1
|
Gembillo G, Sessa C, Santoro D. Advances in the pathophysiology and treatment of focal segmental glomerulosclerosis: The importance of a timely and tailored approach. World J Nephrol 2025; 14. [DOI: 10.5527/wjn.v14.i2.103039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/14/2025] [Accepted: 01/21/2025] [Indexed: 04/09/2025] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) is a histological pattern of glomerular damage that significantly contributes to chronic kidney disease and end-stage renal disease. Its incidence is rising globally, necessitating timely and personalized management strategies. This paper aims to provide an updated overview of the pathophysiology, diagnosis, and therapeutic strategies for FSGS, emphasizing the importance of early interventions and tailored treatments. This editorial synthesizes key findings from recent literature to highlight advancements in understanding and managing FSGS. Emerging evidence supports the role of targeted therapies and personalized approaches in improving outcomes for FSGS patients. Advances include novel biomarkers, genetic testing, and innovative therapeutics such as transient receptor potential ion channel blockers and antisense oligonucleotides for apolipoprotein 1-related FSGS. Effective management of FSGS requires a combination of timely diagnosis, evidence-based therapeutic strategies, and ongoing research to optimize patient outcomes and address gaps in the current understanding of the disease.
Collapse
Affiliation(s)
- Guido Gembillo
- Unit of Nephrology and Dialysis, AOU "G. Martino", University of Messina, Messina 98125, Sicilia, Italy
| | - Concetto Sessa
- Unit of Nephrology and Dialysis, P.O. Maggiore "Nino Baglieri", Ragusa 97100, Sicilia, Italy
| | - Domenico Santoro
- Unit of Nephrology and Dialysis, AOU "G. Martino", University of Messina, Messina 98125, Sicilia, Italy
| |
Collapse
|
2
|
Gupta AK, Minocha E, Koss KM, Naved BA, Safar-Boueri L, Wertheim JA, Gallon L. A kidney organoid-based readout to assess disease activity in primary and recurrent focal segmental glomerulosclerosis. Kidney Int 2025; 107:888-902. [PMID: 39914654 DOI: 10.1016/j.kint.2025.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 02/23/2025]
Abstract
Primary focal segmental glomerulosclerosis (pFSGS) is an acquired kidney disorder that frequently leads to kidney failure and confers an elevated risk of recurrence after kidney transplantation, termed recurrent pFSGS. Unfortunately, there is no diagnostic method to foresee recurrence of pFSGS after kidney transplantation. Progress in developing assays to test disease activity is hampered by few preclinical models to replicate disease and inability of in vitro cultured primary podocytes to remain terminally differentiated. In recent years, advancements in kidney organoid biology have led to the development of kidney tissues with glomeruli and major nephron segments including podocytes. To develop a pFSGS model, we studied the effect of plasma from patients diagnosed with pFSGS on kidney organoids differentiated from human pluripotent stem cells. The pFSGS plasma treatment induced podocytopathy, extracellular matrix protein deposition, fibrosis and apoptosis within organoids, whereas non-recurrent plasma did not affect organoid structure. pFSGS plasma also led to loss of normal expression patterns of podocyte specific proteins, nephrin and podocin within podocytes. Further, cytokine array profiling revealed that pFSGS plasma induced secretion of cytokines associated with inflammation and angiogenesis. Additionally, kidney organoids treated with plasma obtained after therapeutic plasma exchange for recurrent pFSGS led to lower cell death in organoids after sequential exchanges with the final exchange showing the least apoptotic cells without morphological abnormality. Overall, our results demonstrate the potential of kidney organoids in advancing kidney disease modeling. These insights could be applied in clinical settings to assist in gauging FSGS recurrence risk prior to kidney transplantation.
Collapse
Affiliation(s)
- Ashwani Kumar Gupta
- Department of Surgery, University of Arizona College of Medicine, Tucson, Arizona, USA; Bio5 Institute, University of Arizona, Tucson, Arizona, USA; Surgery Service, Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona, USA; Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ekta Minocha
- Department of Surgery, University of Arizona College of Medicine, Tucson, Arizona, USA; Bio5 Institute, University of Arizona, Tucson, Arizona, USA; Surgery Service, Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona, USA
| | - Kyle M Koss
- Department of Surgery, University of Arizona College of Medicine, Tucson, Arizona, USA; Bio5 Institute, University of Arizona, Tucson, Arizona, USA; Surgery Service, Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona, USA; Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Neurobiology, University of Texas Medical Branch, Galveston, Texas, USA; Sealy Institute for Drug Discovery, University of Texas Medical Branch, Galveston, Texas, USA
| | - Bilal A Naved
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, USA
| | - Luisa Safar-Boueri
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jason A Wertheim
- Department of Surgery, University of Arizona College of Medicine, Tucson, Arizona, USA; Bio5 Institute, University of Arizona, Tucson, Arizona, USA; Surgery Service, Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona, USA; Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, USA; Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, USA.
| | - Lorenzo Gallon
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Medicine, Abdominal Organ Transplant Program, University of Illinois Chicago, Chicago, Illinois, USA.
| |
Collapse
|
3
|
Maslyennikov Y, Bărar AA, Rusu CC, Potra AR, Tirinescu D, Ticala M, Urs A, Pralea IE, Iuga CA, Moldovan DT, Kacso IM. The Spectrum of Minimal Change Disease/Focal Segmental Glomerulosclerosis: From Pathogenesis to Proteomic Biomarker Research. Int J Mol Sci 2025; 26:2450. [PMID: 40141093 PMCID: PMC11941885 DOI: 10.3390/ijms26062450] [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: 01/18/2025] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Podocyte injury plays a central role in both focal segmental glomerulosclerosis (FSGS) and minimal change disease (MCD). Pathogenic mechanisms are diverse and incompletely understood, partially overlap between FSGS and MCD, and are not reflected by kidney biopsy. In order to optimize the current variable response to treatment, personalized management should rely on pathogenesis. One promising approach involves identifying biomarkers associated with specific pathogenic pathways. With the advancement of technology, proteomic studies could be a valuable tool to improve knowledge in this area and define valid biomarkers, as they have in other areas of glomerular disease. This work attempts to cover and discuss the main mechanisms of podocyte injury, followed by a review of the recent literature on proteomic biomarker studies in podocytopathies. Most of these studies have been conducted on biofluids, while tissue proteomic studies applied to podocytopathies remain limited. While we recognize the importance of non-invasive biofluid biomarkers, we propose a sequential approach for their development: tissue proteomics could first identify proteins with increased expression that may reflect underlying disease mechanisms; subsequently, the validation of these proteins in urine or plasma could pave the way to a diagnostic and prognostic biomarker-based approach.
Collapse
Affiliation(s)
- Yuriy Maslyennikov
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (Y.M.); (A.A.B.); (C.C.R.); (A.R.P.); (D.T.); (M.T.); (A.U.); (I.M.K.)
| | - Andrada Alina Bărar
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (Y.M.); (A.A.B.); (C.C.R.); (A.R.P.); (D.T.); (M.T.); (A.U.); (I.M.K.)
| | - Crina Claudia Rusu
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (Y.M.); (A.A.B.); (C.C.R.); (A.R.P.); (D.T.); (M.T.); (A.U.); (I.M.K.)
| | - Alina Ramona Potra
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (Y.M.); (A.A.B.); (C.C.R.); (A.R.P.); (D.T.); (M.T.); (A.U.); (I.M.K.)
| | - Dacian Tirinescu
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (Y.M.); (A.A.B.); (C.C.R.); (A.R.P.); (D.T.); (M.T.); (A.U.); (I.M.K.)
| | - Maria Ticala
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (Y.M.); (A.A.B.); (C.C.R.); (A.R.P.); (D.T.); (M.T.); (A.U.); (I.M.K.)
| | - Alexandra Urs
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (Y.M.); (A.A.B.); (C.C.R.); (A.R.P.); (D.T.); (M.T.); (A.U.); (I.M.K.)
| | - Ioana Ecaterina Pralea
- Department of Personalized Medicine and Rare Diseases, MedFuture—Research Centre for Biomedical Research, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (I.E.P.); (C.A.I.)
| | - Cristina Adela Iuga
- Department of Personalized Medicine and Rare Diseases, MedFuture—Research Centre for Biomedical Research, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (I.E.P.); (C.A.I.)
- Department of Drug Analysis, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Diana Tania Moldovan
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (Y.M.); (A.A.B.); (C.C.R.); (A.R.P.); (D.T.); (M.T.); (A.U.); (I.M.K.)
| | - Ina Maria Kacso
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (Y.M.); (A.A.B.); (C.C.R.); (A.R.P.); (D.T.); (M.T.); (A.U.); (I.M.K.)
| |
Collapse
|
4
|
Guaragna MS, Casimiro FMS, Varela P, de S Feltran L, Watanabe A, Neves PDMM, Pesquero JB, Belangero VMS, Nogueira PCK, Onuchic LF. Past and future in vitro and in vivo approaches toward circulating factors and biomarkers in idiopathic nephrotic syndrome. Pediatr Nephrol 2025:10.1007/s00467-024-06643-8. [PMID: 39883133 DOI: 10.1007/s00467-024-06643-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 11/26/2024] [Accepted: 12/09/2024] [Indexed: 01/31/2025]
Abstract
Predicting the risks of progression to chronic kidney disease (CKD) stage 5 in idiopathic nephrotic syndrome (NS) and recurrence of the disease (rNS) following kidney transplantation (KT) is a key assessment to provide essential management information. NS has been categorized etiologically as genetic and immune-based. A genetic cause can be identified in ~ 30% of children with steroid-resistant NS (SRNS), a finding associated with a very low risk of rNS following KT. In immune-based NS, clinical overlap is observed among steroid-sensitive NS, secondary-resistant NS, and SRNS not associated with disease-causing genetic variants (non-monogenic SRNS). While ~ 50% of SRNS patients with no identified monogenic disease respond to intensified immunosuppressive treatments, the ones that do not respond to this therapy have a high risk of progression to CKD stage 5 and post-KT rNS. Secondary-resistant patients who progress to CKD stage 5 display the highest risk of post-KT rNS. The proposed shared underlying mechanism of the immune-based NS associated with post-KT rNS is based on a systemic circulating factor (CF) that affects glomerular permeability by inducing foot process effacement and focal segmental glomerulosclerosis. However, identifying patients without a detected genetic form who will recur post-KT is a major challenge. Extensive efforts, therefore, have been made to identify CFs and biomarkers potentially capable of predicting the risk of progression to CKD stage 5 and post-KT rNS. This review discusses the in vitro and in vivo approaches employed to date to identify and characterize potential CFs and CF-induced biomarkers of recurrent NS and offers an assessment of their potential to improve outcomes of KT in this patient population.
Collapse
Affiliation(s)
- Mara S Guaragna
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, State University of Campinas, Campinas, Brazil
- Center for Molecular Biology and Genetic Engineering, State University of Campinas, Campinas, Brazil
| | - Fernanda M S Casimiro
- Center for Diagnosis and Research On Genetic Diseases, Department of Biophysics, Federal University of São Paulo, São Paulo, Brazil
| | - Patrícia Varela
- Center for Diagnosis and Research On Genetic Diseases, Department of Biophysics, Federal University of São Paulo, São Paulo, Brazil
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luciana de S Feltran
- Division of Pediatric Kidney Transplantation, São Paulo Samaritan Hospital, São Paulo, Brazil
| | - Andreia Watanabe
- Department of Pediatrics, University of São Paulo School of Medicine, São Paulo, Brazil
- Division of Molecular Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Precil D M M Neves
- Division of Molecular Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
- Division of Nephrology, University of São Paulo School of Medicine, Avenida Dr. Arnaldo, 455 - Sala 4304, São Paulo, SP, 01246-903, Brazil
| | - João B Pesquero
- Center for Diagnosis and Research On Genetic Diseases, Department of Biophysics, Federal University of São Paulo, São Paulo, Brazil
| | - Vera M S Belangero
- Department of Pediatrics, School of Medical Sciences, State University of Campinas, Campinas, Brazil
| | - Paulo C K Nogueira
- Division of Pediatric Kidney Transplantation, São Paulo Samaritan Hospital, São Paulo, Brazil
- Department of Pediatric Nephrology, São Paulo Federal University, São Paulo, Brazil
| | - Luiz F Onuchic
- Division of Molecular Medicine, University of São Paulo School of Medicine, São Paulo, Brazil.
- Division of Nephrology, University of São Paulo School of Medicine, Avenida Dr. Arnaldo, 455 - Sala 4304, São Paulo, SP, 01246-903, Brazil.
| |
Collapse
|
5
|
Xie Y, Liu F. Precision medicine for focal segmental glomerulosclerosis. Kidney Res Clin Pract 2024; 43:709-723. [PMID: 38325863 PMCID: PMC11615440 DOI: 10.23876/j.krcp.23.227] [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: 08/24/2023] [Revised: 11/06/2023] [Accepted: 11/15/2023] [Indexed: 02/09/2024] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) is one of the common causes of nephrotic syndrome in adults and children worldwide. FSGS consists of a group of kidney diseases classified based on specific histopathological features. The current classification of FSGS makes it difficult to distinguish individual differences in pathogenesis, disease progression, and response to treatment. In recent years, the spread of next-generation sequencing, updates in biological techniques, and improvements of treatment have changed our understanding of FSGS. In this review, we will discuss the use of genetic testing in patients with FSGS, explore its clinical significance from a genetic identification perspective, and introduce several new biomarkers, that may help in the early diagnosis of FSGS and guide the development of specific or targeted therapies, so as to understand the biological characteristics in FSGS. This will certainly help develop more effective and safer treatments and advance precision medicine.
Collapse
Affiliation(s)
- Yi Xie
- Department of Nephrology, Children’s Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Liu
- Department of Nephrology, Children’s Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
6
|
Meliambro K, He JC, Campbell KN. Podocyte-targeted therapies - progress and future directions. Nat Rev Nephrol 2024; 20:643-658. [PMID: 38724717 DOI: 10.1038/s41581-024-00843-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2024] [Indexed: 09/14/2024]
Abstract
Podocytes are the key target cells for injury across the spectrum of primary and secondary proteinuric kidney disorders, which account for up to 90% of cases of kidney failure worldwide. Seminal experimental and clinical studies have established a causative link between podocyte depletion and the magnitude of proteinuria in progressive glomerular disease. However, no substantial advances have been made in glomerular disease therapies, and the standard of care for podocytopathies relies on repurposed immunosuppressive drugs. The past two decades have seen a remarkable expansion in understanding of the mechanistic basis of podocyte injury, with prospects increasing for precision-based treatment approaches. Dozens of disease-causing genes with roles in the pathogenesis of clinical podocytopathies have been identified, as well as a number of putative glomerular permeability factors. These achievements, together with the identification of novel targets of podocyte injury, the development of potential approaches to harness the endogenous podocyte regenerative potential of progenitor cell populations, ongoing clinical trials of podocyte-specific pharmacological agents and the development of podocyte-directed drug delivery systems, contribute to an optimistic outlook for the future of glomerular disease therapy.
Collapse
Affiliation(s)
- Kristin Meliambro
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John C He
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kirk N Campbell
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
7
|
Hou S, Yang B, Chen Q, Xu Y, Li H. Potential biomarkers of recurrent FSGS: a review. BMC Nephrol 2024; 25:258. [PMID: 39134955 PMCID: PMC11318291 DOI: 10.1186/s12882-024-03695-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024] Open
Abstract
Focal segmental glomerulosclerosis (FSGS), a clinicopathological condition characterized by nephrotic-range proteinuria, has a high risk of progression to end-stage renal disease (ESRD). Meanwhile, the recurrence of FSGS after renal transplantation is one of the main causes of graft loss. The diagnosis of recurrent FSGS is mainly based on renal puncture biopsy transplants, an approach not widely consented by patients with early mild disease. Therefore, there is an urgent need to find definitive diagnostic markers that can act as a target for early diagnosis and intervention in the treatment of patients. In this review, we summarize the domestic and international studies on the pathophysiology, pathogenesis and earliest screening methods of FSGS and describe the functions and roles of specific circulating factors in the progression of early FSGS, in order to provide a new theoretical basis for early diagnosis of FSGS recurrence, as well as aid the exploration of therapeutic targets.
Collapse
Affiliation(s)
- Shuang Hou
- Department of Organ Transplantation, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550000, China
| | - Bo Yang
- Department of Organ Transplantation, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550000, China
| | - Qian Chen
- Department of Organ Transplantation, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550000, China
| | - Yuan Xu
- Department of Organ Transplantation, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550000, China.
| | - Haiyang Li
- Hepatological surgery department, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550000, China.
| |
Collapse
|
8
|
Bonilla M, Efe O, Selvaskandan H, Lerma EV, Wiegley N. A Review of Focal Segmental Glomerulosclerosis Classification With a Focus on Genetic Associations. Kidney Med 2024; 6:100826. [PMID: 38765809 PMCID: PMC11099322 DOI: 10.1016/j.xkme.2024.100826] [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] [Indexed: 05/22/2024] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) defines a distinct histologic pattern observed in kidney tissue that is linked to several distinct underlying causes, all converging on the common factor of podocyte injury. It presents a considerable challenge in terms of classification because of its varied underlying causes and the limited correlation between histopathology and clinical outcomes. Critically, precise nomenclature is key to describe and delineate the pathogenesis, subsequently guiding the selection of suitable and precision therapies. A proposed pathomechanism-based approach has been suggested for FSGS classification. This approach differentiates among primary, secondary, genetic, and undetermined causes, aiming to provide clarity. Genetic FSGS from monogenic mutations can emerge during childhood or adulthood, and it is advisable to conduct genetic testing in cases in which there is a family history of chronic kidney disease, nephrotic syndrome, or resistance to treatment. Genome-wide association studies have identified several genetic risk variants, such as those in apolipoprotein L1 (APOL1), that play a role in the development of FSGS. Currently, no specific treatments have been approved to treat genetic FSGS; however, interventions targeting underlying cofactor deficiencies have shown potential in some cases. Furthermore, encouraging results have emerged from a phase 2 trial investigating inaxaplin, a novel small molecule APOL1 channel inhibitor, in APOL1-associated FSGS.
Collapse
Affiliation(s)
- Marco Bonilla
- Section of Nephrology, Department of Medicine, University of Chicago, Chicago, IL
| | - Orhan Efe
- Division of Nephrology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Haresh Selvaskandan
- IgA Mayer Nephropathy Laboratories, Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- John Walls Renal Unit, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Edgar V. Lerma
- Section of Nephrology, University of Illinois at Chicago/Advocate Christ Medical Center, Oak Lawn, IL
| | - Nasim Wiegley
- University of California Davis School of Medicine, Division of Nephrology, Sacramento, CA
| |
Collapse
|
9
|
Catanese L, Rupprecht H, Huber TB, Lindenmeyer MT, Hengel FE, Amann K, Wendt R, Siwy J, Mischak H, Beige J. Non-Invasive Biomarkers for Diagnosis, Risk Prediction, and Therapy Guidance of Glomerular Kidney Diseases: A Comprehensive Review. Int J Mol Sci 2024; 25:3519. [PMID: 38542491 PMCID: PMC10970781 DOI: 10.3390/ijms25063519] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 04/09/2025] Open
Abstract
Effective management of glomerular kidney disease, one of the main categories of chronic kidney disease (CKD), requires accurate diagnosis, prognosis of progression, assessment of therapeutic efficacy, and, ideally, prediction of drug response. Multiple biomarkers and algorithms for the assessment of specific aspects of glomerular diseases have been reported in the literature. Though, the vast majority of these have not been implemented in clinical practice or are not available on a global scale due to limited access, missing medical infrastructure, or economical as well as political reasons. The aim of this review is to compile all currently available information on the diagnostic, prognostic, and predictive biomarkers currently available for the management of glomerular diseases, and provide guidance on the application of these biomarkers. As a result of the compiled evidence for the different biomarkers available, we present a decision tree for a non-invasive, biomarker-guided diagnostic path. The data currently available demonstrate that for the large majority of patients with glomerular diseases, valid biomarkers are available. However, despite the obvious disadvantages of kidney biopsy, being invasive and not applicable for monitoring, especially in the context of rare CKD etiologies, kidney biopsy still cannot be replaced by non-invasive strategies.
Collapse
Affiliation(s)
- Lorenzo Catanese
- Department of Nephrology, Angiology and Rheumatology, Klinikum Bayreuth GmbH, 95445 Bayreuth, Germany; (L.C.); (H.R.)
- Kuratorium for Dialysis and Transplantation (KfH) Bayreuth, 95445 Bayreuth, Germany
- Department of Nephrology, Medizincampus Oberfranken, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Harald Rupprecht
- Department of Nephrology, Angiology and Rheumatology, Klinikum Bayreuth GmbH, 95445 Bayreuth, Germany; (L.C.); (H.R.)
- Kuratorium for Dialysis and Transplantation (KfH) Bayreuth, 95445 Bayreuth, Germany
- Department of Nephrology, Medizincampus Oberfranken, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Tobias B. Huber
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (T.B.H.); (M.T.L.); (F.E.H.)
| | - Maja T. Lindenmeyer
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (T.B.H.); (M.T.L.); (F.E.H.)
| | - Felicitas E. Hengel
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (T.B.H.); (M.T.L.); (F.E.H.)
| | - Kerstin Amann
- Department of Nephropathology, Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Ralph Wendt
- Division of Nephrology, St. Georg Hospital, 04129 Leipzig, Germany;
| | - Justyna Siwy
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (J.S.); (H.M.)
| | - Harald Mischak
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (J.S.); (H.M.)
| | - Joachim Beige
- Division of Nephrology, St. Georg Hospital, 04129 Leipzig, Germany;
- Department of Internal Medicine II, Martin-Luther-University Halle-Wittenberg, 06108 Halle, Germany
- Kuratorium for Dialysis and Transplantation (KfH) Leipzig, 04129 Leipzig, Germany
| |
Collapse
|
10
|
Catanese L, Siwy J, Wendt R, Amann K, Beige J, Hendry B, Mischak H, Mullen W, Paterson I, Schiffer M, Wolf M, Rupprecht H. Differentiating primary and secondary FSGS using non-invasive urine biomarkers. Clin Kidney J 2024; 17:sfad296. [PMID: 38313685 PMCID: PMC10833144 DOI: 10.1093/ckj/sfad296] [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: 05/17/2023] [Indexed: 02/06/2024] Open
Abstract
Background Focal segmental glomerulosclerosis (FSGS) is divided into genetic, primary (p), uncertain cause, and secondary (s) forms. The subclasses differ in management and prognosis with differentiation often being challenging. We aimed to identify specific urine proteins/peptides discriminating between clinical and biopsy-proven pFSGS and sFSGS. Methods Sixty-three urine samples were collected in two different centers (19 pFSGS and 44 sFSGS) prior to biopsy. Samples were analysed using capillary electrophoresis-coupled mass spectrometry. For biomarker definition, datasets of age-/sex-matched normal controls (NC, n = 98) and patients with other chronic kidney diseases (CKDs, n = 100) were extracted from the urinary proteome database. Independent specificity assessment was performed in additional data of NC (n = 110) and CKD (n = 170). Results Proteomics data from patients with pFSGS were first compared to NC (n = 98). This resulted in 1179 biomarker (P < 0.05) candidates. Then, the pFSGS group was compared to sFSGS, and in a third step, pFSGS data were compared to data from different CKD etiologies (n = 100). Finally, 93 biomarkers were identified and combined in a classifier, pFSGS93. Total cross-validation of this classifier resulted in an area under the receiving operating curve of 0.95. The specificity investigated in an independent set of NC and CKD of other etiologies was 99.1% for NC and 94.7% for CKD, respectively. The defined biomarkers are largely fragments of different collagens (49%). Conclusion A urine peptide-based classifier that selectively detects pFSGS could be developed. Specificity of 95%-99% could be assessed in independent samples. Sensitivity must be confirmed in independent cohorts before routine clinical application.
Collapse
Affiliation(s)
- Lorenzo Catanese
- Department of Nephrology, Angiology and Rheumatology, Klinikum Bayreuth GmbH, Bayreuth, Germany
- Kuratorium for Dialysis and Transplantation (KfH) Bayreuth, Bayreuth, Germany
- Medizincampus Oberfranken, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | | | - Ralph Wendt
- Division of Nephrology, St. Georg Hospital Leipzig, Leipzig, Germany
| | - Kerstin Amann
- Department of Nephropathology, Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Joachim Beige
- Kuratorium for Dialysis and Transplantation (KfH) Renal Unit, Leipzig, Germany
- Department of Internal Medicine II, Martin-Luther-University Halle/Wittenberg, Halle/Saale, Germany
| | | | | | - William Mullen
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | | | - Mario Schiffer
- Department of Nephrology and Hypertension, University Hospital Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Research Center on Rare Kidney Diseases (RECORD), University Hospital Erlangen, Erlangen, Germany
| | | | - Harald Rupprecht
- Department of Nephrology, Angiology and Rheumatology, Klinikum Bayreuth GmbH, Bayreuth, Germany
- Kuratorium for Dialysis and Transplantation (KfH) Bayreuth, Bayreuth, Germany
- Medizincampus Oberfranken, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
11
|
Chhuon C, Herrera-Marcos LV, Zhang SY, Charrière-Bertrand C, Jung V, Lipecka J, Savas B, Nasser N, Pawlak A, Boulmerka H, Audard V, Sahali D, Guerrera IC, Ollero M. Proteomics of Plasma and Plasma-Treated Podocytes: Application to Focal and Segmental Glomerulosclerosis. Int J Mol Sci 2023; 24:12124. [PMID: 37569500 PMCID: PMC10418338 DOI: 10.3390/ijms241512124] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Focal and segmental glomerulosclerosis (FSGS) is a severe form of idiopathic nephrotic syndrome (INS), a glomerulopathy of presumably immune origin that is attributed to extrarenal pathogenic circulating factors. The recurrence of FSGS (rFSGS) after transplant occurs in 30% to 50% of cases. The direct analysis of patient plasma proteome has scarcely been addressed to date, mainly due to the methodological difficulties associated with plasma complexity and dynamic range. In this study, first, we compared different methods of plasma preparation, second, we compared the plasma proteomes of rFSGS and controls using two preparation methods, and third, we analyzed the early proximal signaling events in podocytes subjected to patient plasma, through a combination of phosphoproteomics and lipid-raft proteomics (raftomics). By combining immunodepletion and high pH fractionation, we performed a differential proteomic analysis of soluble plasma proteins and of extracellular vesicles (EV) obtained from healthy controls, non-INS patient controls, and rFSGS patients (n = 4). In both the soluble- and the EV-protein sets from the rFSGS patients, we found a statistically significant increase in a cluster of proteins involved in neutrophil degranulation. A group of lipid-binding proteins, generally associated with lipoproteins, was found to be decreased in the soluble set from the rFSGS patients. In addition, three amino acid transporters involved in mTORC1 activation were found to be significantly increased in the EV from the rFSGS. Next, we incubated human podocytes for 30 min with 10% plasma from both groups of patients. The phosphoproteomics and raftomics of the podocytes revealed profound differences in the proteins involved in the mTOR pathway, in autophagy, and in cytoskeleton organization. We analyzed the correlation between the abundance of plasma and plasma-regulated podocyte proteins. The observed changes highlight some of the mechanisms involved in FSGS recurrence and could be used as specific early markers of circulating-factor activity in podocytes.
Collapse
Affiliation(s)
- Cerina Chhuon
- Proteomic Platform Necker, Université Paris Cité Structure Fédérative de Recherche SFR Necker US24, 75015 Paris, France; (C.C.); (V.J.); (J.L.)
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
| | - Luis Vicente Herrera-Marcos
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
| | - Shao-Yu Zhang
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
| | - Cécile Charrière-Bertrand
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
| | - Vincent Jung
- Proteomic Platform Necker, Université Paris Cité Structure Fédérative de Recherche SFR Necker US24, 75015 Paris, France; (C.C.); (V.J.); (J.L.)
| | - Joanna Lipecka
- Proteomic Platform Necker, Université Paris Cité Structure Fédérative de Recherche SFR Necker US24, 75015 Paris, France; (C.C.); (V.J.); (J.L.)
| | - Berkan Savas
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
| | - Nour Nasser
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
| | - André Pawlak
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
| | - Hocine Boulmerka
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
| | - Vincent Audard
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
- AP-HP, Hôpitaux Universitaires Henri Mondor, Service de Néphrologie, F-94010 Creteil, France
| | - Dil Sahali
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
- AP-HP, Hôpitaux Universitaires Henri Mondor, Service de Néphrologie, F-94010 Creteil, France
| | - Ida Chiara Guerrera
- Proteomic Platform Necker, Université Paris Cité Structure Fédérative de Recherche SFR Necker US24, 75015 Paris, France; (C.C.); (V.J.); (J.L.)
| | - Mario Ollero
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
| |
Collapse
|
12
|
Hernández D, Caballero A. Kidney transplant in the next decade: Strategies, challenges and vision of the future. Nefrologia 2023; 43:281-292. [PMID: 37635014 DOI: 10.1016/j.nefroe.2022.04.008] [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: 11/10/2021] [Accepted: 04/24/2022] [Indexed: 08/29/2023] Open
Abstract
Although the results of kidney transplantation (KT) have improved substantially in recent years, a chronic and inexorable loss of grafts mainly due to the death of the patient and chronic dysfunction of the KT, continues to be observed. The objectives, thus, to optimize this situation in the next decade are fundamentally focused on minimizing the rate of kidney graft loss, improving patient survival, increasing the rate of organ procurement and its distribution, promoting research and training in health professionals and the development of scientific registries providing clinical and reliable information that allow us to optimize our clinical practice in the field of KT. With this perspective, this review will deep into: (1) strategies to avoid chronic dysfunction and graft loss in the medium and long term; (2) to prolong patient survival; (3) strategies to increase the donation, maintenance and allocation of organs; (4) promote clinical and basic research and training activity in KT; and (5) the analysis of the results in KT by optimizing and merging scientific registries.
Collapse
Affiliation(s)
- Domingo Hernández
- Unidad de Gestión Clínica de Nefrología, Hospital Regional Universitario Carlos Haya, Instituto Biomédico de Investigación de Málaga (IBIMA), Universidad de Málaga, REDinREN, Málaga, Spain.
| | - Abelardo Caballero
- Sección de Inmunología, Hospital Regional Universitario Carlos Haya, Instituto Biomédico de Investigación de Málaga (IBIMA), Universidad de Málaga, REDinREN, Málaga, Spain
| |
Collapse
|
13
|
Shen Q, Teng L, Wang Y, Guo L, Xu F, Huang H, Xie W, Zhou Q, Chen Y, Wang J, Mao Y, Chen J, Jiang H. Integrated genomic, transcriptomic and metabolomic analysis reveals MDH2 mutation-induced metabolic disorder in recurrent focal segmental glomerulosclerosis. Front Immunol 2022; 13:962986. [PMID: 36159820 PMCID: PMC9495259 DOI: 10.3389/fimmu.2022.962986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) has an over 30% risk of recurrence after kidney transplantation (Ktx) and is associated with an extremely high risk of graft loss. However, mechanisms remain largely unclear. Thus, this study identifies novel genes related to the recurrence of FSGS (rFSGS). Whole genome-wide sequencing and next-generation RNA sequencing were used to identify the candidate mutant genes associated with rFSGS in peripheral blood mononuclear cells (PBMCs) from patients with biopsy-confirmed rFSGS after KTx. To confirm the functional role of the identified gene with the MDH2 c.26C >T mutation, a homozygous MDH2 c.26C >T mutation in HMy2.CIR cell line was induced by CRISPR/Cas9 and co-cultured with podocytes, mesangial cells, or HK2 cells, respectively, to detect the potential pathogenicity of the c.26C >T variant in MDH2. A total of 32 nonsynonymous single nucleotide polymorphisms (SNPs) and 610 differentially expressed genes (DEGs) related to rFSGS were identified. DEGs are mainly enriched in the immune and metabolomic-related pathways. A variant in MDH2, c.26C >T, was found in all patients with rFSGS, which was also accompanied by lower levels of mRNA expression in PBMCs from relapsed patients compared with patients with remission after KTx. Functionally, co-cultures of HMy2.CIR cells overexpressing the mutant MDH2 significantly inhibited the expression of synaptopodin, podocin, and F-actin by podocytes compared with those co-cultured with WT HMy2.CIR cells or podocytes alone. We identified that MDH2 is a novel rFSGS susceptibility gene in patients with recurrence of FSGS after KTx. Mutation of the MDH2 c.26C >T variant may contribute to progressive podocyte injury in rFSGS patients.
Collapse
Affiliation(s)
- Qixia Shen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Lisha Teng
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Yucheng Wang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Luying Guo
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Feng Xu
- The Centre for Heart and Lung Innovation, The University of British Columbia, Vancouver, BC, Canada
| | - Hongfeng Huang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Wenqing Xie
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Qin Zhou
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Ying Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Junwen Wang
- Department of Health Sciences Research and Center for Individualized Medicine, Mayo Clinic, Scottsdale, AZ, United States
| | - Youying Mao
- Dapartment of Nephrology, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Hong Jiang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
- *Correspondence: Hong Jiang,
| |
Collapse
|
14
|
Trasplante renal en la próxima década: estrategias, retos y visión de futuro. Nefrologia 2022. [DOI: 10.1016/j.nefro.2022.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
15
|
HLA Loci and Recurrence of Focal Segmental Glomerulosclerosis in Pediatric Kidney Transplantation. Transplant Direct 2021; 7:e748. [PMID: 34476293 PMCID: PMC8405131 DOI: 10.1097/txd.0000000000001201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/16/2021] [Indexed: 11/21/2022] Open
Abstract
Recurrent focal segmental glomerulosclerosis (FSGS) after kidney transplantation accounts for the majority of allograft failures in children with primary FSGS. Although current research focuses on FSGS pathophysiology, a common etiology and mechanisms of disease recurrence remain elusive.
Collapse
|
16
|
De Souza L, Prunster J, Chan D, Chakera A, Lim WH. Recurrent glomerulonephritis after kidney transplantation: a practical approach. Curr Opin Organ Transplant 2021; 26:360-380. [PMID: 34039882 DOI: 10.1097/mot.0000000000000887] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW This review will provide a practical approach in the assessment of kidney failure patients with primary glomerulonephritides (GN) being considered for kidney transplantation, focusing on high-risk subtypes of immunoglobulin A nephropathy, focal segmental glomerulosclerosis, idiopathic membranous glomerulonephritis and membranoproliferative glomerulonephritis. RECENT FINDINGS Recurrent glomerulonephritis remains one of the most common causes of allograft loss in kidney transplant recipients. Although the epidemiology and clinical outcomes of glomerulonephritis recurrence occurring after kidney transplantation are relatively well-described, the natural course and optimal treatment strategies of recurrent disease in kidney allografts remain poorly defined. With a greater understanding of the pathophysiology and treatment responses of patients with glomerulonephritis affecting the native kidneys, these discoveries have laid the framework for the potential to improve the management of patients with high-risk glomerulonephritis subtypes being considered for kidney transplantation. SUMMARY Advances in the understanding of the underlying immunopathogenesis of primary GN has the potential to offer novel therapeutic options for kidney patients who develop recurrent disease after kidney transplantation. To test the efficacy of novel treatment options in adequately powered clinical trials requires a more detailed understanding of the clinical and histological characteristics of kidney transplant recipients with recurrent glomerulonephritis.
Collapse
Affiliation(s)
- Laura De Souza
- Department of Renal Medicine, Cairns Hospital, Cairns North, Queensland
| | - Janelle Prunster
- Department of Renal Medicine, Cairns Hospital, Cairns North, Queensland
| | - Doris Chan
- Department of Renal Medicine, Sir Charles Gairdner Hospital, Perth
| | - Aron Chakera
- Department of Renal Medicine, Sir Charles Gairdner Hospital, Perth
| | - Wai H Lim
- Department of Renal Medicine, Sir Charles Gairdner Hospital, Perth
- Medical School, University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
17
|
Kon V, Yang HC, Smith LE, Vickers KC, Linton MF. High-Density Lipoproteins in Kidney Disease. Int J Mol Sci 2021; 22:ijms22158201. [PMID: 34360965 PMCID: PMC8348850 DOI: 10.3390/ijms22158201] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
Decades of epidemiological studies have established the strong inverse relationship between high-density lipoprotein (HDL)-cholesterol concentration and cardiovascular disease. Recent evidence suggests that HDL particle functions, including anti-inflammatory and antioxidant functions, and cholesterol efflux capacity may be more strongly associated with cardiovascular disease protection than HDL cholesterol concentration. These HDL functions are also relevant in non-cardiovascular diseases, including acute and chronic kidney disease. This review examines our current understanding of the kidneys’ role in HDL metabolism and homeostasis, and the effect of kidney disease on HDL composition and functionality. Additionally, the roles of HDL particles, proteins, and small RNA cargo on kidney cell function and on the development and progression of both acute and chronic kidney disease are examined. The effect of HDL protein modification by reactive dicarbonyls, including malondialdehyde and isolevuglandin, which form adducts with apolipoprotein A-I and impair proper HDL function in kidney disease, is also explored. Finally, the potential to develop targeted therapies that increase HDL concentration or functionality to improve acute or chronic kidney disease outcomes is discussed.
Collapse
Affiliation(s)
- Valentina Kon
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (V.K.); (H.-C.Y.)
| | - Hai-Chun Yang
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (V.K.); (H.-C.Y.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Loren E. Smith
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Kasey C. Vickers
- Atherosclerosis Research Unit, Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - MacRae F. Linton
- Atherosclerosis Research Unit, Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
- Correspondence:
| |
Collapse
|
18
|
A Specific Tubular ApoA-I Distribution Is Associated to FSGS Recurrence after Kidney Transplantation. J Clin Med 2021; 10:jcm10102174. [PMID: 34069888 PMCID: PMC8157584 DOI: 10.3390/jcm10102174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 05/14/2021] [Indexed: 01/27/2023] Open
Abstract
A major complication of primary focal segmental glomerulosclerosis (FSGS) is its recurrence after kidney transplantation that happens in 30 to 40% of the patients. The diagnosis of these relapses is not always easy as the histological lesions are not highly specific and appear after the proteinuria increase. Currently, there are no accurate biomarkers to detect FSGS recurrence. Our group identified a modified form of Apolipoprotein A-I (ApoA-I), named ApoA-Ib, specifically present in the urine of recurrent FSGS patients after kidney transplantation. Aberrant forms of ApoA-I have also been described in the urine of native primary FSGS patients; this feature has been associated with prominent staining of ApoA-I at the apical membrane of the tubular cells. In this study, we aim to analyze the ApoA-I distribution in kidney allograft biopsies of recurrent FSGS patients. We detected ApoA-I by immunohistochemistry in kidney allograft biopsies of patients with FSGS relapse after kidney transplantation and in kidney allograft biopsies of patients with a disease different from FSGS in the native kidney (non-FSGS). In recurrent FSGS patients, ApoA-I was prominently localized at the brush border of the tubular cells, while in the non-FSGS patients, ApoA-I was found along the cytoplasm of the tubular cells. The localization of ApoA-I at the brush border of the tubular cells is a specific feature of primary FSGS in relapse. This suggests that ApoA-I staining in kidney biopsies, coupled with ApoA-Ib measurement in urine, could be used as a diagnostic tool of primary FSGS relapse after kidney transplantation due to its highly specific tubular distribution.
Collapse
|
19
|
Shoji J, Mii A, Terasaki M, Shimizu A. Update on Recurrent Focal Segmental Glomerulosclerosis in Kidney Transplantation. Nephron Clin Pract 2020; 144 Suppl 1:65-70. [PMID: 33260184 DOI: 10.1159/000510748] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 08/06/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Focal segmental glomerulosclerosis (FSGS) is a clinicopathological syndrome characterized by nephrotic-range proteinuria with high incidence of progression to end-stage renal disease (ESRD). In primary FSGS, 40-60% of patients develop ESRD within 10-20 years. SUMMARY Recurrence of FSGS after kidney transplantation is frequent and is associated with poor allograft survival. The risk factors for recurrent FSGS include onset of FSGS during childhood, rapid progression of primary FSGS to ESRD, history of recurrent FSGS in previous allograft, and diffuse mesangial hypercellularity or collapsing variant of FSGS in the native kidney. The early histological findings of recurrent FSGS consist of unremarkable glomerular changes on light microscopy but significant podocyte effacement on electron microscopy; the loss of foot processes with eventual dropout of podocytes leads to the development of segmental lesions in the glomerulus. Experimental and clinical data suggest the existence of circulating permeability factors, such as soluble urokinase-type plasminogen activator receptor (suPAR), cardiotrophin-like cytokine factor-1 (CLCF-1), CD40 axis, and apolipoprotein A-Ib (ApoA-Ib), in the pathogenesis of recurrent FSGS. These biomarkers including circulating permeability factors may facilitate earlier diagnosis of FSGS posttransplant and may guide in the development of novel therapies that may be more effective and improve long-term outcomes in kidney transplantation. Key Messages: Several studies have suggested the possible circulating permeability factors, such as suPAR, CLCF-1, CD40 axis, and ApoA-Ib, in the pathogenesis and disease progression of FSGS and recurrent FSGS. Further studies should be performed to elucidate the true essential biomarker(s) associated with the onset and progression of FSGS as well as recurrent FSGS.
Collapse
Affiliation(s)
- Jun Shoji
- Division of Transplant Nephrology, University of California San Francisco, San Francisco, California, USA
| | - Akiko Mii
- Department of Nephrology, Nippon Medical School, Tokyo, Japan
| | - Mika Terasaki
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Akira Shimizu
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan,
| |
Collapse
|
20
|
Jacobs-Cachá C, Vergara A, García-Carro C, Agraz I, Toapanta-Gaibor N, Ariceta G, Moreso F, Serón D, López-Hellín J, Soler MJ. Challenges in primary focal segmental glomerulosclerosis diagnosis: from the diagnostic algorithm to novel biomarkers. Clin Kidney J 2020; 14:482-491. [PMID: 33623672 PMCID: PMC7886539 DOI: 10.1093/ckj/sfaa110] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 05/04/2020] [Indexed: 12/11/2022] Open
Abstract
Primary or idiopathic focal segmental glomerulosclerosis (FSGS) is a kidney entity that involves the podocytes, leading to heavy proteinuria and in many cases progresses to end-stage renal disease. Idiopathic FSGS has a bad prognosis, as it involves young individuals who, in a considerably high proportion (∼15%), are resistant to corticosteroids and other immunosuppressive treatments as well. Moreover, the disease recurs in 30–50% of patients after kidney transplantation, leading to graft function impairment. It is suspected that this relapsing disease is caused by a circulating factor(s) that would permeabilize the glomerular filtration barrier. However, the exact pathologic mechanism is an unsettled issue. Besides its poor outcome, a major concern of primary FSGS is the complexity to confirm the diagnosis, as it can be confused with other variants or secondary forms of FSGS and also with other glomerular diseases, such as minimal change disease. New efforts to optimize the diagnostic approach are arising to improve knowledge in well-defined primary FSGS cohorts of patients. Follow-up of properly classified primary FSGS patients will allow risk stratification for predicting the response to different treatments. In this review we will focus on the diagnostic algorithm used in idiopathic FSGS both in native kidneys and in disease recurrence after kidney transplantation. We will emphasize those potential confusing factors as well as their detection and prevention. In addition, we will also provide an overview of ongoing studies that recruit large cohorts of glomerulopathy patients (Nephrotic Syndrome Study Network and Cure Glomerulonephropathy, among others) and the experimental studies performed to find novel reliable biomarkers to detect primary FSGS.
Collapse
Affiliation(s)
- Conxita Jacobs-Cachá
- Nephrology Research Group, Vall d'hebrón Institut de Recerca (VHIR), Barcelona, Spain.,Department of Nephrology, Hospital Universitari Vall d'Hebron, Universitat Autónoma de Barcelona, Barcelona, Spain.,Red de Investigaciones Renales (RedInRen), Madrid, Spain
| | - Ander Vergara
- Nephrology Research Group, Vall d'hebrón Institut de Recerca (VHIR), Barcelona, Spain.,Department of Nephrology, Hospital Universitari Vall d'Hebron, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Clara García-Carro
- Nephrology Research Group, Vall d'hebrón Institut de Recerca (VHIR), Barcelona, Spain.,Department of Nephrology, Hospital Universitari Vall d'Hebron, Universitat Autónoma de Barcelona, Barcelona, Spain.,Red de Investigaciones Renales (RedInRen), Madrid, Spain
| | - Irene Agraz
- Nephrology Research Group, Vall d'hebrón Institut de Recerca (VHIR), Barcelona, Spain.,Department of Nephrology, Hospital Universitari Vall d'Hebron, Universitat Autónoma de Barcelona, Barcelona, Spain.,Red de Investigaciones Renales (RedInRen), Madrid, Spain
| | - Nestor Toapanta-Gaibor
- Nephrology Research Group, Vall d'hebrón Institut de Recerca (VHIR), Barcelona, Spain.,Department of Nephrology, Hospital Universitari Vall d'Hebron, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Gema Ariceta
- Red de Investigaciones Renales (RedInRen), Madrid, Spain.,Department of Paediatric Nephrology, Hospital Universitari Vall d'Hebron. Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Francesc Moreso
- Nephrology Research Group, Vall d'hebrón Institut de Recerca (VHIR), Barcelona, Spain.,Department of Nephrology, Hospital Universitari Vall d'Hebron, Universitat Autónoma de Barcelona, Barcelona, Spain.,Red de Investigaciones Renales (RedInRen), Madrid, Spain
| | - Daniel Serón
- Nephrology Research Group, Vall d'hebrón Institut de Recerca (VHIR), Barcelona, Spain.,Department of Nephrology, Hospital Universitari Vall d'Hebron, Universitat Autónoma de Barcelona, Barcelona, Spain.,Red de Investigaciones Renales (RedInRen), Madrid, Spain
| | - Joan López-Hellín
- Red de Investigaciones Renales (RedInRen), Madrid, Spain.,Department of Biochemistry, Hospital Universitari Vall d'Hebron, Universitat Autónoma de Barcelona, Barcelona, Spain.,Biochemistry Research Group, Vall d'hebrón Institut de Recerca (VHIR), Barcelona, Spain
| | - Maria José Soler
- Nephrology Research Group, Vall d'hebrón Institut de Recerca (VHIR), Barcelona, Spain.,Department of Nephrology, Hospital Universitari Vall d'Hebron, Universitat Autónoma de Barcelona, Barcelona, Spain.,Red de Investigaciones Renales (RedInRen), Madrid, Spain
| |
Collapse
|
21
|
Abuzeineh M, Aala A, Alasfar S, Alachkar N. Angiotensin II receptor 1 antibodies associate with post-transplant focal segmental glomerulosclerosis and proteinuria. BMC Nephrol 2020; 21:253. [PMID: 32615995 PMCID: PMC7331243 DOI: 10.1186/s12882-020-01910-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/25/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Angiotensin II type 1 receptors (AT1Rs) are expressed on podocytes, endothelial and other cells, and play an essential role in the maintenance of podocyte function and vascular homeostasis. The presence of AT1R antibodies (AT1R-Abs) leads to activation of these receptors resulting in podocyte injury and endothelial cell dysfunction. We assessed the correlation between AT1R-Abs and the risk of post-transplant FSGS. METHODS This is a retrospective study, which included all kidney transplant recipients with positive AT1R-Abs (≥ 9 units/ml), who were transplanted and followed at our center between 2006 and 2016. We assessed the development of biopsy proven FSGS and proteinuria by urine protein to creatinine ratio of ≥1 g/g and reviewed short and long term outcomes. RESULTS We identified 100 patients with positive AT1R-Abs at the time of kidney transplant biopsy or proteinuria. 49% recipients (FSGS group) had biopsy-proven FSGS and/or proteinuria and 51% did not (non-FSGS group). Pre-transplant hypertension was present in 89% of the FSGS group compared to 72% in the non-FSGS group, p = 0.027. Of the FSGS group, 43% were on angiotensin converting enzyme inhibitors or angiotensin receptor blockers prior to transplantation, compared to 25.5% in the non-FSGS group, p = 0.06. Primary idiopathic FSGS was the cause of ESRD in 20% of the FSGS group, compared to 6% in the non-FSGS group, p = 0.03. The allograft loss was significantly higher in the FSGS group 63% compared to 39% in non-FSGS. Odds ratio and 95% confidence interval were 2.66 (1.18-5.99), p = 0.017. CONCLUSIONS Our data suggest a potential association between AT1R-Abs and post-transplant FSGS leading to worse allograft outcome. Therefore, AT1R-Abs may be considered biomarkers for post-transplant FSGS.
Collapse
Affiliation(s)
- Mohammad Abuzeineh
- Department of Medicine, Division of Nephrology, The Johns Hopkins University School of Medicine, 600 N Wolfe St, Carnegie 344B, Baltimore, MD, 21287, USA
| | - Amtul Aala
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Sami Alasfar
- Department of Medicine, Division of Nephrology, The Johns Hopkins University School of Medicine, 600 N Wolfe St, Carnegie 344B, Baltimore, MD, 21287, USA
| | - Nada Alachkar
- Department of Medicine, Division of Nephrology, The Johns Hopkins University School of Medicine, 600 N Wolfe St, Carnegie 344B, Baltimore, MD, 21287, USA.
| |
Collapse
|
22
|
Bukosza EN, Kornauth C, Hummel K, Schachner H, Huttary N, Krieger S, Nöbauer K, Oszwald A, Razzazi Fazeli E, Kratochwill K, Aufricht C, Szénási G, Hamar P, Gebeshuber CA. ECM Characterization Reveals a Massive Activation of Acute Phase Response during FSGS. Int J Mol Sci 2020; 21:ijms21062095. [PMID: 32197499 PMCID: PMC7139641 DOI: 10.3390/ijms21062095] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/16/2020] [Accepted: 03/16/2020] [Indexed: 12/19/2022] Open
Abstract
The glomerular basement membrane (GBM) and extra-cellular matrix (ECM) are essential to maintain a functional interaction between the glomerular podocytes and the fenestrated endothelial cells in the formation of the slit diaphragm for the filtration of blood. Dysregulation of ECM homeostasis can cause Focal segmental glomerulosclerosis (FSGS). Despite this central role, alterations in ECM composition during FSGS have not been analyzed in detail yet. Here, we characterized the ECM proteome changes in miR-193a-overexpressing mice, which suffer from FSGS due to suppression of Wilms' tumor 1 (WT1). By mass spectrometry we identified a massive activation of the acute phase response, especially the complement and fibrinogen pathways. Several protease inhibitors (ITIH1, SERPINA1, SERPINA3) were also strongly increased. Complementary analysis of RNA expression data from both miR-193a mice and human FSGS patients identified additional candidate genes also mainly involved in the acute phase response. In total, we identified more than 60 dysregulated, ECM-associated genes with potential relevance for FSGS progression. Our comprehensive analysis of a murine FSGS model and translational comparison with human data offers novel targets for FSGS therapy.
Collapse
Affiliation(s)
- Eva Nora Bukosza
- Institute of Translational Medicine, Semmelweis University Budapest, Tűzoltó u 37-47, 1094 Budapest, Hungary; (E.N.B.); (G.S.); (P.H.)
- Clinical Institute for Pathology, Medical University Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria; (C.K.); (H.S.); (N.H.); (S.K.); (A.O.)
| | - Christoph Kornauth
- Clinical Institute for Pathology, Medical University Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria; (C.K.); (H.S.); (N.H.); (S.K.); (A.O.)
- Clinical Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Karin Hummel
- Vetcore Facility for Research, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria; (K.H.); (K.N.); (E.R.F.)
| | - Helga Schachner
- Clinical Institute for Pathology, Medical University Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria; (C.K.); (H.S.); (N.H.); (S.K.); (A.O.)
| | - Nicole Huttary
- Clinical Institute for Pathology, Medical University Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria; (C.K.); (H.S.); (N.H.); (S.K.); (A.O.)
| | - Sigurd Krieger
- Clinical Institute for Pathology, Medical University Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria; (C.K.); (H.S.); (N.H.); (S.K.); (A.O.)
| | - Katharina Nöbauer
- Vetcore Facility for Research, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria; (K.H.); (K.N.); (E.R.F.)
| | - André Oszwald
- Clinical Institute for Pathology, Medical University Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria; (C.K.); (H.S.); (N.H.); (S.K.); (A.O.)
| | - Ebrahim Razzazi Fazeli
- Vetcore Facility for Research, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria; (K.H.); (K.N.); (E.R.F.)
| | - Klaus Kratochwill
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1210 Vienna, Austria;
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1210 Vienna, Austria;
| | - Christoph Aufricht
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1210 Vienna, Austria;
| | - Gabor Szénási
- Institute of Translational Medicine, Semmelweis University Budapest, Tűzoltó u 37-47, 1094 Budapest, Hungary; (E.N.B.); (G.S.); (P.H.)
| | - Peter Hamar
- Institute of Translational Medicine, Semmelweis University Budapest, Tűzoltó u 37-47, 1094 Budapest, Hungary; (E.N.B.); (G.S.); (P.H.)
| | - Christoph A. Gebeshuber
- Clinical Institute for Pathology, Medical University Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria; (C.K.); (H.S.); (N.H.); (S.K.); (A.O.)
- Correspondence: ; Tel.: +43-1-40400-51840
| |
Collapse
|
23
|
Jacobs-Cachá C, Puig-Gay N, Helm D, Rettel M, Sellarès J, Meseguer A, Savitski MM, Moreso FJ, Soler MJ, Seron D, Lopez-Hellin J. A misprocessed form of Apolipoprotein A-I is specifically associated with recurrent Focal Segmental Glomerulosclerosis. Sci Rep 2020; 10:1159. [PMID: 31980684 PMCID: PMC6981185 DOI: 10.1038/s41598-020-58197-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 01/10/2020] [Indexed: 12/29/2022] Open
Abstract
Apolipoprotein A-Ib (ApoA-Ib) is a high molecular weight form of Apolipoprotein A-I (ApoA-I) found specifically in the urine of kidney-transplanted patients with recurrent idiopathic focal segmental glomerulosclerosis (FSGS). To determine the nature of the modification present in ApoA-Ib, we sequenced the whole APOA1 gene in ApoA-Ib positive and negative patients, and we also studied the protein primary structure using mass spectrometry. No genetic variations in the APOA1 gene were found in the ApoA-Ib positive patients that could explain the increase in its molecular mass. The mass spectrometry analysis revealed three extra amino acids at the N-Terminal end of ApoA-Ib that were not present in the standard plasmatic form of ApoA-I. These amino acids corresponded to half of the propeptide sequence of the immature form of ApoA-I (proApoA-I) indicating that ApoA-Ib is a misprocessed form of proApoA-I. The description of ApoA-Ib could be relevant not only because it can allow the automated analysis of this biomarker in the clinical practice but also because it has the potential to shed light into the molecular mechanisms that cause idiopathic FSGS, which is currently unknown.
Collapse
Affiliation(s)
- Conxita Jacobs-Cachá
- Nephrology Research Group, Hospital Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain. .,Nephrology Department, Hospital Vall d'Hebrón, Barcelona, Spain.
| | - Natàlia Puig-Gay
- Renal Physiopathology Group-CIBBIM. Hospital Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Dominic Helm
- Proteomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Mandy Rettel
- Proteomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Joana Sellarès
- Nephrology Research Group, Hospital Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain.,Nephrology Department, Hospital Vall d'Hebrón, Barcelona, Spain
| | - Anna Meseguer
- Renal Physiopathology Group-CIBBIM. Hospital Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Mikhail M Savitski
- Proteomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany.,Genome Biology, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Francesc J Moreso
- Nephrology Research Group, Hospital Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain.,Nephrology Department, Hospital Vall d'Hebrón, Barcelona, Spain
| | - Maria José Soler
- Nephrology Research Group, Hospital Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain.,Nephrology Department, Hospital Vall d'Hebrón, Barcelona, Spain
| | - Daniel Seron
- Nephrology Research Group, Hospital Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain.,Nephrology Department, Hospital Vall d'Hebrón, Barcelona, Spain
| | - Joan Lopez-Hellin
- Renal Physiopathology Group-CIBBIM. Hospital Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain. .,Biochemistry Department, Hospital Vall d'Hebrón, Barcelona, Spain.
| |
Collapse
|
24
|
Should high molecular weight forms of apolipoprotein A-I be analyzed in urine of relapsing FSGS patients? Pediatr Nephrol 2019; 34:2423-2424. [PMID: 31342149 DOI: 10.1007/s00467-019-04309-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 07/17/2019] [Indexed: 10/26/2022]
|
25
|
Clark AJ, Jabs K, Hunley TE, Jones DP, VanDeVoorde RG, Anderson C, Du L, Zhong J, Fogo AB, Yang H, Kon V. Urinary apolipoprotein AI in children with kidney disease. Pediatr Nephrol 2019; 34:2351-2360. [PMID: 31230128 PMCID: PMC6801060 DOI: 10.1007/s00467-019-04289-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/15/2019] [Accepted: 06/06/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Although high-density lipoprotein (HDL) modulates many cell types in the cardiovascular system, little is known about HDL in the kidney. We assessed urinary excretion of apolipoprotein AI (apoAI), the main protein in HDL. METHODS We enrolled 228 children with various kidney disorders and 40 controls. Urinary apoAI, albumin, and other markers of kidney damage were measured using ELISA, apoAI isoforms with Western blot, and renal biopsies stained for apoAI. RESULTS Patients followed in nephrology clinic had elevated urinary apoAI vs. controls (median 0.074 μg/mg; interquartile range (IQR) 0.0160-0.560, vs. 0.019 μg/mg; IQR 0.004-0.118, p < 0.001). Patients with tubulopathies, renal dysplasia/congenital anomalies of the kidney and urogenital tract, glomerulonephritis, and nephrotic syndrome (NS) in relapse had the greatest elevations (p ≤ 0.01). Patients with NS in remission, nephrolithiasis, polycystic kidney disease, transplant, or hypertension were not different from controls. Although all NS in relapse had higher apoAI excretion than in remission (0.159 vs. 0.0355 μg/mg, p = 0.01), this was largely driven by patients with focal segmental glomerulosclerosis (FSGS). Many patients, especially with FSGS, had increased urinary apoAI isoforms. Biopsies from FSGS patients showed increased apoAI staining at proximal tubule brush border, compared to diffuse cytoplasmic distribution in minimal change disease. CONCLUSIONS Children with kidney disease have variably increased urinary apoAI depending on underlying disease. Urine apoAI is particularly elevated in diseases affecting proximal tubules. Kidney disease is also associated with high molecular weight (HMW) apoAI isoforms in urine, especially FSGS. Whether abnormal urinary apoAI is a marker or contributor to renal disease awaits further study.
Collapse
Affiliation(s)
- Amanda J. Clark
- Monroe Carrell Children’s Hospital at Vanderbilt, Department of Pediatrics
| | - Kathy Jabs
- Monroe Carrell Children’s Hospital at Vanderbilt, Division of Pediatric Nephrology
| | - Tracy E. Hunley
- Monroe Carrell Children’s Hospital at Vanderbilt, Division of Pediatric Nephrology
| | - Deborah P. Jones
- Monroe Carrell Children’s Hospital at Vanderbilt, Division of Pediatric Nephrology
| | - Rene G. VanDeVoorde
- Monroe Carrell Children’s Hospital at Vanderbilt, Division of Pediatric Nephrology
| | - Carl Anderson
- Monroe Carrell Children’s Hospital at Vanderbilt, Division of Pediatric Nephrology
| | - Liping Du
- Vanderbilt Center for Quantitative Sciences, Department of Biostatistics
| | - Jianyong Zhong
- Monroe Carrell Children’s Hospital at Vanderbilt, Division of Pediatric Nephrology,Vanderbilt University Medical Center, Department of Pathology, Microbiology and Immunology
| | - Agnes B. Fogo
- Monroe Carrell Children’s Hospital at Vanderbilt, Division of Pediatric Nephrology,Vanderbilt University Medical Center, Department of Pathology, Microbiology and Immunology,Vanderbilt University Medical Center, Department of Internal Medicine
| | - Haichun Yang
- Monroe Carrell Children’s Hospital at Vanderbilt, Division of Pediatric Nephrology,Vanderbilt University Medical Center, Department of Pathology, Microbiology and Immunology
| | - Valentina Kon
- Division of Pediatric Nephrology, Monroe Carrell Children's Hospital at Vanderbilt, Nashville, TN, USA.
| |
Collapse
|
26
|
Clark AJ, Yang H, Kon V. Urinary apoAl: novel marker of renal disease? Pediatr Nephrol 2019; 34:2425-2426. [PMID: 31402404 PMCID: PMC6801088 DOI: 10.1007/s00467-019-04328-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 07/26/2019] [Accepted: 07/31/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Amanda J. Clark
- Monroe Carrell Children’s Hospital at Vanderbilt, Department of Pediatrics
| | - Haichun Yang
- Monroe Carrell Children’s Hospital at Vanderbilt, Division of Pediatric Nephrology,Vanderbilt University Medical Center, Department of Pathology, Microbiology and Immunology
| | - Valentina Kon
- Department of Pediatrics, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
27
|
Molecular stratification of idiopathic nephrotic syndrome. Nat Rev Nephrol 2019; 15:750-765. [DOI: 10.1038/s41581-019-0217-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2019] [Indexed: 01/03/2023]
|
28
|
Lim WH, Shingde M, Wong G. Recurrent and de novo Glomerulonephritis After Kidney Transplantation. Front Immunol 2019; 10:1944. [PMID: 31475005 PMCID: PMC6702954 DOI: 10.3389/fimmu.2019.01944] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 08/01/2019] [Indexed: 12/13/2022] Open
Abstract
The prevalence, pathogenesis, predictors, and natural course of patients with recurrent glomerulonephritis (GN) occurring after kidney transplantation remains incompletely understood, including whether there are differences in the outcomes and advances in the treatment options of specific GN subtypes, including those with de novo GN. Consequently, the treatment options and approaches to recurrent disease are largely extrapolated from the general population, with responses to these treatments in those with recurrent or de novo GN post-transplantation poorly described. Given a greater understanding of the pathogenesis of GN and the development of novel treatment options, it is conceivable that these advances will result in an improved structure in the future management of patients with recurrent or de novo GN. This review focuses on the incidence, genetics, characteristics, clinical course, and risk of allograft failure of patients with recurrent or de novo GN after kidney transplantation, ascertaining potential disparities between “high risk” disease subtypes of IgA nephropathy, idiopathic membranous glomerulonephritis, focal segmental glomerulosclerosis, and membranoproliferative glomerulonephritis. We will examine in detail the management of patients with high risk GN, including the pre-transplant assessment, post-transplant monitoring, and the available treatment options for disease recurrence. Given the relative paucity of data of patients with recurrent and de novo GN after kidney transplantation, a global effort in collecting comprehensive in-depth data of patients with recurrent and de novo GN as well as novel trial design to test the efficacy of specific treatment strategy in large scale multicenter randomized controlled trials are essential to address the knowledge deficiency in this disease.
Collapse
Affiliation(s)
- Wai H Lim
- Department of Renal Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia.,School of Medicine, University of Western Australia, Perth, WA, Australia
| | - Meena Shingde
- NSW Health Pathology, Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - Germaine Wong
- Sydney School of Public Health, University of Sydney, Sydney, NSW, Australia.,Centre for Transplant and Renal Research, Westmead Hospital, Sydney, NSW, Australia.,Centre for Kidney Research, The Children's Hospital at Westmead, Sydney, NSW, Australia
| |
Collapse
|
29
|
Anigilaje EA, Olutola A. Prospects of genetic testing for steroid-resistant nephrotic syndrome in Nigerian children: a narrative review of challenges and opportunities. Int J Nephrol Renovasc Dis 2019; 12:119-136. [PMID: 31190951 PMCID: PMC6512787 DOI: 10.2147/ijnrd.s193874] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The prevalence of childhood steroid-resistant nephrotic syndrome (SRNS) ranges from 35% to 92%. This steroid resistance among Nigerian children also reflects underlying renal histopathology, revealing a rare minimal-change disease and a varying burden of membranoproliferative glomerulonephritis and focal segmental glomerulosclerosis (FSGS). FSGS tends to progress to end-stage kidney disease, which requires dialysis and/or renal transplantation. While knowledge of the molecular basis of NS is evolving, recent data support the role of mutant genes that otherwise maintain the structural and functional composition of the glomerular filtration barrier to account for many monogenic forms of FSGS. With the advent of next-generation sequencing, >39 genes are currently associated with SRNS, and the number is likely to increase in the near future. Monogenic FSGS is primarily resistant to steroids, and this foreknowledge obviates the need for steroids, other immunosuppressive therapy, and renal biopsy. Therefore, a multidisciplinary collaboration among cell biologists, molecular physiologists, geneticists, and clinicians holds prospects of fine-tuning the management of SRNS caused by known mutant genes. This article describes the genetics of NS/SRNS in childhood and also gives a narrative review of the challenges and opportunities for molecular testing among children with SRNS in Nigeria. For these children to benefit from genetic diagnosis, Nigeria must aspire to have and develop the manpower and infrastructure required for medical genetics and genomic medicine, leveraging on her existing experiences in genomic medicine. Concerted efforts can be put in place to increase the number of enrollees in Nigeria’s National Health Insurance Scheme (NHIS). The scope of the NHIS can be expanded to cater for the expensive bill of genetic testing within or outside the structure of the National Renal Care Policy proposed by Nigerian nephrologists.
Collapse
Affiliation(s)
- Emmanuel Ademola Anigilaje
- Nephrology Unit, Department of Paediatrics, Faculty of Clinical Sciences, College of Health Sciences, University of Abuja, Abuja, Nigeria,
| | | |
Collapse
|
30
|
Mitrofanova A, Molina J, Varona Santos J, Guzman J, Morales XA, Ducasa GM, Bryn J, Sloan A, Volosenco I, Kim JJ, Ge M, Mallela SK, Kretzler M, Eddy S, Martini S, Wahl P, Pastori S, Mendez AJ, Burke GW, Merscher S, Fornoni A. Hydroxypropyl-β-cyclodextrin protects from kidney disease in experimental Alport syndrome and focal segmental glomerulosclerosis. Kidney Int 2018; 94:1151-1159. [PMID: 30301568 PMCID: PMC6278936 DOI: 10.1016/j.kint.2018.06.031] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 06/14/2018] [Accepted: 06/28/2018] [Indexed: 10/28/2022]
Abstract
Studies suggest that altered renal lipid metabolism plays a role in the pathogenesis of diabetic kidney disease and that genetic or pharmacological induction of cholesterol efflux protects from the development of diabetic kidney disease and focal segmental glomerulosclerosis (FSGS). Here we tested whether altered lipid metabolism contributes to renal failure in the Col4a3 knockout mouse model for Alport Syndrome. There was an eight-fold increase in the cholesterol content in renal cortexes of mice with Alport Syndrome. This was associated with increased glomerular lipid droplets and cholesterol crystals. Treatment of mice with Alport Syndrome with hydroxypropyl-β-cyclodextrin (HPβCD) reduced cholesterol content in the kidneys of mice with Alport Syndrome and protected from the development of albuminuria, renal failure, inflammation and tubulointerstitial fibrosis. Cholesterol efflux and trafficking-related genes were primarily affected in mice with Alport Syndrome and were differentially regulated in the kidney cortex and isolated glomeruli. HPβCD also protected from proteinuria and mesangial expansion in a second model of non-metabolic kidney disease, adriamycin-induced nephropathy. Consistent with our experimental findings, microarray analysis confirmed dysregulation of several lipid-related genes in glomeruli isolated from kidney biopsies of patients with primary FSGS enrolled in the NEPTUNE study. Thus, lipid dysmetabolism occurs in non-metabolic glomerular disorders such as Alport Syndrome and FSGS, and HPβCD improves renal function in experimental Alport Syndrome and FSGS.
Collapse
Affiliation(s)
- Alla Mitrofanova
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Judith Molina
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Javier Varona Santos
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Johanna Guzman
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ximena A Morales
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - G Michelle Ducasa
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jonathan Bryn
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Alexis Sloan
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ion Volosenco
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jin-Ju Kim
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Mengyuan Ge
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Shamroop K Mallela
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Matthias Kretzler
- Division of Nephrology, Departments of Internal Medicine and Computational Medicine and Bioinformatics, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Sean Eddy
- Division of Nephrology, Departments of Internal Medicine and Computational Medicine and Bioinformatics, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Sebastian Martini
- Division of Nephrology, Departments of Internal Medicine and Computational Medicine and Bioinformatics, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Patricia Wahl
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Santiago Pastori
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Armando J Mendez
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - George W Burke
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA; Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA.
| |
Collapse
|
31
|
Puig-Gay N, Jacobs-Cacha C, Sellarès J, Guirado L, González Roncero F, Jiménez C, Zárraga S, Paul J, Lauzurica R, Alonso Á, Fernández A, Beneyto I, Mazuecos A, Hernández D, Rodriguez-Benot A, Franco A, Jimeno L, Crespo M, Meseguer A, Moreso F, Seron D, Lopez-Hellin J, Cantarell C. Apolipoprotein A-Ib as a biomarker of focal segmental glomerulosclerosis recurrence after kidney transplantation: diagnostic performance and assessment of its prognostic value - a multi-centre cohort study. Transpl Int 2018; 32:313-322. [PMID: 30411406 DOI: 10.1111/tri.13372] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/05/2018] [Accepted: 10/31/2018] [Indexed: 01/02/2023]
Abstract
Recurrence of idiopathic focal segmental glomerulosclerosis (FSGS) is a serious complication after kidney transplantation. FSGS relapse is suspected by a sudden increase in proteinuria but there is not an accurate noninvasive diagnostic tool to confirm this entity or to detect patients at risk. We aimed to validate the diagnostic performance of ApoA-Ib to detect FSGS relapses by measuring urinary ApoA-Ib in a retrospective cohort of 61 kidney transplanted patients (37 FSGS and 24 non-FSGS). In addition, to assess the ApoA-Ib predictive ability, ApoA-Ib was measured periodically in a prospective cohort of 13 idiopathic FSGS patients who were followed during 1 year after transplantation. ApoA-Ib had a sensitivity of 93.3% and a specificity of 90.9% to diagnose FSGS relapses, with a high negative predictive value (95.2%), confirming our previous results. In the prospective cohort, ApoA-Ib predated the recurrence in four of five episodes observed. In the nonrelapsing group (n = 9), ApoA-Ib was negative in 37 of 38 samples. ApoA-Ib has the potential to be a good diagnostic biomarker of FSGS relapses, providing a confident criterion to exclude false positives even in the presence of high proteinuria. It has also the potential to detect patients at risk of relapse, even before transplantation.
Collapse
Affiliation(s)
- Natàlia Puig-Gay
- Renal Physiopathology Group-CIBBIM, Hospital Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Conxita Jacobs-Cacha
- Renal Physiopathology Group-CIBBIM, Hospital Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain.,Nephrology, Hospital Vall d'Hebron, Barcelona, Spain
| | | | | | | | - Carlos Jiménez
- Nephrology, Hospital Universitario La Paz, Madrid, Spain
| | | | - Javier Paul
- Nephrology, Hospital Miguel Servet, Zaragoza, Spain
| | | | - Ángel Alonso
- Nephrology, Hospital Universitario de A Coruña, A Coruña, Spain
| | | | - Isabel Beneyto
- Nephrology, Hospital Universitari i Politecnic La Fe, Valencia, Spain
| | | | | | | | | | - Luisa Jimeno
- Nephrology, Hospital Virgen de la Arrixaca, Murcia, Spain
| | | | - Anna Meseguer
- Renal Physiopathology Group-CIBBIM, Hospital Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | | | - Daniel Seron
- Nephrology, Hospital Vall d'Hebron, Barcelona, Spain
| | - Joan Lopez-Hellin
- Renal Physiopathology Group-CIBBIM, Hospital Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | | |
Collapse
|
32
|
Wen Y, Shah S, Campbell KN. Molecular Mechanisms of Proteinuria in Focal Segmental Glomerulosclerosis. Front Med (Lausanne) 2018; 5:98. [PMID: 29713631 PMCID: PMC5912003 DOI: 10.3389/fmed.2018.00098] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 03/26/2018] [Indexed: 01/01/2023] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) is the most common primary glomerular disease resulting in end-stage renal disease in the USA and is increasing in prevalence worldwide. It is a diverse clinical entity with idiopathic, genetic, metabolic, infectious, and other causes that culminate in a characteristic histologic pattern of injury. Proteinuria is a hallmark of FSGS as well as other primary and secondary glomerular disorders. The magnitude of proteinuria at disease onset and during treatment has prognostic implications for renal survival as well as associated cardiovascular morbidity and mortality. Significant advances over the last two decades have shed light on the molecular architecture of the glomerular filtration barrier. The podocyte is the target cell for injury in FSGS. A growing list of disease-causing gene mutations encoding proteins that regulate podocyte survival and homeostasis has been identified in FSGS patients. Several pathogenic and regulatory pathways have been uncovered that result in proteinuria in rodent models and human FSGS. The recurrence of proteinuria and FSGS after kidney transplantation is supporting evidence for the role of a circulating permeability factor in disease pathogenesis. These advances reviewed herein have significant implications for disease classification and therapeutic drug development for FSGS.
Collapse
Affiliation(s)
- Yumeng Wen
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Sapna Shah
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kirk N Campbell
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
33
|
Affiliation(s)
- Arjun Chakraborty
- Department of Surgery, University of California San Francisco, San Francisco, USA
| | - Minnie Sarwal
- Director of Precision Transplant Medicine, University of California San Francisco, San Francisco, USA
| |
Collapse
|
34
|
Multiple Targets for Novel Therapy of FSGS Associated with Circulating Permeability Factor. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6232616. [PMID: 28951873 PMCID: PMC5603123 DOI: 10.1155/2017/6232616] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 05/10/2017] [Accepted: 06/15/2017] [Indexed: 01/13/2023]
Abstract
A plasma component is responsible for altered glomerular permeability in patients with focal segmental glomerulosclerosis. Evidence includes recurrence after renal transplantation, remission after plasmapheresis, proteinuria in infants of affected mothers, transfer of proteinuria to experimental animals, and impaired glomerular permeability after exposure to patient plasma. Therapy may include decreasing synthesis of the injurious agent, removing or blocking its interaction with cells, or blocking signaling or enhancing cell defenses to restore the permeability barrier and prevent progression. Agents that may prevent the synthesis of the permeability factor include cytotoxic agents or aggressive chemotherapy. Extracorporeal therapies include plasmapheresis, immunoadsorption with protein A or anti-immunoglobulin, or lipopheresis. Oral or intravenous galactose also decreases Palb activity. Studies of glomeruli have shown that several strategies prevent the action of FSGS sera. These include blocking receptor-ligand interactions, modulating cell reactions using indomethacin or eicosanoids 20-HETE or 8,9-EET, and enhancing cytoskeleton and protein interactions using calcineurin inhibitors, glucocorticoids, or rituximab. We have identified cardiotrophin-like cytokine factor 1 (CLCF-1) as a candidate for the permeability factor. Therapies specific to CLCF-1 include potential use of cytokine receptor-like factor (CRLF-1) and inhibition of Janus kinase 2. Combined therapy using multiple modalities offers therapy to reverse proteinuria and prevent scarring.
Collapse
|
35
|
Beaudreuil S, Lorenzo HK, Elias M, Nnang Obada E, Charpentier B, Durrbach A. Optimal management of primary focal segmental glomerulosclerosis in adults. Int J Nephrol Renovasc Dis 2017; 10:97-107. [PMID: 28546764 PMCID: PMC5436760 DOI: 10.2147/ijnrd.s126844] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) is a frequent glomerular kidney disease that is revealed by proteinuria or even nephrotic syndrome. A diagnosis can be established from a kidney biopsy that shows focal and segmental glomerulosclerosis. This histopathological lesion may be caused by a primary podocyte injury (idiopathic FSGS) but is also associated with other pathologies (secondary FSGS). The first-line treatment for idiopathic FSGS with nephrotic syndrome is a prolonged course of corticosteroids. However, steroid resistance or steroid dependence is frequent, and despite intensified immunosuppressive treatment, FSGS can lead to end-stage renal failure. In addition, in some cases, FSGS can recur on a graft after kidney transplantation: an unidentified circulating factor may be implicated. Understanding of its physiopathology is unclear, and it remains an important challenge for the scientific community to identify a specific diagnostic biomarker and to develop specific therapeutics. This study reviews the treatment of primary FSGS and the recurrence of FSGS after kidney transplantation in adults.
Collapse
Affiliation(s)
- Séverine Beaudreuil
- Department of Nephrology Dialysis Transplantation, Paris-Sud University Hospital, Le Kremlin Bicêtre.,INSERM Unit 1197, Paris-Sud University Hospital, Villejuif, France
| | - Hans Kristian Lorenzo
- Department of Nephrology Dialysis Transplantation, Paris-Sud University Hospital, Le Kremlin Bicêtre.,INSERM Unit 1197, Paris-Sud University Hospital, Villejuif, France
| | - Michele Elias
- Department of Nephrology Dialysis Transplantation, Paris-Sud University Hospital, Le Kremlin Bicêtre
| | - Erika Nnang Obada
- Department of Nephrology Dialysis Transplantation, Paris-Sud University Hospital, Le Kremlin Bicêtre
| | - Bernard Charpentier
- Department of Nephrology Dialysis Transplantation, Paris-Sud University Hospital, Le Kremlin Bicêtre.,INSERM Unit 1197, Paris-Sud University Hospital, Villejuif, France
| | - Antoine Durrbach
- Department of Nephrology Dialysis Transplantation, Paris-Sud University Hospital, Le Kremlin Bicêtre.,INSERM Unit 1197, Paris-Sud University Hospital, Villejuif, France
| |
Collapse
|
36
|
Abstract
Focal segmental glomerulosclerosis (FSGS) is a leading cause of kidney disease worldwide. The presumed etiology of primary FSGS is a plasma factor with responsiveness to immunosuppressive therapy and a risk of recurrence after kidney transplant-important disease characteristics. In contrast, adaptive FSGS is associated with excessive nephron workload due to increased body size, reduced nephron capacity, or single glomerular hyperfiltration associated with certain diseases. Additional etiologies are now recognized as drivers of FSGS: high-penetrance genetic FSGS due to mutations in one of nearly 40 genes, virus-associated FSGS, and medication-associated FSGS. Emerging data support the identification of a sixth category: APOL1 risk allele-associated FSGS in individuals with sub-Saharan ancestry. The classification of a particular patient with FSGS relies on integration of findings from clinical history, laboratory testing, kidney biopsy, and in some patients, genetic testing. The kidney biopsy can be helpful, with clues provided by features on light microscopy (e.g, glomerular size, histologic variant of FSGS, microcystic tubular changes, and tubular hypertrophy), immunofluorescence (e.g, to rule out other primary glomerulopathies), and electron microscopy (e.g., extent of podocyte foot process effacement, podocyte microvillous transformation, and tubuloreticular inclusions). A complete assessment of renal histology is important for establishing the parenchymal setting of segmental glomerulosclerosis, distinguishing FSGS associated with one of many other glomerular diseases from the clinical-pathologic syndrome of FSGS. Genetic testing is beneficial in particular clinical settings. Identifying the etiology of FSGS guides selection of therapy and provides prognostic insight. Much progress has been made in our understanding of FSGS, but important outstanding issues remain, including the identity of the plasma factor believed to be responsible for primary FSGS, the value of routine implementation of genetic testing, and the identification of more effective and less toxic therapeutic interventions for FSGS.
Collapse
Affiliation(s)
- Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland; and
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jeffrey B. Kopp
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland; and
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
37
|
Wahl P, Ducasa GM, Fornoni A. Systemic and renal lipids in kidney disease development and progression. Am J Physiol Renal Physiol 2016; 310:F433-45. [PMID: 26697982 PMCID: PMC4971889 DOI: 10.1152/ajprenal.00375.2015] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 12/22/2015] [Indexed: 12/14/2022] Open
Abstract
Altered lipid metabolism characterizes proteinuria and chronic kidney diseases. While it is thought that dyslipidemia is a consequence of kidney disease, a large body of clinical and experimental studies support that altered lipid metabolism may contribute to the pathogenesis and progression of kidney disease. In fact, accumulation of renal lipids has been observed in several conditions of genetic and nongenetic origins, linking local fat to the pathogenesis of kidney disease. Statins, which target cholesterol synthesis, have not been proven beneficial to slow the progression of chronic kidney disease. Therefore, other therapeutic strategies to reduce cholesterol accumulation in peripheral organs, such as the kidney, warrant further investigation. Recent advances in the understanding of the biology of high-density lipoprotein (HDL) have revealed that functional HDL, rather than total HDL per se, may protect from both cardiovascular and kidney diseases, strongly supporting a role for altered cholesterol efflux in the pathogenesis of kidney disease. Although the underlying pathophysiological mechanisms responsible for lipid-induced renal damage have yet to be uncovered, several studies suggest novel mechanisms by which cholesterol, free fatty acids, and sphingolipids may affect glomerular and tubular cell function. This review will focus on the clinical and experimental evidence supporting a causative role of lipids in the pathogenesis of proteinuria and kidney disease, with a primary focus on podocytes.
Collapse
Affiliation(s)
- Patricia Wahl
- Peggy and Harold Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, Florida
| | - Gloria Michelle Ducasa
- Peggy and Harold Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, Florida
| | - Alessia Fornoni
- Peggy and Harold Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
38
|
Focal segmental glomerular sclerosis: do not overlook the role of immune response. J Nephrol 2016; 29:525-34. [DOI: 10.1007/s40620-016-0272-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/28/2016] [Indexed: 10/22/2022]
|
39
|
Human Urine Proteomics: Analytical Techniques and Clinical Applications in Renal Diseases. INTERNATIONAL JOURNAL OF PROTEOMICS 2015; 2015:782798. [PMID: 26693351 PMCID: PMC4677025 DOI: 10.1155/2015/782798] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 11/09/2015] [Indexed: 12/14/2022]
Abstract
Urine has been in the center of attention among scientists of clinical proteomics in the past decade, because it is valuable source of proteins and peptides with a relative stable composition and easy to collect in large and repeated quantities with a noninvasive procedure. In this review, we discuss technical aspects of urinary proteomics in detail, including sample preparation, proteomic technologies, and their advantage and disadvantages. Several recent experiments are presented which applied urinary proteome for biomarker discovery in renal diseases including diabetic nephropathy, immunoglobulin A (IgA) nephropathy, focal segmental glomerulosclerosis, lupus nephritis, membranous nephropathy, and acute kidney injury. In addition, several available databases in urinary proteomics are also briefly introduced.
Collapse
|
40
|
Differentially expressed urinary biomarkers in children with idiopathic nephrotic syndrome. Clin Exp Nephrol 2015; 20:273-83. [DOI: 10.1007/s10157-015-1162-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 08/25/2015] [Indexed: 01/01/2023]
|
41
|
Dynamic changes of urinary proteins in focal segmental glomerulosclerosis model. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 845:167-73. [PMID: 25355579 DOI: 10.1007/978-94-017-9523-4_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Compare to blood, which has mechanisms to maintain homeostasis, urine is more likely to reflect changes in the body. As urine accumulates all types of changes, identifying the precise cause of changes in the urine proteome is challenging and crucial in biomarker discovery. To reduce the confounding factors to minimal, some studies used animal model resembling human diseases. This chapter highlights the importance of animal models and introduces a strategic research which focused on adriamycin-induced nephropathy. In this study, urine samples were collected at before adriamycin administration and days 3, 7, 11, 15, and 23 after, urinary proteins were profiled by LC-MS/MS. Of 23 changed proteins with disease development, 13 proteins were identified as stable in normal human urine, meaning that changes in these proteins are more likely to reflect disease. We think this stage-dependent dynamic changes of urine proteome in animal models will help to support the role of urine as key source in biomarker discovery especially in kidney diseases and help to identify corresponding biomarkers for clinical validation.
Collapse
|
42
|
Reiser J, Nast CC, Alachkar N. Permeability factors in focal and segmental glomerulosclerosis. Adv Chronic Kidney Dis 2014; 21:417-21. [PMID: 25168830 DOI: 10.1053/j.ackd.2014.05.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 05/30/2014] [Accepted: 05/30/2014] [Indexed: 01/30/2023]
Abstract
Focal and segmental glomerulosclerosis (FSGS) represents a group of glomerular disorders, identified on kidney biopsy, that progress in the histopathologic pattern of sclerosis in parts of some glomeruli. Damage to podocytes usually marks the beginning of the disease, most evident in primary FSGS. In addition to genetic predisposition, there are many acquired causes that disturb normal podocyte homeostasis and allow for the development of FSGS. The aim of this review was to summarize recent findings of the most relevant circulating permeability factors that may serve as biomarkers of active primary idiopathic FSGS and aid in the diagnosis and prediction of recurrent FSGS after kidney transplantation.
Collapse
|
43
|
Leca N. Focal segmental glomerulosclerosis recurrence in the renal allograft. Adv Chronic Kidney Dis 2014; 21:448-52. [PMID: 25168835 DOI: 10.1053/j.ackd.2014.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 06/06/2014] [Accepted: 06/06/2014] [Indexed: 01/31/2023]
Abstract
Focal segmental glomerulosclerosis (FSGS) represents a common histologic pattern of glomerular injury associated with a multitude of disease mechanisms. The etiology of FSGS is often classified into primary (idiopathic) and secondary forms in response to genetic abnormalities, infections, toxins, and systemic disorders that lead to adaptive changes, glomerular hyperfiltration, and proteinuria. Our understanding of the pathogenic mechanisms responsible for FSGS was substantially enhanced in recent years because of major advances in the cell biology of the podocyte and parietal epithelial cell. Recurrence of FSGS occurs mainly in its primary form and is only rarely described in secondary forms. The re-enactment of pathologic mechanisms of FSGS as recurrent disease after kidney transplantation represents a biologic experiment that can provide unique insight. Nonetheless, recurrent FSGS remains a notable clinical problem that correlates with poorer renal allograft outcomes. This is the focus of this particular review, concentrating on the most recent developments.
Collapse
|
44
|
Fibrinogen alpha chain precursor and apolipoprotein A-I in urine as biomarkers for noninvasive diagnosis of calcium oxalate nephrolithiasis: a proteomics study. BIOMED RESEARCH INTERNATIONAL 2014; 2014:415651. [PMID: 25147800 PMCID: PMC4132411 DOI: 10.1155/2014/415651] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/08/2014] [Accepted: 07/04/2014] [Indexed: 02/08/2023]
Abstract
Calcium oxalate nephrolithiasis is the most common urological disease, but noninvasive and convenient methods of diagnosis are rarely available. Objective. The present study aimed to identify potential urine biomarkers for noninvasive diagnosis of CaOx nephrolithiasis. Methodology. Urine samples from 72 patients with CaOx nephrolithiasis and 30 healthy controls were collected and proteomics analysis was performed using matrix-assisted laser desorption/ionization-time of flight-mass spectrometer (MALDI-TOF-MS). Results. Thirteen proteins/peptides displayed statistically significant differences. The peptides of m/z 1207.23 and 2773.86 were selected by the genetic algorithm (GA) to build a possible diagnostic model. The area under the curve of m/z 1207.23 and 2773.86 was 0.936 and 0.987, respectively. The diagnostic model in distinguishing patients and healthy subjects showed 100% sensitivity and specificity. The peak at m/z 2773.86 was identified as fibrinogen alpha chain (FGA) with the sequence G.EGDFLAEGGGVR.G, and the peak at m/z 2773.86 was identified as apolipoprotein A-I (apoA-I) with the sequence L.PVLESFKVSFLSALEEYTKKLNTQ. Conclusion. The study results strongly suggested that urinary FGA and apoA-I are highly sensitive and specific biomarkers for noninvasive diagnosis of CaOx nephrolithiasis.
Collapse
|
45
|
Dynamic changes of urinary proteins in a focal segmental glomerulosclerosis rat model. Proteome Sci 2014; 12:42. [PMID: 25061428 PMCID: PMC4109389 DOI: 10.1186/1477-5956-12-42] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 07/14/2014] [Indexed: 12/20/2022] Open
Abstract
Background In contrast to blood, which has mechanisms to maintain a homeostatic internal environment, urine is more likely to reflect changes in the body. As urine accumulates all types of changes, identifying the precise cause of changes in the urine proteome is challenging and crucial in biomarker discovery. To reduce the effects of both genetic and environmental factors on the urinary proteome, this study used a rat model of adriamycin-induced nephropathy resembling human focal segmental glomerulosclerosis (FSGS) development. Results Urine samples were collected at before adriamycin administration and day3, 7, 11, 15 and 23 after. Urinary proteins were profiled by liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). Of 23 changed proteins with disease development, 20 have human orthologs, and 13 proteins were identified as stable in normal human urine, meaning that changes in these proteins are more likely to reflect disease. Fifteen of the identified proteins have not been established to function in FSGS development. Seven proteins were selected for verification in ten more rats as markers closely associated with disease severity by western blot. Conclusion We identified proteins changed in different stages of FSGS in rat models, which may aid in biomarker development and the understanding of FSGS pathogenesis.
Collapse
|
46
|
The novel diagnostic biomarkers for focal segmental glomerulosclerosis. Int J Nephrol 2014; 2014:574261. [PMID: 24790760 PMCID: PMC3984796 DOI: 10.1155/2014/574261] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 02/21/2014] [Accepted: 02/24/2014] [Indexed: 11/17/2022] Open
Abstract
Background. Focal segmental glomerulosclerosis (FSGS) is a glomerular injury with various pathogenic mechanisms. Urine proteome panel might help in noninvasive diagnosis and better understanding of pathogenesis of FSGS. Method. We have analyzed the urine sample of 11 biopsy-proven FSGS subjects, 8 healthy controls, and 6 patients with biopsy-proven IgA nephropathy (disease controls) by means of liquid chromatography tandem mass spectrometry (nLC-MS/MS). Multivariate analysis of quantified proteins was performed by principal component analysis (PCA) and partial least squares (PLS). Results. Of the total number of 389 proteins, after multivariate analysis and additional filter criterion and comparing FSGS versus IgA nephropathy and healthy subjects, 77 proteins were considered as putative biomarkers of FSGS. CD59, CD44, IBP7, Robo4, and DPEP1 were the most significant differentially expressed proteins. These proteins are involved in pathogenic pathways: complement pathway, sclerosis, cell proliferation, actin cytoskeleton remodeling, and activity of TRPC6.There was complete absence of DPEP1 in urine proteome of FSGS subjects compared with healthy and disease controls. DPEP1 acts via leukotrienes on TRPC6 and results in increased podocyte motility and proteinuria. Conclusion. The results suggest a panel of candidate biomarkers for noninvasive diagnosis of FSGS, while complete absence of DPEP1 might represent a novel marker of FSGS.
Collapse
|