci in a cross-phenotype analysis of IgAV with IgA nephropathy, including novel loci PAID4, WLS, and ANKRD55.
Collapse
Affiliation(s)
- Lili Liu
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Li Zhu
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Sara Monteiro-Martins
- Institute of Genetic Epidemiology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Aaron Griffin
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Lukas J. Vlahos
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Masashi Fujita
- Division of Neuroimmunology, Department of Neurology, Columbia University, New York, NY, USA
| | - Cecilia Berrouet
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Francesca Zanoni
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Maddalena Marasa
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Jun Y. Zhang
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Xu-jie Zhou
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Yasar Caliskan
- Division of Nephrology, Saint Louis University, Saint Louis, MO, USA
| | | | | | | | - Monica Bodria
- MONICA BODRIA, MD, PHD, Primary Care Unit, Ausl Parma, south east district, Parma, Italy
| | - Aftab Chishti
- Division of Pediatric Nephrology, University of Kentucky, Kentucky Children’s Hospital, Lexington, KY, USA
| | - Ciro Esposito
- Istituti Clinico Scientifici Maugeri IRCCS, University of Pavia, Pavia, Italy
| | - Vittoria Esposito
- Istituti Clinico Scientifici Maugeri IRCCS, University of Pavia, Pavia, Italy
| | - Donna Claes
- Cinncinnati Children’s Hospital, Cincinnati, OH, USA
| | - Vladimir Tesar
- Department of Nephrology, 1st School of Medicine, Charles University Prague, Czech Republic
| | | | - Dmitry Samsonov
- Maria Fareri Children’s Hospital (MCF), New York Medical College, New York, NY, USA
| | - Dorota Kaminska
- Department of Non-Procedural Clinical Sciences, Faculty of Medicine, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Tomasz Hryszko
- 2nd Department of Nephrology, Hypertension and Internal Medicine, Medical University of Bialystok, Poland
| | - Gianluigi Zaza
- Renal, Dialysis and Transplant Unit, Department of Pharmacy, Health and Nutritional Sciences (DFSSN), University of Calabria
| | - Joseph T. Flynn
- Department of Pediatrics, University of Washington; and Division of Nephrology, Seattle Children’s Hospital
| | | | | | - Dana Rizk
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | | | - Luisa Bono
- Nephrology and Dialysis, A.R.N.A.S. Civico and Benfratellio, Palermo, Italy
| | - Laila-Yasmin Mani
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Bruno Vogt
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Fangming Lin
- Division of Pediatric Nephrology, Department of Medicine, Columbia University, New York, NY, USA
| | | | | | | | | | | | | | | | - Scott Wenderfer
- Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Dave Selewski
- Mott Children’s Hospital, University of Michigan, Ann Arbor, MI, USA
| | - Sigrid Lundberg
- Danderyd University Hospital, Karolinska Institute, Stockholm, Sweden
| | - Cynthia Silva
- Connecticut Children’s Medical Center, Hartford, CT, USA
| | - Sherene Mason
- Connecticut Children’s Medical Center, Hartford, CT, USA
| | | | | | - Krzysztof Mucha
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Bartosz Foroncewicz
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Leszek Pączek
- Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Michał Florczak
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | | | - Agnieszka Gradzińska
- Department of Dermatology and Pediatric Dermatology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Maria Szczepańska
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Edyta Machura
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Andrzej Badeński
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Helena Krakowczyk
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Przemysław Sikora
- Department of Pediatric Nephrology, Medical University of Lublin, Lublin, Poland
| | - Norbert Kwella
- Department of Nephrology, Transplantology and Internal Diseases, University of Warmia and Mazury, Olsztyn, Poland
| | - Monika Miklaszewska
- Department of Pediatric Nephrology and Hypertension, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Dorota Drożdż
- Department of Pediatric Nephrology and Hypertension, Jagiellonian University Medical College, Krakow, Poland
| | - Marcin Zaniew
- Department of Pediatrics, University of Zielona Góra, Zielona Góra, Poland
| | - Krzysztof Pawlaczyk
- Department of Nephrology, Transplantology and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna SiniewiczLuzeńczyk
- Department of Paediatrics, Immunology and Nephrology, Polish Mother’s Memorial Hospital Research Institute, Lodz, Poland
| | | | | | - Claudia Izzi
- Department of Medical and Surgical Specialties and Nephrology Unit, University of Brescia-ASST Spedali Civili, Brescia, Italy
| | - Francesco Scolari
- Department of Medical and Surgical Specialties and Nephrology Unit, University of Brescia-ASST Spedali Civili, Brescia, Italy
| | | | | | - Laureline Berthelot
- Nantes University, Inserm, CR2TI Center of Research on Translational Transplantation and Immunology, Nantes, France
| | - Evangeline Pillebout
- Center for Research on Inflammation, Paris Cité University, INSERM and CNRS, Paris, France
| | - Renato C. Monteiro
- Center for Research on Inflammation, Paris Cité University, INSERM and CNRS, Paris, France
| | - Jan Novak
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | - Robert J. Wyatt
- Department of Pediatrics, University of Tennessee Health Sciences Center, Memphis, Tennessee
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, Tennessee
| | | | - Javier Martin
- Institute of Parasitology and Biomedicine Lopez-Neyra, Spanish National Research Council (CSIC), Granada, Spain
| | - Miguel A. González-Gay
- Division of Rheumatology, IIS-Fundación Jiménez Díaz, Madrid, Spain
- Medicine and Psychiatry Department, University of Cantabria, Santander, Spain
| | - Philip L. De Jager
- Division of Neuroimmunology, Department of Neurology, Columbia University, New York, NY, USA
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS – Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Andrea Califano
- Department of Systems Biology, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
- Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Chan Zuckerberg Biohub New York, New York, NY, USA
| | - Ali G. Gharavi
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Krzysztof Kiryluk
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| |
Collapse
|
11
|
Zhou XJ, Zhang H. The Genetics of IgA Nephropathy: Implications for Future Therapies. Semin Nephrol 2024; 44:151567. [PMID: 40087125 DOI: 10.1016/j.semnephrol.2025.151567] [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] [Indexed: 03/16/2025]
Abstract
IgA nephropathy (IgAN), the most prevalent primary glomerulonephritis worldwide, carries a considerable lifetime risk of kidney failure. The etiology of IgAN, however, remains incompletely understood, and effective treatment is lacking. Although the multihit model effectively identifies key steps in IgAN development and, to date, provides the best description of IgAN pathogenesis, it remains under development to fully capture the complexity of immune system dysregulation. Large-scale genome-wide association studies have revealed clues regarding the association between IgAN and genes in both innate and adaptive immune pathways. Hence, genetic investigations may shed light on the aberrant molecular mechanisms, thereby presenting new opportunities for therapeutic advancements. This review discusses the genetic associations that have been robustly connected with IgAN, placing them within the framework of disease mechanism. Altogether, these findings highlight numerous new possibilities for the development of treatments and the road to personalized medicine.
Collapse
Affiliation(s)
- Xu-Jie Zhou
- Renal Division, Peking University First Hospital, Beijing, People's Republic of China; Kidney Genetics Center, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, People's Republic of China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, People's Republic of China
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Beijing, People's Republic of China; Kidney Genetics Center, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, People's Republic of China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, People's Republic of China.
| |
Collapse
|
12
|
Novak J. Pathogenesis of IgA nephropathy: Omics data inform glycomedicine. Nephrology (Carlton) 2024; 29 Suppl 2:18-22. [PMID: 39327757 PMCID: PMC11441619 DOI: 10.1111/nep.14350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/11/2024] [Accepted: 06/25/2024] [Indexed: 09/28/2024]
Affiliation(s)
- Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
13
|
Novak J, Reily C, Steers NJ, Schumann T, Rizk DV, Julian BA, Kiryluk K, Gharavi AG, Green TJ. Emerging Biochemical and Immunologic Mechanisms in the Pathogenesis of IgA Nephropathy. Semin Nephrol 2024; 44:151565. [PMID: 40087124 PMCID: PMC11972156 DOI: 10.1016/j.semnephrol.2025.151565] [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] [Indexed: 03/16/2025]
Abstract
IgA nephropathy is a mesangioproliferative glomerular disease with significant morbidity and mortality. Most patients with IgA nephropathy develop kidney failure in their lifetime, reducing their life expectancy by a decade. Since its first description in 1968, it has been established that kidneys of IgA nephropathy patients are injured as "innocent bystanders" by nephritogenic IgA1-containing immune complexes. Results from clinical, biochemical, immunologic, and genetic studies suggest a multistep pathogenetic mechanism. In genetically predisposed individuals, this process results in formation of circulating immune complexes due to the binding of IgG/IgA autoantibodies to the polymeric IgA1 molecules with incomplete O-glycosylation. This event is followed by the addition of other proteins, such as complement C3, resulting in the formation of nephritogenic immune complexes. These complexes are not effectively removed from the circulation, and some of them pass through the fenestration of glomerular endothelial cells to enter the mesangial space and activate mesangial cells. It is thought that the process is initiated by soluble immune complexes and that their accumulation results in the formation of immunodeposits that further amplify glomerular injury. Here we summarize current understanding of the pathogenesis of IgA nephropathy and discuss experimental model systems that can inform development of new therapeutic strategies and targets.
Collapse
Affiliation(s)
- Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL.
| | - Colin Reily
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL; Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Nicholas J Steers
- Division of Nephrology, Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
| | | | - Dana V Rizk
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Bruce A Julian
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
| | - Ali G Gharavi
- Division of Nephrology, Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
| | - Todd J Green
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
14
|
Suzuki H, Novak J. IgA Nephropathy: Significance of IgA1-Containing Immune Complexes in Clinical Settings. J Clin Med 2024; 13:4495. [PMID: 39124764 PMCID: PMC11313413 DOI: 10.3390/jcm13154495] [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: 05/20/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 08/12/2024] Open
Abstract
IgA nephropathy (IgAN) is considered to be an autoimmune disease characterized by the formation of IgA1-containing immune complexes in the circulation and glomerular immunodeposits. Extensive research has identified multiple genetic, immunological, and environmental factors contributing to disease development and progression. The pathogenesis of IgAN is considered a multifactorial process involving the formation of immune complexes wherein aberrantly O-glycosylated IgA1 is recognized as an autoantigen. Consequently, the clinical presentation of IgAN is highly variable, with a wide spectrum of manifestations ranging from isolated microscopic hematuria or episodic macroscopic hematuria to nephrotic-range proteinuria. Whereas some patients may exhibit a slowly progressive form of IgAN, others may present with a rapidly progressive glomerulonephritis leading to kidney failure. Development of the treatment for IgAN requires an understanding of the characteristics of the pathogenic IgA1-containing immune complexes that enter the glomerular mesangium and induce kidney injury. However, not all details of the mechanisms involved in the production of galactose-deficient IgA1 and immune-complex formation are fully understood. Here, we review what we have learned about the characteristics of nephritogenic IgA1 in the half-century since the first description of IgAN in 1968.
Collapse
Affiliation(s)
- Hitoshi Suzuki
- Department of Nephrology, Juntendo University Urayasu Hospital, 2-1-1 Tomioka, Urayasu 279-0021, Chiba, Japan
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
15
|
Tang C, Chen P, Xu LL, Lv JC, Shi SF, Zhou XJ, Liu LJ, Zhang H. Circulating Proteins and IgA Nephropathy: A Multiancestry Proteome-Wide Mendelian Randomization Study. J Am Soc Nephrol 2024; 35:1045-1057. [PMID: 38687828 PMCID: PMC11377805 DOI: 10.1681/asn.0000000000000379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/23/2024] [Indexed: 05/02/2024] Open
Abstract
Key Points
A multiancestry proteome-wide Mendelian randomization analysis was conducted for IgA nephropathy.The findings from the study would help prioritize new drug targets and drug-repurposing opportunities.
Background
The therapeutic options for IgA nephropathy are rapidly evolving, but early diagnosis and targeted treatment remain challenging. We aimed to identify circulating plasma proteins associated with IgA nephropathy by proteome-wide Mendelian randomization studies across multiple ancestry populations.
Methods
In this study, we applied Mendelian randomization and colocalization analyses to estimate the putative causal effects of 2615 proteins on IgA nephropathy in Europeans and 235 proteins in East Asians. Following two-stage network Mendelian randomization, multitrait colocalization analysis and protein-altering variant annotation were performed to strengthen the reliability of the results. A protein–protein interaction network was constructed to investigate the interactions between the identified proteins and the targets of existing medications.
Results
Putative causal effects of 184 and 13 protein–disease pairs in European and East Asian ancestries were identified, respectively. Two protein–disease pairs showed shared causal effects across them (CFHR1 and FCRL2). Supported by the evidence from colocalization analysis, potential therapeutic targets were prioritized and four drug-repurposing opportunities were suggested. The protein–protein interaction network further provided strong evidence for existing medications and pathways that are known to be therapeutically important.
Conclusions
Our study identified a number of circulating proteins associated with IgA nephropathy and prioritized several potential drug targets that require further investigation.
Collapse
Affiliation(s)
- Chen Tang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China; and Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Chen Z, Xu LL, Du W, Ouyang Y, Gu X, Fang Z, Yu X, Li J, Xie L, Jin Y, Ma J, Wang Z, Pan X, Zhang W, Ren H, Wang W, Chen X, Zhou XJ, Zhang H, Chen N, Xie J. Uromodulin and progression of IgA nephropathy. Clin Kidney J 2024; 17:sfae209. [PMID: 39145144 PMCID: PMC11322676 DOI: 10.1093/ckj/sfae209] [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: 03/23/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND This study investigates the link between genetic variants associated with kidney function and immunoglobulin A (IgA) nephropathy (IgAN) progression. METHODS We recruited 961 biopsy-proven IgAN patients and 651 non-IgAN end-stage renal disease (ESRD) patients from Ruijin Hospital. Clinical and renal pathological data were collected. The primary outcome was the time to ESRD. A healthy population was defined as estimated glomerular filtration rate >60 mL/min/1.73 m2 without albuminuria or hematuria. Fifteen single-nucleotide polymorphisms (SNPs) were selected from a genome-wide association study of kidney function and genotyped by the SNaPshot. Immunohistochemistry in renal tissue and ELISA in urine samples were performed to explore the potential functions of genetic variations. RESULTS The rs77924615-G was independently associated with an increased risk for ESRD in IgAN patients after adjustments for clinical and pathologic indices, and treatment (adjusted hazard ratio 2.10; 95% confidence interval 1.14-3.88). No significant differences in ESRD-free survival time were found among different genotypes in non-IgAN ESRD patients (log-rank, P = .480). Moreover, rs77924615 exhibited allele-specific enhancer activity by dual-luciferase reporter assay. Accordingly, the urinary uromodulin-creatinine ratio (uUCR) was significantly higher in healthy individuals with rs77924615 AG or GG than in individuals with AA. Furthermore, uromodulin expression in tubular epithelial cells was higher in patients with rs77924615 AG or GG. Finally, we confirmed that an increased uUCR (P = .009) was associated with faster IgAN progression. CONCLUSION The SNP rs77924615, which modulates the enhancer activity of the UMOD gene, is associated with renal function deterioration in IgAN patients by increasing uromodulin levels in both the renal tubular epithelium and urine.
Collapse
Affiliation(s)
- Zijin Chen
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin-lin Xu
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
| | - Wen Du
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Ouyang
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangchen Gu
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengying Fang
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xialian Yu
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junru Li
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Xie
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanmeng Jin
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Ma
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaohui Wang
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxia Pan
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Zhang
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Ren
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiming Wang
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaonong Chen
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xu-jie Zhou
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
| | - Hong Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
| | - Nan Chen
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingyuan Xie
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
Luo Y, Khan A, Liu L, Lee CH, Perreault GJ, Pomenti SF, Gourh P, Kiryluk K, Bernstein EJ. Cross-Phenotype GWAS Supports Shared Genetic Susceptibility to Systemic Sclerosis and Primary Biliary Cholangitis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.01.24309721. [PMID: 39006426 PMCID: PMC11245064 DOI: 10.1101/2024.07.01.24309721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Objective An increased risk of primary biliary cholangitis (PBC) has been reported in patients with systemic sclerosis (SSc). Our study aims to investigate the shared genetic susceptibility between the two disorders and to define candidate causal genes using cross-phenotype GWAS meta-analysis. Methods We performed cross-phenotype GWAS meta-analysis and colocalization analysis for SSc and PBC. We performed both genome-wide and locus-based analysis, including tissue and pathway enrichment analyses, fine-mapping, colocalization analyses with expression quantitative trait loci (eQTL) and protein quantitative trait loci (pQTL) datasets, and phenome-wide association studies (PheWAS). Finally, we used an integrative approach to prioritize candidate causal genes from the novel loci. Results We detected a strong genetic correlation between SSc and PBC (rg = 0.84, p = 1.7 × 10-6). In the cross-phenotype GWAS meta-analysis, we identified 44 non-HLA loci that reached genome-wide significance (p < 5 × 10-8). Evidence of shared causal variants between SSc and PBC was found for nine loci, five of which were novel. Integrating multiple sources of evidence, we prioritized CD40, ERAP1, PLD4, SPPL3, and CCDC113 as novel candidate causal genes. The CD40 risk locus colocalized with trans-pQTLs of multiple plasma proteins involved in B cell function. Conclusion Our study supports a strong shared genetic susceptibility between SSc and PBC. Through cross-phenotype analyses, we have prioritized several novel candidate causal genes and pathways for these disorders.
Collapse
Affiliation(s)
- Yiming Luo
- Division of Rheumatology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Lili Liu
- Division of Nephrology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Cue Hyunkyu Lee
- Department of Biostatistics, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY
| | - Gabriel J Perreault
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Sydney F Pomenti
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Pravitt Gourh
- Scleroderma Genomics and Health Disparities Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Elana J Bernstein
- Division of Rheumatology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
18
|
Fan Y, Wang Y, Xiao H, Sun H. Advancements in understanding the role of intestinal dysbacteriosis mediated mucosal immunity in IgA nephropathy. BMC Nephrol 2024; 25:203. [PMID: 38907188 PMCID: PMC11191200 DOI: 10.1186/s12882-024-03646-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024] Open
Abstract
IgA nephropathy, presently recognized as the foremost primary glomerular disorder, emerges as a principal contributor to renal failure globally, with its pathogenesis yet to be fully elucidated. Extensive research has highlighted the critical role of gut microbiome in the onset and progression of IgA nephropathy, underscoring its importance in accurately delineating the disease's etiology. For example, gut microbiome dysbacteriosis can lead to the production of nephritogenic IgA1 antibodies, which form immune complexes that deposit in the kidneys, causing inflammation and damage. The gut microbiome, a source of numerous bioactive compounds, interacts with the host and plays a regulatory role in gut-immune axis modulation, earning it the moniker of the "second brain." Recent investigations have particularly emphasized a significant correlation between IgA nephropathy and gut microbiome dysbacteriosis. This article offers a detailed overview of the pathogenic mechanisms of IgA nephropathy, specifically focusing on elucidating how alterations in the gut microbiome are associated with anomalies in the intestinal mucosal system in IgA nephropathy. Additionally, it describes the possible influence of gut microbiome on recurrent IgA nephropathy following kidney transplantation. Furthermore, it compiles potential therapeutic interventions, offering both theoretical and practical foundations for the management of IgA nephropathy. Lastly, the challenges currently faced in the therapeutic approaches to IgA nephropathy are discussed.
Collapse
Affiliation(s)
- Yitao Fan
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730030, Gansu, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Yan Wang
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730030, Gansu, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Han Xiao
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730030, Gansu, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Hui Sun
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730030, Gansu, China.
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
19
|
Trachtman H, Komers R, Inrig J. Sparsentan: the first and only non-immunosuppressive therapy for the reduction of proteinuria in IgA nephropathy. Expert Rev Clin Immunol 2024; 20:571-576. [PMID: 38362830 DOI: 10.1080/1744666x.2024.2319132] [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: 12/30/2023] [Accepted: 02/12/2024] [Indexed: 02/17/2024]
Abstract
INTRODUCTION IgA nephropathy is one of the most common forms of glomerular disease. Patients with persistent proteinuria are at increased risk of progression to kidney failure. There is a significant need for safe and effective therapies to lower proteinuria in these patients. Sparsentan is a non-immunosuppressive agent that acts as a dual angiotensin and endothelin receptor antagonist. It lowers proteinuria in experimental models of glomerular disease and in affected patients. AREAS COVERED This review covers the immunological and non-immunological actions of sparsentan in glomerular disease. It reviews the clinical trials that evaluated the impact of the drug in pediatric and adult patients with IgA nephropathy. It places the use of sparsentan in an overall treatment paradigm for the full spectrum of patients with IgA nephropathy including nonspecific renoprotective agents such as inhibitors of the renin-angiotensin-aldosterone axis and SGLT2 transporter and immunosuppressive drugs. The review represents a search of the current literature about the effect of the drug on normal physiology and the pathogenesis of IgA nephropathy. EXPERT OPINION The safety, tolerability, and therapeutic efficacy of sparsentan have been demonstrated in long-term studies of patients with primary glomerular diseases extending over 5 years. The evidence in support of a beneficial treatment effect of sparsentan is stronger in IgAN than in FSGS. It is anticipated that sparsentan will supplant the use of ACEI or ARB as the first-line therapy to reduce proteinuria prior to the implementation of immunosuppressive agents in patients with IgA nephropathy. It may be combined with other renoprotective drugs like SGLT2 inhibitors. Practice guidelines are needed to promote safe and effective use of this new drug by nephrologists caring for patients with IgAN in all clinical settings.
Collapse
Affiliation(s)
- Howard Trachtman
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | | | - Jula Inrig
- Travere Therapeutics, Inc, San Diego, CA, USA
| |
Collapse
|
20
|
Koehler S, Hengel FE, Dumoulin B, Damashek L, Holzman LB, Susztak K, Huber TB. The 14th International Podocyte Conference 2023: from podocyte biology to glomerular medicine. Kidney Int 2024; 105:935-952. [PMID: 38447880 DOI: 10.1016/j.kint.2024.01.042] [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: 10/30/2023] [Revised: 12/11/2023] [Accepted: 01/02/2024] [Indexed: 03/08/2024]
Abstract
The 14th International Podocyte Conference took place in Philadelphia, Pennsylvania, USA from May 23 to 26, 2023. It commenced with an early-career researchers' meeting on May 23, providing young scientists with a platform to present and discuss their research findings. Throughout the main conference, 29 speakers across 9 sessions shared their insights on podocyte biology, glomerular medicine, novel technologic advancements, and translational approaches. Additionally, the event featured 3 keynote lectures addressing engineered chimeric antigen receptor T cell- and mRNA-based therapies and the use of biobanks for enhanced disease comprehension. Furthermore, 4 brief oral abstract sessions allowed scientists to present their findings to a broad audience. The program also included a panel discussion addressing the challenges of conducting human research within the American Black community. Remarkably, after a 5-year hiatus from in-person conferences, the 14th International Podocyte Conference successfully convened scientists from around the globe, fostering the presentation and discussion of crucial research findings, as summarized in this review. Furthermore, to ensure continuous and sustainable education, research, translation, and trial medicine related to podocyte and glomerular diseases for the benefit of patients, the International Society of Glomerular Disease was officially launched during the conference.
Collapse
Affiliation(s)
- Sybille Koehler
- III. Department of Medicine and Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Felicitas E Hengel
- III. Department of Medicine and Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Bernhard Dumoulin
- III. Department of Medicine and Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Laurel Damashek
- International Society of Glomerular Disease, Florence, Massachusetts, USA
| | - Lawrence B Holzman
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Institute of Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tobias B Huber
- III. Department of Medicine and Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; International Society of Glomerular Disease, Florence, Massachusetts, USA.
| |
Collapse
|
21
|
Yamada K, Huang ZQ, Reily C, Green TJ, Suzuki H, Novak J, Suzuki Y. LIF/JAK2/STAT1 Signaling Enhances Production of Galactose-Deficient IgA1 by IgA1-Producing Cell Lines Derived From Tonsils of Patients With IgA Nephropathy. Kidney Int Rep 2024; 9:423-435. [PMID: 38344714 PMCID: PMC10851019 DOI: 10.1016/j.ekir.2023.11.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/13/2023] [Accepted: 11/06/2023] [Indexed: 02/28/2024] Open
Abstract
Introduction Galactose-deficient IgA1 (Gd-IgA1) plays a key role in the pathogenesis of IgA nephropathy (IgAN). Tonsillectomy has been beneficial to some patients with IgAN, possibly due to the removal of tonsillar cytokine-activated cells producing Gd-IgA1. To test this hypothesis, we used immortalized IgA1-producing cell lines derived from tonsils of patients with IgAN or obstructive sleep apnea (OSA) and assessed the effect of leukemia inhibitory factor (LIF) or oncostatin M (OSM) on Gd-IgA1 production. Methods Gd-IgA1 production was measured by lectin enzyme-linked immunosorbent assay; JAK-STAT signaling in cultured cells was assessed by immunoblotting of cell lysates; and validated by using small interfering RNA (siRNA) knock-down and small-molecule inhibitors. Results IgAN-derived cells produced more Gd-IgA1 than the cells from patients with OSA, and exhibited elevated Gd-IgA1 production in response to LIF, but not OSM. This effect was associated with dysregulated STAT1 phosphorylation, as confirmed by STAT1 siRNA knock-down. JAK2 inhibitor, AZD1480 exhibited a dose-dependent inhibition of the LIF-induced Gd-IgA1 overproduction. Unexpectedly, high concentrations of AZD1480, but only in the presence of LIF, reduced Gd-IgA1 production in the cells derived from patients with IgAN to that of the control cells from patients with OSA. Based on modeling LIF-LIFR-gp130-JAK2 receptor complex, we postulate that LIF binding to LIFR may sequester gp130 and/or JAK2 from other pathways; and when combined with JAK2 inhibition, enables full blockade of the aberrant O-glycosylation pathways in IgAN. Conclusion In summary, IgAN cells exhibit LIF-mediated overproduction of Gd-IgA1 due to abnormal signaling. JAK2 inhibitors can counter these LIF-induced effects and block Gd-IgA1 synthesis in IgAN.
Collapse
Affiliation(s)
- Koshi Yamada
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Zhi-Qiang Huang
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Colin Reily
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Todd J. Green
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hitoshi Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yusuke Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
22
|
Xu R, Zhang J, Hu X, Xu P, Huang S, Cui S, Guo Y, Yang H, Chen X, Jiang C. Yi-shen-hua-shi granules modulate immune and inflammatory damage via the ALG3/PPARγ/NF-κB pathway in the treatment of immunoglobulin a nephropathy. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117204. [PMID: 37757993 DOI: 10.1016/j.jep.2023.117204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/02/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Controversy persists regarding the treatment of immunoglobulin A nephropathy (IgAN), thereby highlighting the demand for safer more effective therapeutic drugs. Although supplementary treatment using Yi-Shen-Hua-Shi (YSHS) granules has distinct advantages with respect to improving renal function in IgAN, a lack of clarity regarding the underlying mechanisms limits their clinical application. AIM OF THE STUDY In this study, we aimed to elucidate the therapeutic mechanisms underlying the efficacy of YSHS granules in the treatment of IgAN. MATERIALS AND METHODS A rat model of IgAN was established based on lipopolysaccharide, carbon tetrachloride, and bovine serum albumin induction. In order to evaluate the effects of YSHS granules, we performed a range of techniques, including immunofluorescence assays, hematoxylin and eosin staining, and flow cytometry, to assess inflammation, immunity, and other relevant factors. Direct data-independent acquisition-mass spectrometry (DIA-MS) analysis and parallel reaction monitoring (PRM) were used for functional characterization and quantitative validation of differentially expressed proteins (DEPs), and Western blot analysis is used to identify downstream proteins associated with DEPs. RESULTS Compared with the model group, the levels of proteinuria, urine red blood cells, serum creatinine, blood urea nitrogen, low-density lipoprotein-cholesterol, triglycerides, and pathological kidney damage were reduced in the YSHS group. A high dose of YSHS granules was found to raise the levels of CD8 T cells and reduce the CD4/CD8 ratio in the peripheral serum. To examine the mechanisms underlying the therapeutic effects YSHS granules, we performed direct DIA-MS analysis to identify proteins that were differentially expressed among the model, YSHS, and control groups. A total of 29 proteins were identified as being commonly expressed in all three groups. Further KEGG and protein-protein interaction (PPI) network analysis revealed that YSHS granules can contribute to the regulation of N-glycosylation-associated proteins, such as ALG3 and STT3A, in rats with IgAN. Detected changes in the expression of ALG3 and STT3A were consistent with the PRM results. We also established that the administration of YSHS granules can contribute to regulation of the ALG3-associated PPAR-γ/NF-κB signaling pathway. CONCLUSIONS Our findings in this study provide evidence to indicate the efficacy of YSHS granules in the treatment of IgAN, the putative underlying mechanisms of which involve the modulation of N-glycosylation, mediated via the PPAR-γ/NF-κB pathway.
Collapse
Affiliation(s)
- Rongjia Xu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Jiajia Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Xingge Hu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Penghao Xu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Shiqi Huang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Shiyan Cui
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yuxin Guo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Hongtao Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Beijing, China
| | - Chen Jiang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
23
|
Yamada K, Novak J, Suzuki Y. GWAS-follow-up Studies Identified a Connection between Abnormal LIF/JAK2/STAT1 Signaling and Overproduction of Galactose-Deficient IgA1 in the Tonsillar IgA1-Secreting Cells from Patients with IgA Nephropathy. JOURNAL OF CLINICAL RESEARCH & BIOETHICS 2024; 15:478. [PMID: 38440092 PMCID: PMC10911063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Affiliation(s)
- Koshi Yamada
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yusuke Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Xu LL, Zhou XJ, Zhang H. An Update on the Genetics of IgA Nephropathy. J Clin Med 2023; 13:123. [PMID: 38202130 PMCID: PMC10780034 DOI: 10.3390/jcm13010123] [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: 11/09/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Immunoglobulin A (IgA) nephropathy (IgAN), the most common form of glomerulonephritis, is one of the leading causes of end-stage kidney disease (ESKD). It is widely believed that genetic factors play a significant role in the development of IgAN. Previous studies of IgAN have provided important insights to unravel the genetic architecture of IgAN and its potential pathogenic mechanisms. The genome-wide association studies (GWASs) together have identified over 30 risk loci for IgAN, which emphasizes the importance of IgA production and regulation in the pathogenesis of IgAN. Follow-up fine-mapping studies help to elucidate the candidate causal variant and the potential pathogenic molecular pathway and provide new potential therapeutic targets. With the rapid development of next-generation sequencing technologies, linkage studies based on whole-genome sequencing (WGS)/whole-exome sequencing (WES) also identify rare variants associated with IgAN, accounting for some of the missing heritability. The complexity of pathogenesis and phenotypic variability may be better understood by integrating genetics, epigenetics, and environment. We have compiled a review summarizing the latest advancements in genetic studies on IgAN. We similarly summarized relevant studies examining the involvement of epigenetics in the pathogenesis of IgAN. Future directions and challenges in this field are also proposed.
Collapse
Affiliation(s)
- Lin-Lin Xu
- Renal Division, Peking University First Hospital, Beijing 100034, China; (L.-L.X.); (H.Z.)
- Kidney Genetics Center, Peking University Institute of Nephrology, Beijing 100034, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing 100034, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing 100034, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing 100034, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100034, China
| | - Xu-Jie Zhou
- Renal Division, Peking University First Hospital, Beijing 100034, China; (L.-L.X.); (H.Z.)
- Kidney Genetics Center, Peking University Institute of Nephrology, Beijing 100034, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing 100034, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing 100034, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing 100034, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100034, China
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Beijing 100034, China; (L.-L.X.); (H.Z.)
- Kidney Genetics Center, Peking University Institute of Nephrology, Beijing 100034, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing 100034, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing 100034, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing 100034, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100034, China
| |
Collapse
|
25
|
Meng MJ, Hu L, Fan Y, Gao H, Chen HZ, Chen CM, Qi Z, Liu B. Efficacy of prednisone combined with mycophenolate mofetil for immunoglobulin A nephropathy with moderate-to-severe renal dysfunction. World J Clin Cases 2023; 11:8300-8309. [PMID: 38130628 PMCID: PMC10731213 DOI: 10.12998/wjcc.v11.i35.8300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 12/14/2023] Open
Abstract
BACKGROUND Immunoglobulin A nephropathy (IgAN) is a common form of chronic glomerulonephritis. Currently, IgAN is one of the main causes of chronic renal failure in China; its prognosis varies greatly between patients, with renal function at the time of diagnosis and prognosis being strongly correlated. Mycophenolate mofetil (MMF) is a drug with a good immunomodulatory effect and is commonly used clinically. However, its effects in IgAN have not yet been clearly demonstrated. Therefore, herein, we retrospectively compared the effectiveness and safety of prednisone alone or combined with MMF for the treatment of primary IgAN with moderate-to-severe renal impairment. AIM To evaluate the effectiveness and safety of prednisone and MMF in treating IgAN with moderate-to-severe renal dysfunction. METHODS Between January 2011 and December 2020, 200 patients with moderate-to-severe IgAN were included in this study, all of whom were admitted to Wuxi People's Hospital affiliated with Nanjing Medical University. All patients underwent a renal puncture biopsy, which revealed primary IgAN with a glomerular filtration rate (GFR) of 30-60 mL/min. The patients were divided into a glucocorticoid therapy group (GTG) and an immunosuppressive therapy group (ITG) according to the different treatment regimens, with 100 patients in each group. Based on general treatments, such as angiotensin-converting enzyme inhibitors/ angiotensin receptor blockers, patients in the GTG were administered prednisone 0.5-0.8 mg/ (kg·d-1) for 4-8 wk, which was reduced by 5 mg every two weeks until the maintenance(30 mg/d) dose was reached and maintained for 12 mo. In the ITG, MMF was administered at 1.0 g/d for 6-12 mo, followed by a maintenance dosage of 0.5 g/d for 12 mo. Age, sex, blood pressure, 24-h urinary egg white measurement, serum creatinine (Scr), blood uric acid, blood albumin, blood potassium (K), hemoglobin, GFR, alanine aminotransferase, total cholesterol (T-CHO), fasting blood glucose, and body mass index were recorded. The 24-h urinary protein, Scr, and GFR levels were recorded 3, 6, 9, and 12 mo after treatment. Follow-up data were also collected. RESULTS No discernible differences existed between the two groups in terms of age, sex, blood pressure, creatinine, 24-h urinary protein level, GFR, or other biochemical indicators at the time of enrollment. Both regimens significantly reduced the 24-h urinary protein quantitation and stabilized renal function. Nine months after treatment, the 24-h urinary protein and Scr of the ITG decreased more significantly than those of the GTG. By the 12th month of treatment, the 24-h urinary protein and Scr in both groups continued to decrease compared to those by the 9th month. In addition, the overall response rate in the ITG was significantly higher than that in the GTG. The occurrence of side effects did not vary significantly between the two regimens; however, endpoint events were significantly more common in the GTG than in the ITG. The follow-up time for the GTG was noticeably lower than that for the ITG. CONCLUSION Prednisone combined with MMF was effective for the treatment of IgAN with moderate-to-severe renal dysfunction.
Collapse
Affiliation(s)
- Mei-Juan Meng
- Department of Nephrology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, Jiangsu Province, China
| | - Ling Hu
- Department of Nephrology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, Jiangsu Province, China
| | - Yun Fan
- Department of Nephrology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, Jiangsu Province, China
| | - Han Gao
- Department of Nephrology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, Jiangsu Province, China
| | - Han-Zhi Chen
- Department of Nephrology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, Jiangsu Province, China
| | - Cai-Mei Chen
- Department of Nephrology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, Jiangsu Province, China
| | - Zhen Qi
- Department of Nephrology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, Jiangsu Province, China
| | - Bin Liu
- Department of Nephrology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, Jiangsu Province, China
| |
Collapse
|
26
|
Rizk DV, Novak L, Hall SD, Moldoveanu Z, Julian BA, Novak J, Haas M. Colocalization of IgG and IgA Heavy Chains with Kappa and Lambda Light Chains in Glomerular Deposits of IgA Nephropathy Patients Using High-Resolution Confocal Microscopy and Correlation with Oxford MEST-C Scores. J Clin Med 2023; 12:7361. [PMID: 38068413 PMCID: PMC10707091 DOI: 10.3390/jcm12237361] [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: 10/18/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 02/12/2024] Open
Abstract
Routine immunofluorescence microscopy of glomerular immunodeposits in IgA nephropathy shows IgA, C3, and lambda light chains, and sometimes IgG, IgM, and kappa light chains. However, a previous study using high-resolution confocal microscopy showed IgG in all IgA nephropathy cases, likely representing autoantibodies specific for galactose-deficient IgA1. Here, we used high-resolution confocal microscopy to examine the composition of glomerular immunodeposits and colocalization of kappa and lambda light chains with IgA or IgG heavy chains in kidney-biopsy samples from twenty patients with IgA nephropathy, seventeen without IgG, and nine with no or trace kappa light chains by routine immunofluorescence microscopy. IgG was detected in all biopsies by high-resolution confocal microscopy. Single-optical-plane images showed similar colocalization of IgG heavy chains with kappa and lambda light chains. Colocalization of IgA heavy chains was greater with lambda light chains than with kappa light chains. Colocalization of IgG heavy chain with kappa light chains was higher than with lambda light chains in biopsies with endocapillary hypercellularity and crescents, i.e., biopsies with active lesions. We confirmed the utility of high-resolution confocal microscopy to detect components of glomerular immunodeposits not apparent on routine immunofluorescence microscopy and for colocalization of different components, potentially clarifying the pathogenesis of IgA nephropathy.
Collapse
Affiliation(s)
- Dana V. Rizk
- Department of Medicine, Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Lea Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (L.N.); (S.D.H.); (Z.M.); (J.N.)
| | - Stacy D. Hall
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (L.N.); (S.D.H.); (Z.M.); (J.N.)
| | - Zina Moldoveanu
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (L.N.); (S.D.H.); (Z.M.); (J.N.)
| | - Bruce A. Julian
- Department of Medicine, Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (L.N.); (S.D.H.); (Z.M.); (J.N.)
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (L.N.); (S.D.H.); (Z.M.); (J.N.)
| | - Mark Haas
- Department of Pathology & Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| |
Collapse
|
27
|
Liao Q, Shen J, Chen Y, Shu Y. Mendelian randomization study on the causal effect of serum IgA levels on H7N9 avian influenza A virus susceptibility. J Med Virol 2023; 95:e29266. [PMID: 38009617 DOI: 10.1002/jmv.29266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/13/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023]
Abstract
Avian influenza A viruses (IAVs) that cross the species barrier to infect humans have the potential to initiate a new pandemic. However, the host factors influencing avian IAV infection remain poorly understood. To address this knowledge gap, we conducted a two-sample Mendelian randomization (MR) analysis by integrating our in-house genome-wide association study (GWAS) of avian IAV H7N9 susceptibility (with 217 cases and 116 controls) with the largest GWAS of serum IgA levels to date (sample size 41 263). Using the inverse-variance weighted (IVW) method, we discovered that genetically decreased serum IgA levels were associated with an increased risk of H7N9 infection (β = -2.528, 95% confidence interval [CI]: -4.572 to -0.484; p = 0.015). Consistent results were obtained from three other MR methods, including robust IVW estimation (β = -2.506, 95% CI: -4.109 to -0.902; p = 0.002), generalized summary-data-based MR (GSMR) (β = -2.238, 95% CI: -4.106 to -0.602; p = 0.019), and MR-pleiotropy residual sum and outlier (MR-PRESSO) (β = -2.528, 95% CI: -4.396 to -0.892; p = 0.026). In conclusion, our analysis provided compelling evidence support a causal relationship between genetically predicted serum IgA levels and avian IAV H7N9 susceptibility.
Collapse
Affiliation(s)
- Qijun Liao
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, People's Republic of China
- BGI Research, Shenzhen, People's Republic of China
| | - Juan Shen
- BGI Research, Shenzhen, People's Republic of China
| | - Yongkun Chen
- Department of Pathogen Biology, Shenzhen University Health Science Center, Shenzhen, People's Republic of China
| | - Yuelong Shu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, People's Republic of China
- Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
28
|
Zhang Y, Li Q, Shi S, Liu L, Lv J, Zhu L, Zhang H. Clinical and pathological characteristics in elderly patients with IgA nephropathy. Clin Kidney J 2023; 16:1974-1979. [PMID: 37915928 PMCID: PMC10616429 DOI: 10.1093/ckj/sfad203] [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: 03/07/2023] [Indexed: 11/03/2023] Open
Abstract
Background Immunoglobulin A nephropathy (IgAN) is the most common cause of primary glomerulonephritis, with highly variable manifestations. Although the peak incidence of IgAN is in young adults, the diagnosis among elderly people is increasing. Here we explored the effect of aging on IgAN features, as well as cellular senescence in the kidney of IgAN. Methods A total of 910 patients with IgAN were enrolled, which contained 182 individuals in each age stage (aged ≥60, 50-59, 40-49, 30-39 and 20-29 years). Clinical and pathological manifestations at the time of renal biopsy were compared. Additionally, 38 patients with IgAN (19 aged over or equal to 60 years and 19 aged below 60 years) were randomly selected for p16INK4a staining by immunohistochemistry. The percentage of p16INK4a-positive cells in glomeruli, renal tubule and interstitium were separately quantified. Results Compared with young IgAN patients, elderly patients presented with higher levels of circulating IgA, uric acid and proteinuria, but lower estimated glomerular filtration rates (eGFR), as well as lower red blood cell counts, platelet counts and lymphocyte counts. Moreover, elderly IgAN patients showed higher incidence of hypertension, and lower incidence of prodromic infection. Regarding histological lesions in the kidney, young IgAN patients had higher degree of IgA and C3 deposits, while elderly IgAN patients had more severe Oxford-E lesions, but less severe Oxford-S lesions. The percentage of glomerular and tubular p16INK4a-positive cells in elderly patients showed an increasing trend, but statistical significance was not reached. The percentage of p16INK4a-positive nuclei in renal interstitium was positively associated with T score, while increased percentage of p16INK4a-positive nuclei in renal tubule was associated with eGFR and 24-h urinary protein level. Conclusion In our IgAN cohort, elderly IgAN patients presented with some aging-related features, and both aging- and IgAN-induced pathological injury contributed to the kidney lesions in patients with IgAN.
Collapse
Affiliation(s)
- Yongji Zhang
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Science, Beijing, China
| | - Qianqian Li
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Science, Beijing, China
| | - Sufang Shi
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Science, Beijing, China
| | - Lijun Liu
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Science, Beijing, China
| | - Jicheng Lv
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Science, Beijing, China
| | - Li Zhu
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Science, Beijing, China
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Science, Beijing, China
| |
Collapse
|
29
|
Rajasekaran A, Green TJ, Renfrow MB, Julian BA, Novak J, Rizk DV. Current Understanding of Complement Proteins as Therapeutic Targets for the Treatment of Immunoglobulin A Nephropathy. Drugs 2023; 83:1475-1499. [PMID: 37747686 PMCID: PMC10807511 DOI: 10.1007/s40265-023-01940-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2023] [Indexed: 09/26/2023]
Abstract
Immunoglobulin A nephropathy (IgAN) is the most common primary glomerulonephritis worldwide and a frequent cause of kidney failure. Currently, the diagnosis necessitates a kidney biopsy, with routine immunofluorescence microscopy revealing IgA as the dominant or co-dominant immunoglobulin in the glomerular immuno-deposits, often with IgG and sometimes IgM or both. Complement protein C3 is observed in most cases. IgAN leads to kidney failure in 20-40% of patients within 20 years of diagnosis and reduces average life expectancy by about 10 years. There is increasing clinical, biochemical, and genetic evidence that the complement system plays a paramount role in the pathogenesis of IgAN. The presence of C3 in the kidney immuno-deposits differentiates the diagnosis of IgAN from subclinical glomerular mesangial IgA deposition. Markers of complement activation via the lectin and alternative pathways in kidney-biopsy specimens are associated with disease activity and are predictive of poor outcome. Levels of select complement proteins in the circulation have also been assessed in patients with IgAN and found to be of prognostic value. Ongoing genetic studies have identified at least 30 loci associated with IgAN. Genes within some of these loci encode complement-system regulating proteins that can interact with immune complexes. The growing appreciation for the central role of complement components in IgAN pathogenesis highlighted these pathways as potential treatment targets and sparked great interest in pharmacological agents targeting the complement cascade for the treatment of IgAN, as evidenced by the plethora of ongoing clinical trials.
Collapse
Affiliation(s)
- Arun Rajasekaran
- Division of Nephrology, Department of Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Todd J Green
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Matthew B Renfrow
- Department of Biochemistry and Molecular Genetics, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Bruce A Julian
- Division of Nephrology, Department of Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jan Novak
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dana V Rizk
- Division of Nephrology, Department of Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
30
|
Zhang X, Wan Z, Lin M, Li Y, Wu X, Jiang J, Lin S, Chi X. Immunoglobulin A and complement C4 are involved in the progression of liver fibrosis in patients with chronic hepatitis B. Int Immunopharmacol 2023; 122:110604. [PMID: 37451022 DOI: 10.1016/j.intimp.2023.110604] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 06/28/2023] [Accepted: 07/02/2023] [Indexed: 07/18/2023]
Abstract
OBJECTIVE To explore the relationship between immunoglobulin A (IgA), complement C4, and liver fibrosis (L.F.) progression (LFP) in patients with chronic hepatitis B (CHB). METHODS This is a retrospective cohort study of CHB patients who received liver biopsies. Relevant data, including demographics, clinical serum markers, and immunological results obtained during liver biopsies, were collected and analyzed to assess and verify the relationship between IgA, C4, and LFP. RESULTS This study included 1,938 CHB patients, of whom 132 received two liver biopsies (group 1). Thirty (22.7%) of these patients were diagnosed with LFP (increase in L.F. stage (Scheuer score F ≥ 1)). IgA (C-IgA) and C4 (C-C4) change values between the first and second biopsies were independent risk factors for LFP. IgA levels increased, and C4 levels decreased during the second liver puncture. The remaining 1,806 patients received one liver puncture (group 2). They were divided into the following subgroups: A (F ≤ 1), B (1 < F ≤ 3), and C (F > 3) to verify whether the same trend was observed by cross-sectional study. IgA levels were highest, and C4 levels were lowest in group C (IgA: C > B > A, p < 0.05; C4: C < B < A, p < 0.05). CONCLUSIONS The findings of this study suggest that serum IgA and C4 levels are independent risk factors for LFP that could serve as future targets for L.F. management and treatment.
Collapse
Affiliation(s)
- Xiujuan Zhang
- The Second Clinical College of Guangzhou University of Chinese Medicine, 510120, Guangzhou, PR China; Department of Hepatology Diseases, Guangdong Provincial Hospital of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 510120, Guangzhou, PR China
| | - Zemin Wan
- Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 510120, Guangzhou, PR China
| | - Ming Lin
- Department of Hepatology Diseases, Guangdong Provincial Hospital of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 510120, Guangzhou, PR China
| | - Yingxian Li
- Department of Medical Education, Guangdong Provincial Hospital of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 510120, Guangzhou, PR China
| | - Xiaoju Wu
- Department of Hepatology Diseases, Guangdong Provincial Hospital of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 510120, Guangzhou, PR China
| | - Junmin Jiang
- Department of Hepatology Diseases, Guangdong Provincial Hospital of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 510120, Guangzhou, PR China
| | - Shanshan Lin
- Department of Hepatology Diseases, Guangdong Provincial Hospital of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 510120, Guangzhou, PR China
| | - Xiaoling Chi
- Department of Hepatology Diseases, Guangdong Provincial Hospital of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 510120, Guangzhou, PR China.
| |
Collapse
|
31
|
Wang YN, Gan T, Qu S, Xu LL, Hu Y, Liu LJ, Shi SF, Lv JC, Tsoi LC, Patrick MT, He K, Berthier CC, Xu HJ, Zhou XJ, Zhang H. MTMR3 risk alleles enhance Toll Like Receptor 9-induced IgA immunity in IgA nephropathy. Kidney Int 2023; 104:562-576. [PMID: 37414396 DOI: 10.1016/j.kint.2023.06.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 05/29/2023] [Accepted: 06/15/2023] [Indexed: 07/08/2023]
Abstract
Multiple genome-wide association studies (GWASs) have reproducibly identified the MTMR3/HORMAD2/LIF/OSM locus to be associated with IgA nephropathy (IgAN). However, the causal variant(s), implicated gene(s), and altered mechanisms remain poorly understood. Here, we performed fine-mapping analyses based on GWAS datasets encompassing 2762 IgAN cases and 5803 control individuals, and identified rs4823074 as the candidate causal variant that intersects the MTMR3 promoter in B-lymphoblastoid cells. Mendelian randomization studies suggested the risk allele may modulate disease susceptibility by affecting serum IgA levels through increased MTMR3 expression. Consistently, elevated MTMR3 expression in peripheral blood mononuclear cells was observed in patients with IgAN. Further mechanistic studies in vitro demonstrated that MTMR3 increased IgA production dependent upon its phosphatidylinositol 3-phosphate binding domain. Moreover, our study provided the in vivo functional evidence that Mtmr3-/- mice exhibited defective Toll Like Receptor 9-induced IgA production, glomerular IgA deposition, as well as mesangial cell proliferation. RNA-seq and pathway analyses showed that MTMR3 deficiency resulted in an impaired intestinal immune network for IgA production. Thus, our results support the role of MTMR3 in IgAN pathogenesis by enhancing Toll Like Receptor 9-induced IgA immunity.
Collapse
Affiliation(s)
- Yan-Na Wang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China; Peking University Institute of Nephrology, Peking University, Beijing, People's Republic of China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, People's Republic of China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, People's Republic of China
| | - Ting Gan
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China; Peking University Institute of Nephrology, Peking University, Beijing, People's Republic of China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, People's Republic of China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, People's Republic of China
| | - Shu Qu
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China; Peking University Institute of Nephrology, Peking University, Beijing, People's Republic of China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, People's Republic of China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, People's Republic of China
| | - Lin-Lin Xu
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China; Peking University Institute of Nephrology, Peking University, Beijing, People's Republic of China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, People's Republic of China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, People's Republic of China
| | - Yong Hu
- Beijing Institute of Biotechnology, Beijing, People's Republic of China
| | - Li-Jun Liu
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China; Peking University Institute of Nephrology, Peking University, Beijing, People's Republic of China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, People's Republic of China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, People's Republic of China
| | - Su-Fang Shi
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China; Peking University Institute of Nephrology, Peking University, Beijing, People's Republic of China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, People's Republic of China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, People's Republic of China
| | - Ji-Cheng Lv
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China; Peking University Institute of Nephrology, Peking University, Beijing, People's Republic of China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, People's Republic of China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, People's Republic of China
| | - Lam C Tsoi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, USA; Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew T Patrick
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, USA; Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Kevin He
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA; Kidney Epidemiology and Cost Center, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Celine C Berthier
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Hu-Ji Xu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai, People's Republic of China
| | - Xu-Jie Zhou
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China; Peking University Institute of Nephrology, Peking University, Beijing, People's Republic of China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, People's Republic of China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, People's Republic of China.
| | - Hong Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China; Peking University Institute of Nephrology, Peking University, Beijing, People's Republic of China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, People's Republic of China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, People's Republic of China.
| |
Collapse
|
32
|
Li J, Zhao M, Luo W, Huang J, Zhao B, Zhou Z. B cell metabolism in autoimmune diseases: signaling pathways and interventions. Front Immunol 2023; 14:1232820. [PMID: 37680644 PMCID: PMC10481957 DOI: 10.3389/fimmu.2023.1232820] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/09/2023] [Indexed: 09/09/2023] Open
Abstract
Autoimmune diseases are heterogeneous disorders believed to stem from the immune system's inability to distinguish between auto- and foreign- antigens. B lymphocytes serve a crucial role in humoral immunity as they generate antibodies and present antigens. Dysregulation of B cell function induce the onset of autoimmune disorders by generating autoantibodies and pro-inflammatory cytokines, resulting in an imbalance in immune regulation. New research in immunometabolism shows that cellular metabolism plays an essential role in controlling B lymphocytes immune reactions by providing the energy and substrates for B lymphocytes activation, differentiation, and function. However, dysregulated immunometabolism lead to autoimmune diseases by disrupting self-tolerance mechanisms. This review summarizes the latest research on metabolic reprogramming of B lymphocytes in autoimmune diseases, identifying crucial pathways and regulatory factors. Moreover, we consider the potential of metabolic interventions as a promising therapeutic strategy. Understanding the metabolic mechanisms of B cells brings us closer to developing novel therapies for autoimmune disorders.
Collapse
Affiliation(s)
- Jingyue Li
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Mingjiu Zhao
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Wenjun Luo
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jiaqi Huang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Xiangya School of Public Health, Central South University, Changsha, China
| | - Bin Zhao
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Furong Laboratory, Central South University, Changsha, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
33
|
Xu LL, Zhang D, Weng HY, Wang LZ, Chen RY, Chen G, Shi SF, Liu LJ, Zhong XH, Hong SD, Duan LX, Lv JC, Zhou XJ, Zhang H. Machine learning in predicting T-score in the Oxford classification system of IgA nephropathy. Front Immunol 2023; 14:1224631. [PMID: 37600788 PMCID: PMC10437057 DOI: 10.3389/fimmu.2023.1224631] [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: 05/18/2023] [Accepted: 07/21/2023] [Indexed: 08/22/2023] Open
Abstract
Background Immunoglobulin A nephropathy (IgAN) is one of the leading causes of end-stage kidney disease (ESKD). Many studies have shown the significance of pathological manifestations in predicting the outcome of patients with IgAN, especially T-score of Oxford classification. Evaluating prognosis may be hampered in patients without renal biopsy. Methods A baseline dataset of 690 patients with IgAN and an independent follow-up dataset of 1,168 patients were used as training and testing sets to develop the pathology T-score prediction (T pre) model based on the stacking algorithm, respectively. The 5-year ESKD prediction models using clinical variables (base model), clinical variables and real pathological T-score (base model plus T bio), and clinical variables and T pre (base model plus T pre) were developed separately in 1,168 patients with regular follow-up to evaluate whether T pre could assist in predicting ESKD. In addition, an external validation set consisting of 355 patients was used to evaluate the performance of the 5-year ESKD prediction model using T pre. Results The features selected by AUCRF for the T pre model included age, systolic arterial pressure, diastolic arterial pressure, proteinuria, eGFR, serum IgA, and uric acid. The AUC of the T pre was 0.82 (95% CI: 0.80-0.85) in an independent testing set. For the 5-year ESKD prediction model, the AUC of the base model was 0.86 (95% CI: 0.75-0.97). When the T bio was added to the base model, there was an increase in AUC [from 0.86 (95% CI: 0.75-0.97) to 0.92 (95% CI: 0.85-0.98); P = 0.03]. There was no difference in AUC between the base model plus T pre and the base model plus T bio [0.90 (95% CI: 0.82-0.99) vs. 0.92 (95% CI: 0.85-0.98), P = 0.52]. The AUC of the 5-year ESKD prediction model using T pre was 0.93 (95% CI: 0.87-0.99) in the external validation set. Conclusion A pathology T-score prediction (T pre) model using routine clinical characteristics was constructed, which could predict the pathological severity and assist clinicians to predict the prognosis of IgAN patients lacking kidney pathology scores.
Collapse
Affiliation(s)
- Lin-Lin Xu
- Renal Division, Peking University First Hospital, Kidney Genetics Center, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| | - Di Zhang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, China
- WeGene, Shenzhen Zaozhidao Technology, Shenzhen, China
- Shenzhen WeGene Clinical Laboratory, Shenzhen, China
| | - Hao-Yi Weng
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, China
- WeGene, Shenzhen Zaozhidao Technology, Shenzhen, China
- Shenzhen WeGene Clinical Laboratory, Shenzhen, China
| | - Li-Zhong Wang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, China
- WeGene, Shenzhen Zaozhidao Technology, Shenzhen, China
- Shenzhen WeGene Clinical Laboratory, Shenzhen, China
| | - Ruo-Yan Chen
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, China
- WeGene, Shenzhen Zaozhidao Technology, Shenzhen, China
- Shenzhen WeGene Clinical Laboratory, Shenzhen, China
| | - Gang Chen
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, China
- WeGene, Shenzhen Zaozhidao Technology, Shenzhen, China
- Shenzhen WeGene Clinical Laboratory, Shenzhen, China
| | - Su-Fang Shi
- Renal Division, Peking University First Hospital, Kidney Genetics Center, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| | - Li-Jun Liu
- Renal Division, Peking University First Hospital, Kidney Genetics Center, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| | - Xu-Hui Zhong
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Shen-Da Hong
- Institute of Medical Technology, Health Science Center of Peking University, Beijing, China
| | - Li-Xin Duan
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ji-Cheng Lv
- Renal Division, Peking University First Hospital, Kidney Genetics Center, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| | - Xu-Jie Zhou
- Renal Division, Peking University First Hospital, Kidney Genetics Center, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Kidney Genetics Center, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| |
Collapse
|
34
|
Yadav K, Ramachandran R, Kumar V, Yadav AK, Pal D, Gopalakrishnan N, Sharma S, Priyamvada PS, Lahiri A, Sahay M, Raju SB, Sreelatha M, Manorajan R, Mukhopadhyay P, Prasad N, Meena P, Kohli HS, Vikrant S, Jha V. Indian TrANslational GlomerulonephrItis BioLogy nEtwork (I-TANGIBLE): Design and Methods. Indian J Nephrol 2023; 33:277-282. [PMID: 37781560 PMCID: PMC10503576 DOI: 10.4103/ijn.ijn_305_23] [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: 07/13/2023] [Revised: 07/14/2023] [Accepted: 07/19/2023] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND AND AIM Primary glomerular disease accounts for one-sixth of all chronic kidney diseases (CKDs) in India. We remain limited in our ability to effectively treat these conditions because of lack of understanding of the disease mechanisms and lack of predictors to identify the clinical course and therapeutic responsiveness. We propose to develop a network of investigators in glomerular diseases, collect information in a systematic fashion to understand the clinical outcomes, answer translational research questions better, and identify and recruit patients for clinical trials. MATERIALS AND METHODS This is a prospective, observational study. The Indian TrANslational GlomerulonephrItis BioLogy nEtwork (I-TANGIBLE) cohort will enroll patients (>18 years) with biopsy-proven minimal change disease (MCD), focal segmental glomerulonephritis (FSGS), membranous nephropathy (MN), IgA nephropathy (IgAN), or membranoproliferative glomerulonephritis (MPGN) (immune complex- and complement-mediated), with first biopsy taken within 2 years of enrollment. Patients with estimated glomerular filtration (eGFR) rate <15 ml/min/1.73 m2 for >3 months at the time of screening, kidney transplant or bone marrow transplant recipients, patients with active malignancy, and patients with active hepatitis B/C replication or human immunodeficiency virus (HIV)-I/II will be excluded. Clinical details including history, medication history and details, and family history will be obtained. Consenting patient's blood and urine samples will be collected and stored, aligned to their clinical follow-up. EXPECTED OUTCOMES The network will allow accurate ascertainment of disease burden of glomerular diseases across study sites, establishment of the treatment pattern of common glomerular diseases, investigation of medium- and long-term outcomes (remission, relapse, rate of eGFR decline), and building a suitable infrastructure to carry out clinical trials in primary glomerular disease.
Collapse
Affiliation(s)
- Kavita Yadav
- Chronic Kidney Disease, George Institute for Global Health India, New Delhi, India
| | - Raja Ramachandran
- Departments of Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Vinod Kumar
- Dermatology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ashok K. Yadav
- Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Deeksha Pal
- Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Sourabh Sharma
- Department of Nephrology, VMMC and Safdarjung Hospital, New Delhi, India
| | - P. S. Priyamvada
- Department of Nephrology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Arpita Lahiri
- Department of Nephrology, Institute of Postgraduate Medical Education and Research, Kolkata, West Bengal, India
| | - Manisha Sahay
- Department of Nephrology, Osmania Medical College, Hyderabad, Telangana, India
| | - Sree Bhushan Raju
- Department of Nephrology, Nizam’s Institute of Medical Sciences, Hyderabad, Telangana, India
| | - M Sreelatha
- Department of Nephrology, Government Medical College, Calicut, Kerala, India
| | - R Manorajan
- Department of Nephrology, Madurai Medical College, Madurai, Tamil Nadu, India
| | | | - Narayan Prasad
- Department of Nephrology, Sanjay Gandhi PGIMS, Lucknow, Uttar Pradesh, India
| | - Priti Meena
- Department of Nephrology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Harbir S. Kohli
- Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sanjay Vikrant
- Department of Nephrology, All India Institute of Medical Sciences, Bilaspur, Himachal Pradesh, India
| | - Vivekanand Jha
- Chronic Kidney Disease, George Institute for Global Health India, UNSW, New Delhi, India
- Prasanna School of Public Health, Manipal Academy of Higher Education, Manipal, Karnataka, India
- Department of Medicine, School of Public Health, Imperial College, London, UK
| |
Collapse
|
35
|
Kiryluk K, Sanchez-Rodriguez E, Zhou XJ, Zanoni F, Liu L, Mladkova N, Khan A, Marasa M, Zhang JY, Balderes O, Sanna-Cherchi S, Bomback AS, Canetta PA, Appel GB, Radhakrishnan J, Trimarchi H, Sprangers B, Cattran DC, Reich H, Pei Y, Ravani P, Galesic K, Maixnerova D, Tesar V, Stengel B, Metzger M, Canaud G, Maillard N, Berthoux F, Berthelot L, Pillebout E, Monteiro R, Nelson R, Wyatt RJ, Smoyer W, Mahan J, Samhar AA, Hidalgo G, Quiroga A, Weng P, Sreedharan R, Selewski D, Davis K, Kallash M, Vasylyeva TL, Rheault M, Chishti A, Ranch D, Wenderfer SE, Samsonov D, Claes DJ, Akchurin O, Goumenos D, Stangou M, Nagy J, Kovacs T, Fiaccadori E, Amoroso A, Barlassina C, Cusi D, Del Vecchio L, Battaglia GG, Bodria M, Boer E, Bono L, Boscutti G, Caridi G, Lugani F, Ghiggeri G, Coppo R, Peruzzi L, Esposito V, Esposito C, Feriozzi S, Polci R, Frasca G, Galliani M, Garozzo M, Mitrotti A, Gesualdo L, Granata S, Zaza G, Londrino F, Magistroni R, Pisani I, Magnano A, Marcantoni C, Messa P, Mignani R, Pani A, Ponticelli C, Roccatello D, Salvadori M, Salvi E, Santoro D, Gembillo G, Savoldi S, Spotti D, Zamboli P, Izzi C, et alKiryluk K, Sanchez-Rodriguez E, Zhou XJ, Zanoni F, Liu L, Mladkova N, Khan A, Marasa M, Zhang JY, Balderes O, Sanna-Cherchi S, Bomback AS, Canetta PA, Appel GB, Radhakrishnan J, Trimarchi H, Sprangers B, Cattran DC, Reich H, Pei Y, Ravani P, Galesic K, Maixnerova D, Tesar V, Stengel B, Metzger M, Canaud G, Maillard N, Berthoux F, Berthelot L, Pillebout E, Monteiro R, Nelson R, Wyatt RJ, Smoyer W, Mahan J, Samhar AA, Hidalgo G, Quiroga A, Weng P, Sreedharan R, Selewski D, Davis K, Kallash M, Vasylyeva TL, Rheault M, Chishti A, Ranch D, Wenderfer SE, Samsonov D, Claes DJ, Akchurin O, Goumenos D, Stangou M, Nagy J, Kovacs T, Fiaccadori E, Amoroso A, Barlassina C, Cusi D, Del Vecchio L, Battaglia GG, Bodria M, Boer E, Bono L, Boscutti G, Caridi G, Lugani F, Ghiggeri G, Coppo R, Peruzzi L, Esposito V, Esposito C, Feriozzi S, Polci R, Frasca G, Galliani M, Garozzo M, Mitrotti A, Gesualdo L, Granata S, Zaza G, Londrino F, Magistroni R, Pisani I, Magnano A, Marcantoni C, Messa P, Mignani R, Pani A, Ponticelli C, Roccatello D, Salvadori M, Salvi E, Santoro D, Gembillo G, Savoldi S, Spotti D, Zamboli P, Izzi C, Alberici F, Delbarba E, Florczak M, Krata N, Mucha K, Pączek L, Niemczyk S, Moszczuk B, Pańczyk-Tomaszewska M, Mizerska-Wasiak M, Perkowska-Ptasińska A, Bączkowska T, Durlik M, Pawlaczyk K, Sikora P, Zaniew M, Kaminska D, Krajewska M, Kuzmiuk-Glembin I, Heleniak Z, Bullo-Piontecka B, Liberek T, Dębska-Slizien A, Hryszko T, Materna-Kiryluk A, Miklaszewska M, Szczepańska M, Dyga K, Machura E, Siniewicz-Luzeńczyk K, Pawlak-Bratkowska M, Tkaczyk M, Runowski D, Kwella N, Drożdż D, Habura I, Kronenberg F, Prikhodina L, van Heel D, Fontaine B, Cotsapas C, Wijmenga C, Franke A, Annese V, Gregersen PK, Parameswaran S, Weirauch M, Kottyan L, Harley JB, Suzuki H, Narita I, Goto S, Lee H, Kim DK, Kim YS, Park JH, Cho B, Choi M, Van Wijk A, Huerta A, Ars E, Ballarin J, Lundberg S, Vogt B, Mani LY, Caliskan Y, Barratt J, Abeygunaratne T, Kalra PA, Gale DP, Panzer U, Rauen T, Floege J, Schlosser P, Ekici AB, Eckardt KU, Chen N, Xie J, Lifton RP, Loos RJF, Kenny EE, Ionita-Laza I, Köttgen A, Julian BA, Novak J, Scolari F, Zhang H, Gharavi AG. Genome-wide association analyses define pathogenic signaling pathways and prioritize drug targets for IgA nephropathy. Nat Genet 2023; 55:1091-1105. [PMID: 37337107 PMCID: PMC11824687 DOI: 10.1038/s41588-023-01422-x] [Show More Authors] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 05/05/2023] [Indexed: 06/21/2023]
Abstract
IgA nephropathy (IgAN) is a progressive form of kidney disease defined by glomerular deposition of IgA. Here we performed a genome-wide association study of 10,146 kidney-biopsy-diagnosed IgAN cases and 28,751 controls across 17 international cohorts. We defined 30 genome-wide significant risk loci explaining 11% of disease risk. A total of 16 loci were new, including TNFSF4/TNFSF18, REL, CD28, PF4V1, LY86, LYN, ANXA3, TNFSF8/TNFSF15, REEP3, ZMIZ1, OVOL1/RELA, ETS1, IGH, IRF8, TNFRSF13B and FCAR. The risk loci were enriched in gene orthologs causing abnormal IgA levels when genetically manipulated in mice. We also observed a positive genetic correlation between IgAN and serum IgA levels. High polygenic score for IgAN was associated with earlier onset of kidney failure. In a comprehensive functional annotation analysis of candidate causal genes, we observed convergence of biological candidates on a common set of inflammatory signaling pathways and cytokine ligand-receptor pairs, prioritizing potential new drug targets.
Collapse
Affiliation(s)
- Krzysztof Kiryluk
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA.
- Institute for Genomic Medicine, Columbia University, New York City, NY, USA.
| | - Elena Sanchez-Rodriguez
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Xu-Jie Zhou
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Francesca Zanoni
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Lili Liu
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Nikol Mladkova
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Atlas Khan
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Maddalena Marasa
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Jun Y Zhang
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Olivia Balderes
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Simone Sanna-Cherchi
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
- Institute for Genomic Medicine, Columbia University, New York City, NY, USA
| | - Andrew S Bomback
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Pietro A Canetta
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Gerald B Appel
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Jai Radhakrishnan
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Hernan Trimarchi
- Nephrology Service, Hospital Británico de Buenos Aires, Buenos Aires, Argentina
| | - Ben Sprangers
- Department of Microbiology and Immunology, Laboratory of Molecular Immunology, KU Leuven, Leuven, Belgium
- Division of Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Daniel C Cattran
- Department of Nephrology, University of Toronto, Toronto General Hospital, Toronto, Ontario, Canada
| | - Heather Reich
- Department of Nephrology, University of Toronto, Toronto General Hospital, Toronto, Ontario, Canada
| | - York Pei
- Department of Nephrology, University of Toronto, Toronto General Hospital, Toronto, Ontario, Canada
| | - Pietro Ravani
- Division of Nephrology, Department of Internal Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Dita Maixnerova
- 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Vladimir Tesar
- 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Benedicte Stengel
- Centre for Research in Epidemiology and Population Health (CESP), Paris-Saclay University, Versailles Saint Quentin University, INSERM Clinical Epidemiology Team, Villejuif, France
| | - Marie Metzger
- Centre for Research in Epidemiology and Population Health (CESP), Paris-Saclay University, Versailles Saint Quentin University, INSERM Clinical Epidemiology Team, Villejuif, France
| | - Guillaume Canaud
- Université de Paris, Hôpital Necker-Enfants Malades, Paris, France
| | - Nicolas Maillard
- Nephrology, Dialysis, and Renal Transplantation Department, University North Hospital, Saint Etienne, France
| | - Francois Berthoux
- Nephrology, Dialysis, and Renal Transplantation Department, University North Hospital, Saint Etienne, France
| | | | - Evangeline Pillebout
- Center for Research on Inflammation, University of Paris, INSERM and CNRS, Paris, France
| | - Renato Monteiro
- Center for Research on Inflammation, University of Paris, INSERM and CNRS, Paris, France
| | - Raoul Nelson
- Division of Pediatric Nephrology, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Robert J Wyatt
- Division of Pediatric Nephrology, University of Tennessee Health Sciences Center, Memphis, TN, USA
- Children's Foundation Research Center, Le Bonheur Children's Hospital, Memphis, TN, USA
| | - William Smoyer
- Division of Pediatric Nephrology, Nationwide Children's Hospital, Columbus, OH, USA
| | - John Mahan
- Division of Pediatric Nephrology, Nationwide Children's Hospital, Columbus, OH, USA
| | - Al-Akash Samhar
- Division of Pediatric Nephrology, Driscoll Children's Hospital, Corpus Christi, TX, USA
| | - Guillermo Hidalgo
- Division of Pediatric Nephrology, Department of Pediatrics, HMH Hackensack University Medical Center, Hackensack, NJ, USA
| | - Alejandro Quiroga
- Division of Pediatric Nephrology, Helen DeVos Children's Hospital, Grand Rapids, MI, USA
| | - Patricia Weng
- Division of Pediatric Nephrology, Mattel Children's Hospital, Los Angeles, CA, USA
| | - Raji Sreedharan
- Division of Pediatric Nephrology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - David Selewski
- Division of Pediatric Nephrology, Mott Children's Hospital, Ann Arbor, MI, USA
| | - Keefe Davis
- Division of Pediatric Nephrology, Department of Pediatrics, The Medical University of South Carolina (MUSC), Charleston, SC, USA
| | - Mahmoud Kallash
- Division of Pediatric Nephrology, SUNY Buffalo, Buffalo, NY, USA
| | - Tetyana L Vasylyeva
- Division of Pediatric Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, OH, USA
| | - Michelle Rheault
- Division of Pediatric Nephrology, University of Minnesota, Minneapolis, MN, USA
| | - Aftab Chishti
- Division of Pediatric Nephrology, University of Kentucky, Lexington, KY, USA
| | - Daniel Ranch
- Division of Pediatric Nephrology, Department of Pediatrics, University of Kentucky, Lexington, KY, USA
| | - Scott E Wenderfer
- Division of Pediatric Nephrology, Baylor College of Medicine/Texas Children's Hospital, Houston, TX, USA
| | - Dmitry Samsonov
- Division of Pediatric Nephrology, Boston Children's Hospital, Boston, MA, USA
| | - Donna J Claes
- Division of Pediatric Nephrology, Department of Pediatrics, New York Medical College, New York City, NY, USA
| | - Oleh Akchurin
- Division of Pediatric Nephrology, Department of Pediatrics, Weill Cornell Medical College, New York City, NY, USA
| | | | - Maria Stangou
- The Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Judit Nagy
- 2nd Department of Internal Medicine, Nephrological and Diabetological Center, University of Pécs, Pécs, Hungary
| | - Tibor Kovacs
- 2nd Department of Internal Medicine, Nephrological and Diabetological Center, University of Pécs, Pécs, Hungary
| | - Enrico Fiaccadori
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Antonio Amoroso
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Cristina Barlassina
- Renal Division, Dipartimento di Medicina, Chirurgia e Odontoiatria, San Paolo Hospital, School of Medicine, University of Milan, Milan, Italy
| | - Daniele Cusi
- Renal Division, Dipartimento di Medicina, Chirurgia e Odontoiatria, San Paolo Hospital, School of Medicine, University of Milan, Milan, Italy
| | | | | | | | - Emanuela Boer
- Division of Nephrology and Dialysis, Gorizia Hospital, Gorizia, Italy
| | - Luisa Bono
- Nephrology and Dialysis, A.R.N.A.S. Civico and Benfratelli, Palermo, Italy
| | - Giuliano Boscutti
- Nephrology, Dialysis and Renal Transplant Unit, S. Maria della Misericordia Hospital, ASUFC, Udine, Italy
| | - Gianluca Caridi
- Division of Nephrology, Dialysis and Transplantation, IRCCS Giannina Gaslini Institute, Genova, Italy
| | - Francesca Lugani
- Division of Nephrology, Dialysis and Transplantation, IRCCS Giannina Gaslini Institute, Genova, Italy
| | - GianMarco Ghiggeri
- Division of Nephrology, Dialysis and Transplantation, IRCCS Giannina Gaslini Institute, Genova, Italy
| | - Rosanna Coppo
- Regina Margherita Children's Hospital, Torino, Italy
| | - Licia Peruzzi
- Regina Margherita Children's Hospital, Torino, Italy
| | | | | | | | | | - Giovanni Frasca
- Division of Nephrology, Dialysis and Renal Transplantation, Riuniti Hospital, Ancona, Italy
| | | | - Maurizio Garozzo
- Unità Operativa di Nefrologia e Dialisi, Ospedale di Acireale, Acireale, Italy
| | - Adele Mitrotti
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Simona Granata
- Renal Unit, Department of Medicine, University of Verona, Verona, Italy
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University of Verona, Verona, Italy
| | | | - Riccardo Magistroni
- Department of Surgical, Medical, Dental, Oncologic and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Isabella Pisani
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Andrea Magnano
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Piergiorgio Messa
- Nephrology Dialysis and Kidney Transplant Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - Renzo Mignani
- Azienda Unità Sanitaria Locale Rimini, Rimini, Italy
| | - Antonello Pani
- Department of Nephrology and Dialysis, G. Brotzu Hospital, Cagliari, Italy
| | | | - Dario Roccatello
- Nephrology and Dialysis Unit, G. Bosco Hub Hospital (ERK-net Member) and University of Torino, Torino, Italy
| | - Maurizio Salvadori
- Division of Nephrology and Renal Transplantation, Carreggi Hospital, Florence, Italy
| | - Erica Salvi
- Renal Division, DMCO (Dipartimento di Medicina, Chirurgia e Odontoiatria), San Paolo Hospital, School of Medicine, University of Milan, Milan, Italy
| | - Domenico Santoro
- Unit of Nephrology and Dialysis, AOU G Martino, University of Messina, Messina, Italy
| | - Guido Gembillo
- Unit of Nephrology and Dialysis, AOU G Martino, University of Messina, Messina, Italy
| | - Silvana Savoldi
- Unit of Nephrology and Dialysis, ASL TO4-Consultorio Cirié, Turin, Italy
| | | | | | - Claudia Izzi
- Department of Medical and Surgical Specialties and Nephrology Unit, University of Brescia-ASST Spedali Civili, Brescia, Italy
| | - Federico Alberici
- Department of Medical and Surgical Specialties and Nephrology Unit, University of Brescia-ASST Spedali Civili, Brescia, Italy
| | - Elisa Delbarba
- Department of Medical and Surgical Specialties and Nephrology Unit, University of Brescia-ASST Spedali Civili, Brescia, Italy
| | - Michał Florczak
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Natalia Krata
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Krzysztof Mucha
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Leszek Pączek
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Stanisław Niemczyk
- Department of Internal Disease, Nephrology and Dialysotherapy, Military Institute of Medicine, Warsaw, Poland
| | - Barbara Moszczuk
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
- Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland
| | | | | | | | - Teresa Bączkowska
- Department of Transplantation Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Magdalena Durlik
- Department of Transplantation Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Krzysztof Pawlaczyk
- Department of Nephrology, Transplantology and Internal Medicine, Poznan Medical University, Poznan, Poland
| | - Przemyslaw Sikora
- Department of Pediatric Nephrology, Medical University of Lublin, Lublin, Poland
| | - Marcin Zaniew
- Department of Pediatrics, University of Zielona Góra, Zielona Góra, Poland
| | - Dorota Kaminska
- Clinical Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Magdalena Krajewska
- Clinical Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Izabella Kuzmiuk-Glembin
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdansk, Gdansk, Poland
| | - Zbigniew Heleniak
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdansk, Gdansk, Poland
| | - Barbara Bullo-Piontecka
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdansk, Gdansk, Poland
| | - Tomasz Liberek
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdansk, Gdansk, Poland
| | - Alicja Dębska-Slizien
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdansk, Gdansk, Poland
| | - Tomasz Hryszko
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Bialystok, Bialystok, Poland
| | | | - Monika Miklaszewska
- Department of Pediatric Nephrology and Hypertension, Jagiellonian University Medical College, Krakow, Poland
| | - Maria Szczepańska
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
| | - Katarzyna Dyga
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
| | - Edyta Machura
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
| | - Katarzyna Siniewicz-Luzeńczyk
- Department of Pediatrics, Immunology and Nephrology, Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Monika Pawlak-Bratkowska
- Department of Pediatrics, Immunology and Nephrology, Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Marcin Tkaczyk
- Department of Pediatrics, Immunology and Nephrology, Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Dariusz Runowski
- Department of Nephrology, Kidney Transplantation and Hypertension, Children's Memorial Health Institute, Warsaw, Poland
| | - Norbert Kwella
- Department of Nephrology, Hypertension and Internal Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Dorota Drożdż
- Department of Pediatric Nephrology and Hypertension, Jagiellonian University Medical College, Krakow, Poland
| | - Ireneusz Habura
- Department of Nephrology, Karol Marcinkowski Hospital, Zielona Góra, Poland
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Larisa Prikhodina
- Division of Inherited and Acquired Kidney Diseases, Veltischev Research and Clinical Institute for Pediatrics of the Pirogov Russian National Research Medical University, Moscow, Russia
| | - David van Heel
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Bertrand Fontaine
- Sorbonne University, INSERM, Center of Research in Myology, Institute of Myology, University Hospital Pitie-Salpetriere, Paris, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Service of Neuro-Myology, University Hospital Pitie-Salpetriere, Paris, France
| | - Chris Cotsapas
- Departments of Neurology and Genetics, Yale University, New Haven, CT, USA
| | | | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Vito Annese
- CBP American Hospital, Dubai, United Arab Emirates
| | - Peter K Gregersen
- Robert S. Boas Center for Genomics and Human Genetics, Feinstein Institutes for Medical Research, North Shore LIJ Health System, New York City, NY, USA
| | | | - Matthew Weirauch
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Leah Kottyan
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - John B Harley
- US Department of Veterans Affairs Medical Center and Cincinnati Education and Research for Veterans Foundation, Cincinnati, OH, USA
| | - Hitoshi Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Ichiei Narita
- Division of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Shin Goto
- Division of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hajeong Lee
- Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dong Ki Kim
- Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yon Su Kim
- Biomedical Science, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin-Ho Park
- Department of Family Medicine, Seoul National University College of Medicine and Seoul National University Hospital, Seoul, Republic of Korea
| | - BeLong Cho
- Department of Family Medicine, Seoul National University College of Medicine and Seoul National University Hospital, Seoul, Republic of Korea
- Institute on Aging, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Murim Choi
- Biomedical Science, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ans Van Wijk
- Amsterdam University Medical Centre, VU University Medical Center (VUMC), Amsterdam, the Netherlands
| | - Ana Huerta
- Hospital Universitario Puerta del Hierro Majadahonda, REDINREN, IISCIII, Madrid, Spain
| | - Elisabet Ars
- Molecular Biology Laboratory and Nephrology Department, Fundació Puigvert, Instituto de Investigaciones Biomédicas Sant Pau, Universitat Autònoma de Barcelona, REDINREN, IISCIII, Barcelona, Spain
| | - Jose Ballarin
- Molecular Biology Laboratory and Nephrology Department, Fundació Puigvert, Instituto de Investigaciones Biomédicas Sant Pau, Universitat Autònoma de Barcelona, REDINREN, IISCIII, Barcelona, Spain
| | - Sigrid Lundberg
- Department of Nephrology, Danderyd University Hospital, and Department of Clinical Sciences, Karolinska Institutet, Stockholm, Sweden
| | - Bruno Vogt
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Laila-Yasmin Mani
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Yasar Caliskan
- Division of Nephrology, Saint Louis University, Saint Louis, MO, USA
| | - Jonathan Barratt
- John Walls Renal Unit, University Hospitals of Leicester, Leicester, UK
| | | | | | - Daniel P Gale
- Department of Renal Medicine, University College London, London, UK
| | | | - Thomas Rauen
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Jürgen Floege
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Pascal Schlosser
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Arif B Ekici
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Nan Chen
- Department of Nephrology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jingyuan Xie
- Department of Nephrology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Richard P Lifton
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York City, NY, USA
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Eimear E Kenny
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- Department of Genetics and Genomic Sciences, Mount Sinai Health System, New York City, NY, USA
- Center for Population Genomic Health, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Iuliana Ionita-Laza
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York City, NY, USA
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Bruce A Julian
- Departments of Microbiology and Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jan Novak
- Departments of Microbiology and Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Francesco Scolari
- Department of Medical and Surgical Specialties and Nephrology Unit, University of Brescia-ASST Spedali Civili, Brescia, Italy
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Ali G Gharavi
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA.
- Institute for Genomic Medicine, Columbia University, New York City, NY, USA.
| |
Collapse
|
36
|
Mucha K, Pac M, Pączek L. Omics are Getting Us Closer to Understanding IgA Nephropathy. Arch Immunol Ther Exp (Warsz) 2023; 71:12. [PMID: 37060455 PMCID: PMC10105675 DOI: 10.1007/s00005-023-00677-w] [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: 10/23/2021] [Accepted: 03/02/2023] [Indexed: 04/16/2023]
Abstract
During the last decade, thanks to omics technologies, new light has been shed on the pathogenesis of many diseases. Genomics, epigenomics, transcriptomics, and proteomics have helped to provide a better understanding of the origin and heterogeneity of several diseases. However, the risk factors for most autoimmune diseases remain unknown. The successes and pitfalls of omics have also been observed in nephrology, including immunoglobulin A nephropathy (IgAN), the most common form of glomerulonephritis and a principal cause of end-stage renal disease worldwide. Unfortunately, the immense progress in basic research has not yet been followed by the satisfactory development of a targeted treatment. Although, most omics studies describe changes in the immune system, there is still insufficient data to apply their results in the constantly evolving multi-hit pathogenesis model and thus do to provide a complete picture of the disease. Here, we describe recent findings regarding the pathophysiology of IgAN and link omics studies with immune system dysregulation. This review provides insights into specific IgAN markers, which may lead to the identification of potential targets for personalised treatment in the future.
Collapse
Affiliation(s)
- Krzysztof Mucha
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland.
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.
| | - Michał Pac
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Leszek Pączek
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
37
|
Novak J, Renfrow MB, King RG, Reily C, Green TJ. Protein-based profiling of the human IgA1 clonal repertoire revealed shared clones of serum polymeric IgA1 and milk secretory IgA1. Cell Mol Immunol 2023; 20:305-307. [PMID: 36596872 PMCID: PMC9971200 DOI: 10.1038/s41423-022-00965-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 01/05/2023] Open
Grants
- R01 GM098539 NIGMS NIH HHS
- R01 AI149431 NIAID NIH HHS
- K01 DK106341 NIDDK NIH HHS
- R01 AI162236 NIAID NIH HHS
- R01 DK078244 NIDDK NIH HHS
- R01 DK082753 NIDDK NIH HHS
- R56 DK078244 NIDDK NIH HHS
- NIH: AI149431, GM098539, DK078244, DK082753, DK106341, and AI162236
- NIH: AI149431, GM098539, DK078244, DK082753, and AI162236
- NIH: DK106341
- NIH: AI149431, DK082753, DK106341
Collapse
Affiliation(s)
- Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Matthew B Renfrow
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - R Glenn King
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Colin Reily
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Todd J Green
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
38
|
Mestecky J, Julian BA, Raska M. IgA Nephropathy: Pleiotropic impact of Epstein-Barr virus infection on immunopathogenesis and racial incidence of the disease. Front Immunol 2023; 14:1085922. [PMID: 36865536 PMCID: PMC9973316 DOI: 10.3389/fimmu.2023.1085922] [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: 10/31/2022] [Accepted: 01/25/2023] [Indexed: 02/09/2023] Open
Abstract
IgA nephropathy (IgAN) is an autoimmune disease in which poorly galactosylated IgA1 is the antigen recognized by naturally occurring anti-glycan antibodies, leading to formation of nephritogenic circulating immune complexes. Incidence of IgAN displays geographical and racial disparity: common in Europe, North America, Australia, and east Asia, uncommon in African Americans, many Asian and South American countries, Australian Aborigines, and rare in central Africa. In analyses of sera and cells from White IgAN patients, healthy controls, and African Americans, IgAN patients exhibited substantial enrichment for IgA-expressing B cells infected with Epstein-Barr virus (EBV), leading to enhanced production of poorly galactosylated IgA1. Disparities in incidence of IgAN may reflect a previously disregarded difference in the maturation of the IgA system as related to the timing of EBV infection. Compared with populations with higher incidences of IgAN, African Americans, African Blacks, and Australian Aborigines are more frequently infected with EBV during the first 1-2 years of life at the time of naturally occurring IgA deficiency when IgA cells are less numerous than in late childhood or adolescence. Therefore, in very young children EBV enters "non-IgA" cells. Ensuing immune responses prevent infection of IgA B cells during later exposure to EBV at older ages. Our data implicate EBV-infected cells as the source of poorly galactosylated IgA1 in circulating immune complexes and glomerular deposits in patients with IgAN. Thus, temporal differences in EBV primo-infection as related to naturally delayed maturation of the IgA system may contribute to geographic and racial variations in incidence of IgAN.
Collapse
Affiliation(s)
- Jiri Mestecky
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
- Laboratory of Cellular and Molecular Immunology Institute of Microbiology, Czech Academy of Sciences, Prague, Czechia
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Bruce A. Julian
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Milan Raska
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, Czechia
| |
Collapse
|