51
|
Speeckaert MM, Seegmiller J, Glorieux G, Lameire N, Van Biesen W, Vanholder R, Delanghe JR. Measured Glomerular Filtration Rate: The Query for a Workable Golden Standard Technique. J Pers Med 2021; 11:949. [PMID: 34683089 PMCID: PMC8541429 DOI: 10.3390/jpm11100949] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 01/12/2023] Open
Abstract
Inulin clearance has, for a long time, been considered as the reference method to determine measured glomerular filtration rates (mGFRs). However, given the known limitations of the standard marker, serum creatinine, and of inulin itself, and the frequent need for accurate GFR estimations, several other non-radioactive (iohexol and iothalamate) and radioactive (51Cr-EDTA, 99mTc-DTPA, 125I iothalamate) exogenous mGFR filtration markers are nowadays considered the most accurate options to evaluate GFR. The availability of 51Cr-EDTA is limited, and all methods using radioactive tracers necessitate specific safety precautions. Serum- or plasma-based certified reference materials for iohexol and iothalamate and evidence-based protocols to accurately and robustly measure GFR (plasma vs. urinary clearance, single-sample vs. multiple-sample strategy, effect of sampling time delay) are lacking. This leads to substantial variation in reported mGFR results across studies and questions the scientific reliability of the alternative mGFR methods as the gold standard to evaluate kidney function. On top of the scientific discussion, regulatory issues are further narrowing the clinical use of mGFR methods. Therefore, this review is a call for standardization of mGFR in terms of three aspects: the marker, the analytical method to assess concentrations of that marker, and the procedure to determine GFR in practice. Moreover, there is also a need for an endogenous filtration marker or a panel of filtration markers from a single blood draw that would allow estimation of GFR as accurately as mGFR, and without the need for application of anthropometric, clinical, and demographic characteristics.
Collapse
Affiliation(s)
- Marijn M. Speeckaert
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium; (G.G.); (N.L.); (W.V.B.); (R.V.)
- Research Foundation Flanders, 1000 Brussels, Belgium
| | - Jesse Seegmiller
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Griet Glorieux
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium; (G.G.); (N.L.); (W.V.B.); (R.V.)
| | - Norbert Lameire
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium; (G.G.); (N.L.); (W.V.B.); (R.V.)
| | - Wim Van Biesen
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium; (G.G.); (N.L.); (W.V.B.); (R.V.)
| | - Raymond Vanholder
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium; (G.G.); (N.L.); (W.V.B.); (R.V.)
| | - Joris R. Delanghe
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, Belgium;
| |
Collapse
|
52
|
Kruglova MP, Ivanov AV, Virus ED, Bulgakova PO, Samokhin AS, Fedoseev AN, Grachev SV, Kubatiev AA. Urine S-Adenosylmethionine are Related to Degree of Renal Insufficiency in Patients with Chronic Kidney Disease. Lab Med 2021; 52:47-56. [PMID: 32702115 DOI: 10.1093/labmed/lmaa034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE To determine whether urine S-adenosylmethionine (SAM) might be an indicator of chronic kidney disease (CKD). METHODS We investigated urine levels of SAM and related metabolites (S-adenosylhomocysteine and homocysteine cysteine) in 62 patients (average age, 65.9 years) with CKD (stages II-V). RESULTS Patients with stages III-V CKD stages have significantly decreased urine levels and SAM/S-adenosylhomocysteine ratio and also cysteine/homocysteine ratio in blood plasma (P <.05), compared with patients with stage II CKD. Urine SAM levels allowed us to distinguish patients with mildly decreased kidney function from those with moderate to severe renal impairment (AUC, 0.791; sensitivity, 85%; specificity, 78.6%). CONCLUSIONS Our study results demonstrate that urine SAM is a potent biomarker for monitoring renal function decline at early CKD stages. Urine SAM testing confers an additional advantage to healthcare professionals in that it is noninvasive.
Collapse
Affiliation(s)
| | | | - Edward Danielevich Virus
- Department of Molecular and Cell Pathophysiology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Polina Olegovna Bulgakova
- Department of Molecular and Cell Pathophysiology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Andrey Segeevich Samokhin
- Department of Molecular and Cell Pathophysiology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | | | | | - Aslan Amirkhanovich Kubatiev
- Department of Molecular and Cell Pathophysiology, Institute of General Pathology and Pathophysiology, Moscow, Russia.,Russian Medical Academy of Postdoctoral Education, Moscow, Russia
| |
Collapse
|
53
|
Siwy J, Wendt R, Albalat A, He T, Mischak H, Mullen W, Latosinska A, Lübbert C, Kalbitz S, Mebazaa A, Peters B, Stegmayr B, Spasovski G, Wiech T, Staessen JA, Wolf J, Beige J. CD99 and polymeric immunoglobulin receptor peptides deregulation in critical COVID-19: A potential link to molecular pathophysiology? Proteomics 2021; 21:e2100133. [PMID: 34383378 PMCID: PMC8420529 DOI: 10.1002/pmic.202100133] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/28/2021] [Accepted: 08/09/2021] [Indexed: 11/11/2022]
Abstract
Identification of significant changes in urinary peptides may enable improved understanding of molecular disease mechanisms. We aimed towards identifying urinary peptides associated with critical course of COVID-19 to yield hypotheses on molecular pathophysiological mechanisms in disease development. In this multicentre prospective study urine samples of PCR-confirmed COVID-19 patients were collected in different centres across Europe. The urinary peptidome of 53 patients at WHO stages 6-8 and 66 at WHO stages 1-3 COVID-19 disease was analysed using capillary electrophoresis coupled to mass spectrometry. 593 peptides were identified significantly affected by disease severity. These peptides were compared with changes associated with kidney disease or heart failure. Similarities with kidney disease were observed, indicating comparable molecular mechanisms. In contrast, convincing similarity to heart failure could not be detected. The data for the first time showed deregulation of CD99 and polymeric immunoglobulin receptor peptides and of known peptides associated with kidney disease, including collagen and alpha-1-antitrypsin. Peptidomic findings were in line with the pathophysiology of COVID-19. The clinical corollary is that COVID-19 induces specific inflammation of numerous tissues including endothelial lining. Restoring these changes, especially in CD99, PIGR and alpha-1-antitripsin, may represent a valid and effective therapeutic approach in COVID-19, targeting improvement of endothelial integrity. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
| | - Ralph Wendt
- Department of Infectious Diseases/Tropical Medicine, Nephrology and Rheumatology, Hospital St. Georg, Leipzig, Germany
| | - Amaya Albalat
- School of Natural Sciences, University of Stirling, Stirling, UK
| | - Tianlin He
- Mosaiques diagnostics GmbH, Hannover, Germany
| | | | - William Mullen
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, UK
| | | | - Christoph Lübbert
- Department of Infectious Diseases/Tropical Medicine, Nephrology and Rheumatology, Hospital St. Georg, Leipzig, Germany.,Division of Infectious Diseases and Tropical Medicine, Department of Oncology, Gastroenterology, Hepatology, Pneumology and Infectious Diseases, Leipzig University Hospital, Leipzig, Germany.,Interdisciplinary Center for Infectious Diseases, Leipzig University Hospital, Leipzig, Germany
| | - Sven Kalbitz
- Department of Infectious Diseases/Tropical Medicine, Nephrology and Rheumatology, Hospital St. Georg, Leipzig, Germany
| | - Alexandre Mebazaa
- Department of Anesthesiology and Intensive Care, Saint Louis-Lariboisière - Fernand Widal University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France.,Université de Paris, Paris, France.,INSERM UMR-S 942 - MASCOT, Paris, France
| | - Björn Peters
- Department of Nephrology, Skaraborg Hospital, Skövde, Sweden.,Department of Molecular and Clinical Medicine, Institute of Medicine, the Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Bernd Stegmayr
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Goce Spasovski
- Department of Nephrology, Medical Faculty, University St.Cyril and Methodius, Umeå, Sweden
| | - Thorsten Wiech
- Nephropathology Section, Institute for Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan A Staessen
- Alliance for the Promotion of Preventive Medicine (APPREMED), Mechelen, Belgium.,Biomedical Sciences Group, Faculty of Medicine, University of Leuven, Leuven, Belgium
| | - Johannes Wolf
- Department of Laboratory Medicine, Hospital St. Georg, Leipzig, Germany.,ImmunoDeficiencyCenter Leipzig (IDCL) at Hospital St. Georg Leipzig, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Leipzig, Germany
| | - Joachim Beige
- Department of Infectious Diseases/Tropical Medicine, Nephrology and Rheumatology, Hospital St. Georg, Leipzig, Germany.,Kuratorium for Dialysis and Transplantation (KfH) Renal Unit, Hospital St. Georg, Leipzig, Germany.,Martin-Luther-University Halle/Wittenberg, Halle/Saale, Germany
| |
Collapse
|
54
|
Urine peptidome analysis in cardiorenal syndrome reflects molecular processes. Sci Rep 2021; 11:16219. [PMID: 34376786 PMCID: PMC8355128 DOI: 10.1038/s41598-021-95695-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023] Open
Abstract
The cardiorenal syndrome (CRS) is defined as the confluence of heart-kidney dysfunction. This study investigates the molecular differences at the level of the urinary peptidome between CRS patients and controls and their association to disease pathophysiology. The urinary peptidome of CRS patients (n = 353) was matched for age and sex with controls (n = 356) at a 1:1 ratio. Changes in the CRS peptidome versus controls were identified after applying the Mann-Whitney test, followed by correction for multiple testing. Proteasix tool was applied to investigate predicted proteases involved in CRS-associated peptide generation. Overall, 559 differentially excreted urinary peptides were associated with CRS patients. Of these, 193 peptides were specifically found in CRS when comparing with heart failure and chronic kidney disease urinary peptide profiles. Proteasix predicted 18 proteases involved in > 1% of proteolytic cleavage events including multiple forms of MMPs, proprotein convertases, cathepsins and kallikrein 4. Forty-four percent of the cleavage events were produced by 3 proteases including MMP13, MMP9 and MMP2. Pathway enrichment analysis supported that ECM-related pathways, fibrosis and inflammation were represented. Collectively, our study describes the changes in urinary peptides of CRS patients and potential proteases involved in their generation, laying the basis for further validation.
Collapse
|
55
|
Catanese L, Siwy J, Mavrogeorgis E, Amann K, Mischak H, Beige J, Rupprecht H. A Novel Urinary Proteomics Classifier for Non-Invasive Evaluation of Interstitial Fibrosis and Tubular Atrophy in Chronic Kidney Disease. Proteomes 2021; 9:32. [PMID: 34287333 PMCID: PMC8293473 DOI: 10.3390/proteomes9030032] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 12/25/2022] Open
Abstract
Non-invasive urinary peptide biomarkers are able to detect and predict chronic kidney disease (CKD). Moreover, specific urinary peptides enable discrimination of different CKD etiologies and offer an interesting alternative to invasive kidney biopsy, which cannot always be performed. The aim of this study was to define a urinary peptide classifier using mass spectrometry technology to predict the degree of renal interstitial fibrosis and tubular atrophy (IFTA) in CKD patients. The urinary peptide profiles of 435 patients enrolled in this study were analyzed using capillary electrophoresis coupled with mass spectrometry (CE-MS). Urine samples were collected on the day of the diagnostic kidney biopsy. The proteomics data were divided into a training (n = 200) and a test (n = 235) cohort. The fibrosis group was defined as IFTA ≥ 15% and no fibrosis as IFTA < 10%. Statistical comparison of the mass spectrometry data enabled identification of 29 urinary peptides with differential occurrence in samples with and without fibrosis. Several collagen fragments and peptide fragments of fetuin-A and others were combined into a peptidomic classifier. The classifier separated fibrosis from non-fibrosis patients in an independent test set (n = 186) with area under the curve (AUC) of 0.84 (95% CI: 0.779 to 0.889). A significant correlation of IFTA and FPP_BH29 scores could be observed Rho = 0.5, p < 0.0001. We identified a peptidomic classifier for renal fibrosis containing 29 peptide fragments corresponding to 13 different proteins. Urinary proteomics analysis can serve as a non-invasive tool to evaluate the degree of renal fibrosis, in contrast to kidney biopsy, which allows repeated measurements during the disease course.
Collapse
Affiliation(s)
- Lorenzo Catanese
- Department of Nephrology, Angiology and Rheumatology, Klinikum Bayreuth GmbH, 95447 Bayreuth, Germany; (L.C.); (H.R.)
- Kuratorium for Dialysis and Transplantation (KfH) Bayreuth, 95445 Bayreuth, Germany
- Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Justyna Siwy
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (E.M.); (H.M.)
| | - Emmanouil Mavrogeorgis
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (E.M.); (H.M.)
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Kerstin Amann
- Department of Nephropathology, Institute of Pathology, University of Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Harald Mischak
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (E.M.); (H.M.)
| | - Joachim Beige
- Department of Infectious Diseases/Tropical Medicine, Nephrology/KfH Renal Unit and Rheumatology, St. Georg Hospital Leipzig, 04129 Leipzig, Germany;
- Kuratorium for Dialysis and Transplantation (KfH) Renal Unit, Hospital St. Georg, 04129 Leipzig, Germany
- Department of Internal Medicine II, Martin-Luther-University Halle/Wittenberg, 06108 Halle/Saale, Germany
| | - Harald Rupprecht
- Department of Nephrology, Angiology and Rheumatology, Klinikum Bayreuth GmbH, 95447 Bayreuth, Germany; (L.C.); (H.R.)
- Kuratorium for Dialysis and Transplantation (KfH) Bayreuth, 95445 Bayreuth, Germany
- Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
56
|
Farzamikia N, Baradaran B, Mostafavi S, Ahmadian E, Hosseiniyan Khatibi SM, Zununi Vahed S, Ardalan M. Podocyte-derived microparticles in IgA nephropathy. Biomed Pharmacother 2021; 141:111891. [PMID: 34237594 DOI: 10.1016/j.biopha.2021.111891] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/06/2021] [Accepted: 06/28/2021] [Indexed: 12/21/2022] Open
Abstract
Microparticles are a general term for different types of cell plasma membrane-originated vesicles that are released into the extracellular environment. The paracrine action of these nano-sized vesicles is crucial for intercellular communications through the transfer of diverse lipids, cytosolic proteins, RNA as well as microRNAs. The progression of different diseases influences the composition, occurrence, and functions of these cell-derived particles. Podocyte injury has been shown to have an important role in the pathophysiology of many glomerular diseases including IgA nephropathy (IgAN). This review would focus on the possible potential of podocyte-derived microparticles detected in urine to be used as a diagnostic tool in IgAN.
Collapse
Affiliation(s)
- Negin Farzamikia
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soroush Mostafavi
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | |
Collapse
|
57
|
Barutta F, Bellini S, Canepa S, Durazzo M, Gruden G. Novel biomarkers of diabetic kidney disease: current status and potential clinical application. Acta Diabetol 2021; 58:819-830. [PMID: 33528734 DOI: 10.1007/s00592-020-01656-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022]
Abstract
Diabetic kidney disease (DKD) is a leading cause of end-stage renal disease (ESRD). Although both albuminuria and glomerular filtration rate (GFR) are well-established diagnostic/prognostic biomarkers of DKD, they have important limitations. There is, thus, increasing quest to find novel biomarkers to identify the disease in an early stage and to improve risk stratification. In this review, we will outline the major pitfalls of currently available markers, describe promising novel biomarkers, and discuss their potential clinical relevance. In particular, we will focus on the importance of recent advancements in multi-omic technologies in the discovery of new DKD biomarkers. In addition, we will provide an update on new emerging approaches to explore renal function and structure, using functional tests and imaging.
Collapse
Affiliation(s)
- Federica Barutta
- Department of Medical Sciences, University of Turin, Turin, Italy.
| | - Stefania Bellini
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Silvia Canepa
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Marilena Durazzo
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Gabriella Gruden
- Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
58
|
Zheng X, Zhu L, Li T, Xu W, Liu D, Sheng J, Cao H, Shi Y, Wang F. Improve Stability of Bioactive Peptides by Enzymatic Modular Synthesis of Peptides with O-Linked Sialyl Lewis x. ACS Catal 2021. [DOI: 10.1021/acscatal.1c00955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Xiaoju Zheng
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lin Zhu
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Tianlu Li
- Laboratory of Carbohydrate Chemistry and Glycobiology, National Glycoengineering Research Center, Shandong University, Jinan 250012, China
| | - Wenjia Xu
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Dongke Liu
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Juzheng Sheng
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Hongzhi Cao
- Laboratory of Carbohydrate Chemistry and Glycobiology, National Glycoengineering Research Center, Shandong University, Jinan 250012, China
| | - Yikang Shi
- Laboratory of Carbohydrate Chemistry and Glycobiology, National Glycoengineering Research Center, Shandong University, Jinan 250012, China
| | - Fengshan Wang
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Laboratory of Carbohydrate Chemistry and Glycobiology, National Glycoengineering Research Center, Shandong University, Jinan 250012, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate Based Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
59
|
Wendt R, Thijs L, Kalbitz S, Mischak H, Siwy J, Raad J, Metzger J, Neuhaus B, Leyen HVD, Dudoignon E, Mebazaa A, Spasovski G, Milenkova M, Canevska-Talevska A, Czerwieńska B, Wiecek A, Peters B, Nilsson Å, Schwab M, Rothfuss K, Lübbert C, Staessen JA, Beige J. A urinary peptidomic profile predicts outcome in SARS-CoV-2-infected patients. EClinicalMedicine 2021; 36:100883. [PMID: 33969282 PMCID: PMC8092440 DOI: 10.1016/j.eclinm.2021.100883] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/07/2021] [Accepted: 04/16/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND COVID-19 prediction models based on clinical characteristics, routine biochemistry and imaging, have been developed, but little is known on proteomic markers reflecting the molecular pathophysiology of disease progression. METHODS The multicentre (six European study sites) Prospective Validation of a Proteomic Urine Test for Early and Accurate Prognosis of Critical Course Complications in Patients with SARS-CoV-2 Infection Study (Crit-COV-U) is recruiting consecutive patients (≥ 18 years) with PCR-confirmed SARS-CoV-2 infection. A urinary proteomic biomarker (COV50) developed by capillary-electrophoresis-mass spectrometry (CE-MS) technology, comprising 50 sequenced peptides and identifying the parental proteins, was evaluated in 228 patients (derivation cohort) with replication in 99 patients (validation cohort). Death and progression along the World Health Organization (WHO) Clinical Progression Scale were assessed up to 21 days after the initial PCR test. Statistical methods included logistic regression, receiver operating curve (ROC) analysis and comparison of the area under the curve (AUC). FINDINGS In the derivation cohort, 23 patients died, and 48 developed worse WHO scores. The odds ratios (OR) for death per 1 standard deviation (SD) increment in COV50 were 3·52 (95% CI, 2·02-6·13, p <0·0001) unadjusted and 2·73 (1·25-5·95, p = 0·012) adjusted for sex, age, baseline WHO score, body mass index (BMI) and comorbidities. For WHO scale progression, the corresponding OR were 2·63 (1·80-3·85, p<0·0001) and 3·38 (1·85-6·17, p<0·0001), respectively. The area under the curve (AUC) for COV50 as a continuously distributed variable was 0·80 (0·72-0·88) for mortality and 0·74 (0·66-0·81) for worsening WHO score. The optimised COV50 thresholds for mortality and worsening WHO score were 0·47 and 0·04 with sensitivity/specificity of 87·0 (74·6%) and 77·1 (63·9%), respectively. On top of covariates, COV50 improved the AUC, albeit borderline for death, from 0·78 to 0·82 (p = 0·11) and 0·84 (p = 0·052) for mortality and from 0·68 to 0·78 (p = 0·0097) and 0·75 (p = 0·021) for worsening WHO score. The validation cohort findings were confirmatory. INTERPRETATION This first CRIT-COV-U report proves the concept that urinary proteomic profiling generates biomarkers indicating adverse COVID-19 outcomes, even at an early disease stage, including WHO stages 1-3. These findings need to be consolidated in an upcoming final dataset. FUNDING The German Federal Ministry of Health funded the study.
Collapse
Affiliation(s)
- Ralph Wendt
- Department of Infectious Diseases/Tropical Medicine, Nephrology/KfH Renal Unit and Rheumatology, St. Georg Hospital Leipzig, Delitzscher Strasse 141, Leipzig DE 04129, Germany
| | - Lutgarde Thijs
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Diseases, University of Leuven, Belgium
| | - Sven Kalbitz
- Department of Infectious Diseases/Tropical Medicine, Nephrology/KfH Renal Unit and Rheumatology, St. Georg Hospital Leipzig, Delitzscher Strasse 141, Leipzig DE 04129, Germany
| | - Harald Mischak
- Mosaiques-Diagnostics GmbH, Hannover, Germany
- Institute of Cardiovascular and Medical Sciences, Glasgow, United Kingdom
| | | | - Julia Raad
- Mosaiques-Diagnostics GmbH, Hannover, Germany
| | | | - Barbara Neuhaus
- Hannover Clinical Trial Center, Medizinische Hochschule, Hannover, Germany
| | | | - Emmanuel Dudoignon
- Department of Anaesthesiology and Intensive Care, Hôpital Saint Louis-Lariboisière, U942 Inserm MASCOT, Université de Paris, Paris, France
| | - Alexandre Mebazaa
- Department of Anaesthesiology and Intensive Care, Hôpital Saint Louis-Lariboisière, U942 Inserm MASCOT, Université de Paris, Paris, France
| | - Goce Spasovski
- Department Nephrology, Cyril and Methodius University, Skopje, Republic of North Macedonia
| | - Mimoza Milenkova
- Department Nephrology, Cyril and Methodius University, Skopje, Republic of North Macedonia
| | | | - Beata Czerwieńska
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, Katowice, Poland
| | - Andrzej Wiecek
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, Katowice, Poland
| | - Björn Peters
- Department of Nephrology, Skaraborg Hospital, Skövde, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Åsa Nilsson
- Research and Development Centre (FoU), Skaraborg Hospital, Skövde, Sweden
| | - Matthias Schwab
- Margarete-Fischer-Bosch Institute for Clinical Pharmacology and Department for Gastroenterology, Hepatology and Endocrinology, Robert Bosch Hospital, Stuttgart, Germany
- Departments of Clinical Pharmacology, and of Biochemistry and Pharmacy, University of Tuebingen, Germany
| | - Katja Rothfuss
- Margarete-Fischer-Bosch Institute for Clinical Pharmacology and Department for Gastroenterology, Hepatology and Endocrinology, Robert Bosch Hospital, Stuttgart, Germany
| | - Christoph Lübbert
- Department of Infectious Diseases/Tropical Medicine, Nephrology/KfH Renal Unit and Rheumatology, St. Georg Hospital Leipzig, Delitzscher Strasse 141, Leipzig DE 04129, Germany
- Division of Infectious Diseases and Tropical Medicine, Leipzig University Medical Center, Leipzig, Germany
| | - Jan A. Staessen
- Non-Profit Research Institute Alliance for the Promotion of Preventive Medicine, Mechelen, Belgium
- Belgium, Biomedical Sciences Group, Faculty of Medicine, University of Leuven, Leuven, Belgium
| | - Joachim Beige
- Department of Infectious Diseases/Tropical Medicine, Nephrology/KfH Renal Unit and Rheumatology, St. Georg Hospital Leipzig, Delitzscher Strasse 141, Leipzig DE 04129, Germany
- Martin-Luther-University Halle-Wittenberg, Halle an der Saale, Germany
- Corresponding author at: Department of Infectious Diseases/Tropical Medicine, Nephrology/KfH Renal Unit and Rheumatology, St. Georg Hospital Leipzig, Delitzscher Strasse 141, Leipzig DE 04129, Germany.
| | | |
Collapse
|
60
|
Trindade F, Barros AS, Silva J, Vlahou A, Falcão-Pires I, Guedes S, Vitorino C, Ferreira R, Leite-Moreira A, Amado F, Vitorino R. Mining the Biomarker Potential of the Urine Peptidome: From Amino Acids Properties to Proteases. Int J Mol Sci 2021; 22:5940. [PMID: 34073067 PMCID: PMC8197949 DOI: 10.3390/ijms22115940] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/11/2022] Open
Abstract
Native biofluid peptides offer important information about diseases, holding promise as biomarkers. Particularly, the non-invasive nature of urine sampling, and its high peptide concentration, make urine peptidomics a useful strategy to study the pathogenesis of renal conditions. Moreover, the high number of detectable peptides as well as their specificity set the ground for the expansion of urine peptidomics to the identification of surrogate biomarkers for extra-renal diseases. Peptidomics further allows the prediction of proteases (degradomics), frequently dysregulated in disease, providing a complimentary source of information on disease pathogenesis and biomarkers. Then, what does urine peptidomics tell us so far? In this paper, we appraise the value of urine peptidomics in biomarker research through a comprehensive analysis of all datasets available to date. We have mined > 50 papers, addressing > 30 different conditions, comprising > 4700 unique peptides. Bioinformatic tools were used to reanalyze peptide profiles aiming at identifying disease fingerprints, to uncover hidden disease-specific peptides physicochemical properties and to predict the most active proteases associated with their generation. The molecular patterns found in this study may be further validated in the future as disease biomarker not only for kidney diseases but also for extra-renal conditions, as a step forward towards the implementation of a paradigm of predictive, preventive and personalized (3P) medicine.
Collapse
Affiliation(s)
- Fábio Trindade
- UnIC—Cardiovascular Research and Development Centre, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (A.S.B.); (I.F.-P.); (A.L.-M.)
| | - António S. Barros
- UnIC—Cardiovascular Research and Development Centre, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (A.S.B.); (I.F.-P.); (A.L.-M.)
| | - Jéssica Silva
- iBiMED—Department of Medical Sciences, Institute of Biomedicine, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Antonia Vlahou
- Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, 115 27 Athens, Greece;
| | - Inês Falcão-Pires
- UnIC—Cardiovascular Research and Development Centre, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (A.S.B.); (I.F.-P.); (A.L.-M.)
| | - Sofia Guedes
- LAQV-REQUIMTE, Departamento de Química, Universidade de Aveiro, 3810-193 Aveiro, Portugal; (S.G.); (R.F.); (F.A.)
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal;
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal
- Center for Neurosciences and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Rita Ferreira
- LAQV-REQUIMTE, Departamento de Química, Universidade de Aveiro, 3810-193 Aveiro, Portugal; (S.G.); (R.F.); (F.A.)
| | - Adelino Leite-Moreira
- UnIC—Cardiovascular Research and Development Centre, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (A.S.B.); (I.F.-P.); (A.L.-M.)
| | - Francisco Amado
- LAQV-REQUIMTE, Departamento de Química, Universidade de Aveiro, 3810-193 Aveiro, Portugal; (S.G.); (R.F.); (F.A.)
| | - Rui Vitorino
- UnIC—Cardiovascular Research and Development Centre, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (A.S.B.); (I.F.-P.); (A.L.-M.)
- iBiMED—Department of Medical Sciences, Institute of Biomedicine, University of Aveiro, 3810-193 Aveiro, Portugal;
- LAQV-REQUIMTE, Departamento de Química, Universidade de Aveiro, 3810-193 Aveiro, Portugal; (S.G.); (R.F.); (F.A.)
| |
Collapse
|
61
|
He T, Mischak M, Clark AL, Campbell RT, Delles C, Díez J, Filippatos G, Mebazaa A, McMurray JJV, González A, Raad J, Stroggilos R, Bosselmann HS, Campbell A, Kerr SM, Jackson CE, Cannon JA, Schou M, Girerd N, Rossignol P, McConnachie A, Rossing K, Schanstra JP, Zannad F, Vlahou A, Mullen W, Jankowski V, Mischak H, Zhang Z, Staessen JA, Latosinska A. Urinary peptides in heart failure: a link to molecular pathophysiology. Eur J Heart Fail 2021; 23:1875-1887. [PMID: 33881206 PMCID: PMC9291452 DOI: 10.1002/ejhf.2195] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/23/2021] [Accepted: 04/18/2021] [Indexed: 02/06/2023] Open
Abstract
Aims Heart failure (HF) is a major public health concern worldwide. The diversity of HF makes it challenging to decipher the underlying complex pathological processes using single biomarkers. We examined the association between urinary peptides and HF with reduced (HFrEF), mid‐range (HFmrEF) and preserved (HFpEF) ejection fraction, defined based on the European Society of Cardiology guidelines, and the links between these peptide biomarkers and molecular pathophysiology. Methods and results Analysable data from 5608 participants were available in the Human Urinary Proteome database. The urinary peptide profiles from participants diagnosed with HFrEF, HFmrEF, HFpEF and controls matched for sex, age, estimated glomerular filtration rate, systolic and diastolic blood pressure, diabetes and hypertension were compared applying the Mann–Whitney test, followed by correction for multiple testing. Unsupervised learning algorithms were applied to investigate groups of similar urinary profiles. A total of 577 urinary peptides significantly associated with HF were sequenced, 447 of which (77%) were collagen fragments. In silico analysis suggested that urinary biomarker abnormalities in HF principally reflect changes in collagen turnover and immune response, both associated with fibrosis. Unsupervised clustering separated study participants into two clusters, with 83% of non‐HF controls allocated to cluster 1, while 65% of patients with HF were allocated to cluster 2 (P < 0.0001). No separation based on HF subtype was detectable. Conclusions Heart failure, irrespective of ejection fraction subtype, was associated with differences in abundance of urinary peptides reflecting collagen turnover and inflammation. These peptides should be studied as tools in early detection, prognostication, and prediction of therapeutic response.
Collapse
Affiliation(s)
- Tianlin He
- Mosaiques Diagnostics GmbH, Hannover, Germany.,Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University Hospital, Aachen, Germany
| | | | - Andrew L Clark
- Academic Cardiology Department, Hull York Medical School in the University of Hull, Kingston upon Hull, UK
| | - Ross T Campbell
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Javier Díez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, IdiSNA and CIBERCV, Pamplona, Spain.,Departments of Nephrology and Cardiology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gerasimos Filippatos
- Heart Failure Unit, Department of Cardiology, Athens University Hospital Attikon, Athens, Greece
| | - Alexandre Mebazaa
- Université de Paris, Unité Inserm MASCOT, Department of Anaesthesiology and Intensive Care, Saint Louis-Lariboisière - Fernand Widal University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France.,F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
| | - John J V McMurray
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, IdiSNA and CIBERCV, Pamplona, Spain
| | - Julia Raad
- Mosaiques Diagnostics GmbH, Hannover, Germany
| | - Rafael Stroggilos
- Biotechnology Division, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Helle S Bosselmann
- Department of Cardiology, Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics & Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Shona M Kerr
- MRC Human Genetics Unit, Institute of Genetics & Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | | | | | - Morten Schou
- Herlev-Gentofte Hospital, Department of Cardiology, Herlev, Denmark
| | - Nicolas Girerd
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques- Plurithématique 1433, and Inserm 1116 DCAC, CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
| | - Patrick Rossignol
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques- Plurithématique 1433, and Inserm 1116 DCAC, CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
| | - Alex McConnachie
- Robertson Centre for Biostatistics, Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Kasper Rossing
- Department of Cardiology, Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Joost P Schanstra
- Institut National de la Santé et de la Recherche Médicale, U1048, Institute of Cardiovascular and Metabolic Disease, Toulouse, France
| | - Faiez Zannad
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques- Plurithématique 1433, and Inserm 1116 DCAC, CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
| | - Antonia Vlahou
- Biotechnology Division, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - William Mullen
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Vera Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University Hospital, Aachen, Germany
| | - Harald Mischak
- Mosaiques Diagnostics GmbH, Hannover, Germany.,Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Zhenyu Zhang
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Jan A Staessen
- Non-Profit Research Institution Alliance for the Promotion of Preventive Medicine, Mechelen, Belgium.,Biomedical Sciences Group, Faculty of Medicine, University of Leuven, Leuven, Belgium
| | | |
Collapse
|
62
|
Tan S, Zeng Y, Kuang S, Li J. Serum Human Epididymis Protein 4 is a Potential Biomarker for Early Chronic Kidney Disease in an Obese Population. Diabetes Metab Syndr Obes 2021; 14:1601-1608. [PMID: 33889001 PMCID: PMC8057804 DOI: 10.2147/dmso.s300940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/10/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND At present, it is difficult to clinically diagnose early chronic kidney disease (CKD). As a novel biomarker of malignancies in the female reproductive tract, the human epididymis protein 4 (HE4) has been reported to be significantly expressed in CKD patients. AIM We sought to assess whether HE4 can be used as a potential biomarker of early-stage CKD. METHODS The association between serum HE4 levels and CKD was analyzed in a retrospective study. A cohort of 506 patients with diabetic nephropathy who were hospitalized at Weihai Central Hospital, China, from January 2016 to November 2019 were included. RESULTS Serum HE4 levels were increased with increasing stage of CKD and significantly elevated in patients with CKD3-5 than CKD1-2 (P<0.001). In multivariate linear regression analyses, HE4 levels were strongly correlated with the estimated glomerular filtration rate (eGFR) in CKD patients (Model 2, P<0.001). HE4 (area under the curve; AUC=0.934) had better diagnostic value than serum creatinine (SCr; AUC=0.770) and blood urea nitrogen (BUN; AUC=0.647) for patients with early-stage CKD (CKD1-2). Additionally, HE4 levels increased with increasing glomerular lesion (GL) and renal interstitial fibrosis (IF)/tubular atrophy (TA) scores in 51 CKD patients (P<0.001). CONCLUSION Serum HE4 levels can be positively associated with the severity of CKD and are a very valuable clinical biomarker for predicting early-stage CKD.
Collapse
Affiliation(s)
- Shubo Tan
- Department of Urology, Second Affiliated Hospital of the University of South China, Hengyang City, 421000, People’s Republic of China
| | - Yongmao Zeng
- Department of Urology, Second Affiliated Hospital of the University of South China, Hengyang City, 421000, People’s Republic of China
| | - Shiliang Kuang
- Department of Urology, Second Affiliated Hospital of the University of South China, Hengyang City, 421000, People’s Republic of China
| | - Jianjun Li
- Department of Urology, Second Affiliated Hospital of the University of South China, Hengyang City, 421000, People’s Republic of China
| |
Collapse
|
63
|
Tailliar M, Schanstra JP, Dierckx T, Breuil B, Hanouna G, Charles N, Bascands JL, Dussol B, Vazi A, Chiche L, Siwy J, Faguer S, Daniel L, Daugas E, Jourde-Chiche N. Urinary Peptides as Potential Non-Invasive Biomarkers for Lupus Nephritis: Results of the Peptidu-LUP Study. J Clin Med 2021; 10:jcm10081690. [PMID: 33920017 PMCID: PMC8071029 DOI: 10.3390/jcm10081690] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/16/2022] Open
Abstract
Background: Lupus nephritis (LN) is a severe manifestation of Systemic Lupus Erythematosus (SLE). The therapeutic strategy relies on kidney biopsy (KB) results. We tested whether urinary peptidome analysis could non-invasively differentiate active from non-active LN. Design: Urinary samples were collected from 93 patients (55 with active LN and 38 with non-active LN), forming a discovery (n = 42) and an independent validation (n = 51) cohort. Clinical characteristics were collected at inclusion and prospectively for 24 months. The urinary peptidome was analyzed by capillary-electrophoresis coupled to mass-spectrometry, comparing active LN to non-active LN, and assessing chronic lesions and response to therapy. The value of previously validated prognostic (CKD273) and differential diagnostic (LN172) signatures was evaluated. Results: Urinary peptides could not discriminate between active and non-active LN or predict early response to therapy. Tubulo-interstitial fibrosis was correlated to the CKD273. The LN172 score identified 92.5% of samples as LN. Few patients developed new-onset CKD. Conclusions: We validated the CKD273 and LN172 classifiers but did not identify a robust signature that could predict active LN and replace KB. The value of urinary peptidome to predict long-term CKD, or renal flares in SLE, remains to be evaluated.
Collapse
Affiliation(s)
- Maxence Tailliar
- AP-HM, Centre de Néphrologie et Transplantation Rénale, Hôpital de la Conception, 13005 Marseille, France; (M.T.); (B.D.)
| | - Joost P. Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institut of Metabolic and Cardiovascular Disease (I2MC), 31432 Toulouse, France; (J.P.S.); (B.B.); (S.F.)
- Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Tim Dierckx
- Laboratory of Clinical and Epidemiological Virology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium;
| | - Benjamin Breuil
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institut of Metabolic and Cardiovascular Disease (I2MC), 31432 Toulouse, France; (J.P.S.); (B.B.); (S.F.)
- Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Guillaume Hanouna
- AP-HP, Service de Néphrologie, Hôpital Bichat, DMU VICTOIRE, 75018 Paris, France; (G.H.); (E.D.)
| | - Nicolas Charles
- Centre de Recherche sur l’Inflammation, Université de Paris, INSERM UMRS1149, CNRS ERL8252, Labex INFLAMEX, DHU FIRE, 75890 Paris, France;
| | - Jean-Loup Bascands
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1188-Université de La Réunion, 97490 Saint-Denis, France;
| | - Bertrand Dussol
- AP-HM, Centre de Néphrologie et Transplantation Rénale, Hôpital de la Conception, 13005 Marseille, France; (M.T.); (B.D.)
- Centre d’Investigation Clinique, CHU Conception, AP-HM, 13005 Marseille, France;
| | - Alain Vazi
- Centre d’Investigation Clinique, CHU Conception, AP-HM, 13005 Marseille, France;
| | - Laurent Chiche
- Médecine Interne, Hôpital Européen, 13003 Marseille, France;
| | - Justyna Siwy
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany;
| | - Stanislas Faguer
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institut of Metabolic and Cardiovascular Disease (I2MC), 31432 Toulouse, France; (J.P.S.); (B.B.); (S.F.)
- Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
- CHU de Toulouse, Service de Néphrologie, 31300 Toulouse, France
| | - Laurent Daniel
- AP-HM, Laboratoire d’Ananatomie Pathologique, Hôpital de la Timone, 13005 Marseille, France;
- Center for CardioVascular and Nutrition Research (C2VN), Aix-Marseille University, INSERM, INRAE, 13005 Marseille, France
| | - Eric Daugas
- AP-HP, Service de Néphrologie, Hôpital Bichat, DMU VICTOIRE, 75018 Paris, France; (G.H.); (E.D.)
- Centre de Recherche sur l’Inflammation, Université de Paris, INSERM UMRS1149, CNRS ERL8252, Labex INFLAMEX, DHU FIRE, 75890 Paris, France;
| | - Noémie Jourde-Chiche
- AP-HM, Centre de Néphrologie et Transplantation Rénale, Hôpital de la Conception, 13005 Marseille, France; (M.T.); (B.D.)
- Center for CardioVascular and Nutrition Research (C2VN), Aix-Marseille University, INSERM, INRAE, 13005 Marseille, France
- Correspondence:
| | | |
Collapse
|
64
|
Mavrogeorgis E, Mischak H, Beige J, Latosinska A, Siwy J. Understanding glomerular diseases through proteomics. Expert Rev Proteomics 2021; 18:137-157. [PMID: 33779448 DOI: 10.1080/14789450.2021.1908893] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Chronic kidney disease is avery common and complex chronic disease. Uncovering the pathological patterns of CKD on the molecular level of bio-fluids and tissue appears to be both vital and promising for a more favorable outcome. We reviewed recently discovered proteomics biomarkers for CKD to provide new insight into disease pathology. AREAS COVERED We review the application of proteome analysis in the context of CKD with various etiologies within the last 5 years. Proteins and peptides associated with CKD as derived from multiple sources (urine, blood and tissue) are reported along with their various biological pathways. EXPERT OPINION A systematic and theoretical comprehension of the CKD pathology is essential for its successful management. The underlying complexity of the disease further requires specific conditions for reliable and interpretable results. In this context, clinical proteomics has resulted in first encouraging findings in CKD. A more complete understanding of the biological pathways related to the disease, based on the scope of a holistic proteomic approach, could improve substantially the management of CKD, especially when in conjunction with the current trend of personalized medicine.
Collapse
Affiliation(s)
| | - H Mischak
- Mosaiques Diagnostics GmbH, Hannover, Germany.,Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - J Beige
- Division of Nephrology and KfH Renal Unit, Hospital St. Georg, Leipzig, Germany.,Department of Internal Medicine 2 (Nephrology, Rheumatology, Endocrinology), Martin-Luther-University Halle, Wittenberg, Germany
| | | | - J Siwy
- Mosaiques Diagnostics GmbH, Hannover, Germany
| |
Collapse
|
65
|
Abstract
Interstitial fibrosis with tubule atrophy (IF/TA) is the response to virtually any sustained kidney injury and correlates inversely with kidney function and allograft survival. IF/TA is driven by various pathways that include hypoxia, renin-angiotensin-aldosterone system, transforming growth factor (TGF)-β signaling, cellular rejection, inflammation and others. In this review we will focus on key pathways in the progress of renal fibrosis, diagnosis and therapy of allograft fibrosis. This review discusses the role and origin of myofibroblasts as matrix producing cells and therapeutic targets in renal fibrosis with a particular focus on renal allografts. We summarize current trends to use multi-omic approaches to identify new biomarkers for IF/TA detection and to predict allograft survival. Furthermore, we review current imaging strategies that might help to identify and follow-up IF/TA complementary or as alternative to invasive biopsies. We further discuss current clinical trials and therapeutic strategies to treat kidney fibrosis.Supplemental Visual Abstract; http://links.lww.com/TP/C141.
Collapse
|
66
|
Vanholder R, Argilés A, Jankowski J. A history of uraemic toxicity and of the European Uraemic Toxin Work Group (EUTox). Clin Kidney J 2021; 14:1514-1523. [PMID: 34413975 PMCID: PMC8371716 DOI: 10.1093/ckj/sfab011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Indexed: 12/13/2022] Open
Abstract
The uraemic syndrome is a complex clinical picture developing in the advanced stages of chronic kidney disease, resulting in a myriad of complications and a high early mortality. This picture is to a significant extent defined by retention of metabolites and peptides that with a preserved kidney function are excreted or degraded by the kidneys. In as far as those solutes have a negative biological/biochemical impact, they are called uraemic toxins. Here, we describe the historical evolution of the scientific knowledge about uraemic toxins and the role played in this process by the European Uraemic Toxin Work Group (EUTox) during the last two decades. The earliest knowledge about a uraemic toxin goes back to the early 17th century when the existence of what would later be named as urea was recognized. It took about two further centuries to better define the role of urea and its link to kidney failure, and one more century to identify the relevance of post-translational modifications caused by urea such as carbamoylation. The knowledge progressively extended, especially from 1980 on, by the identification of more and more toxins and their adverse biological/biochemical impact. Progress of knowledge was paralleled and impacted by evolution of dialysis strategies. The last two decades, when insights grew exponentially, coincide with the foundation and activity of EUTox. In the final section, we summarize the role and accomplishments of EUTox and the part it is likely to play in future action, which should be organized around focus points like biomarker and potential target identification, intestinal generation, toxicity mechanisms and their correction, kidney and extracorporeal removal, patient-oriented outcomes and toxin characteristics in acute kidney injury and transplantation.
Collapse
Affiliation(s)
- Raymond Vanholder
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Angel Argilés
- RD-Néphrologie, Montpellier, France.,Néphrologie Dialyse St Guilhem, Sète, France
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research, University Hospital, RWTH Aachen, Aachen, Germany.,School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | | |
Collapse
|
67
|
Latosinska A, Siwy J, Faguer S, Beige J, Mischak H, Schanstra JP. Value of Urine Peptides in Assessing Kidney and Cardiovascular Disease. Proteomics Clin Appl 2021; 15:e2000027. [PMID: 32710812 DOI: 10.1002/prca.202000027] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/18/2020] [Indexed: 12/14/2022]
Abstract
Urinary peptides gained significant attention as potential biomarkers especially in the context of kidney and cardiovascular disease. In this manuscript the recent literature since 2015 on urinary peptide investigation in human kidney and cardiovascular disease is reviewed. The technology most commonly used in this context is capillary electrophoresis coupled mass spectrometry, in part owed to the large database available and the well-defined dataspace. Several studies based on over 1000 subjects are reported in the recent past, especially examining CKD273, a classifier for assessment of chronic kidney disease based on 273 urine peptides. Interestingly, the most abundant urinary peptides are generally collagen fragments, which may have gone undetected for some time as they are typically modified via proline hydroxylation. The data available suggest that urinary peptides specifically depict inflammation and fibrosis, and may serve as a non-invasive tool to assess fibrosis, which appears to be a key driver in kidney and cardiovascular disease. The recent successful completion of the first urinary peptide guided intervention trial, PRIORITY, is expected to further spur clinical application of urinary peptidomics, aiming especially at early detection of chronic diseases, prediction of progression, and prognosis of drug response.
Collapse
Affiliation(s)
| | - Justyna Siwy
- Mosaiques Diagnostics GmbH, Rotenburger Straße 20, 30659, Hannover, Germany
| | - Stanislas Faguer
- Département de Néphrologie et Transplantation d'organes, Centre de référence des maladies rénales rares, Centre Hospitalier Universitaire de Toulouse, 1, Avenue Jean Poulhes, Toulouse, 31059, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, 1 Avenue Jean Poulhès, BP 84225, Toulouse Cedex 4, 31432, France
- Université Toulouse III Paul-Sabatier, Route de Narbonne, Toulouse, 31330, France
| | - Joachim Beige
- Department of Nephrology and Kuratorium for Dialysis and Transplantation Renal Unit, Hospital St Georg, Delitzscher Str. 141, 04129, Leipzig, Germany
- Department of Nephrology, Martin-Luther-University Halle/Wittenberg, Universitätsplatz 10, 06108, Halle (Saale), Germany
| | - Harald Mischak
- Mosaiques Diagnostics GmbH, Rotenburger Straße 20, 30659, Hannover, Germany
| | - Joost P Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, 1 Avenue Jean Poulhès, BP 84225, Toulouse Cedex 4, 31432, France
- Université Toulouse III Paul-Sabatier, Route de Narbonne, Toulouse, 31330, France
| |
Collapse
|
68
|
Zhou IY, Montesi SB, Akam EA, Caravan P. Molecular Imaging of Fibrosis. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00077-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
69
|
He T, Pejchinovski M, Mullen W, Beige J, Mischak H, Jankowski V. Peptides in Plasma, Urine, and Dialysate: Toward Unravelling Renal Peptide Handling. Proteomics Clin Appl 2021; 15:e2000029. [PMID: 32618437 DOI: 10.1002/prca.202000029] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/11/2020] [Indexed: 12/28/2022]
Abstract
PURPOSE The peptidomes of spent hemodialysate, urine, and plasma are investigated, to shed light on peptide handling in the kidney. EXPERIMENTAL DESIGN Fifteen plasma, 15 urine, and 13 spent hemodialysate samples are collected from age- and sex-matched subjects with chronic kidney disease. Peptide identification and quantification are performed with capillary electrophoresis-coupled mass spectrometry. RESULTS A total of 6278 urinary peptides, 1743 plasma peptides, and 1727 peptides from spent hemodialysate are detected. Of these, sequences can be assigned to 1580, 419, and 352 peptides, respectively. A strong correlation in peptide abundance between urine and spent hemodialysate (p = 3 × 10-21 , Rho = 0.52), a moderately strong correlation between spent hemodialysate and plasma (p = 4.5 × 10-5 , Rho = 0.30), and no significant correlation between urine and plasma (p = 0.11, Rho = 0.094) are found. Collagen and fibrinogen alpha peptides are highly abundant in all three body fluids. In spent hemodialysate, thymosin ß4 is one of the most abundant peptides, which is shown to be negatively associated with the estimated glomerular filtration rate (Rho = -0.39, p-value = 3.9 × 10-81 ). CONCLUSION AND CLINICAL RELEVANCE The correlation of peptide abundance in these three body fluids is lower than expected, supporting the hypothesis that tubular reabsorption has a major impact on urinary peptide content. Further investigation of thymosin ß4 in hemodialysis is thus warranted.
Collapse
Affiliation(s)
- Tianlin He
- Mosaiques Diagnostics GmbH, Hannover, Germany
- Institute for Molecular Cardiovascular Research (IMCAR), University of Aachen, Aachen, Germany
| | | | - William Mullen
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, UK
| | - Joachim Beige
- Department of Nephrology and Kuratorium for Dialysis and Transplantation (KfH) Renal Unit, Hospital St. Georg, Leipzig, Germany
| | - Harald Mischak
- Mosaiques Diagnostics GmbH, Hannover, Germany
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, UK
| | - Vera Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University of Aachen, Aachen, Germany
| |
Collapse
|
70
|
Beige J, Drube J, von der Leyen H, Pape L, Rupprecht H. Früherkennung mittels Urinproteomanalyse. Internist (Berl) 2020; 61:1094-1105. [DOI: 10.1007/s00108-020-00863-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
71
|
Provenzano M, Rotundo S, Chiodini P, Gagliardi I, Michael A, Angotti E, Borrelli S, Serra R, Foti D, De Sarro G, Andreucci M. Contribution of Predictive and Prognostic Biomarkers to Clinical Research on Chronic Kidney Disease. Int J Mol Sci 2020; 21:E5846. [PMID: 32823966 PMCID: PMC7461617 DOI: 10.3390/ijms21165846] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/09/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic kidney disease (CKD), defined as the presence of albuminuria and/or reduction in estimated glomerular filtration rate (eGFR) < 60 mL/min/1.73 m2, is considered a growing public health problem, with its prevalence and incidence having almost doubled in the past three decades. The implementation of novel biomarkers in clinical practice is crucial, since it could allow earlier diagnosis and lead to an improvement in CKD outcomes. Nevertheless, a clear guidance on how to develop biomarkers in the setting of CKD is not yet available. The aim of this review is to report the framework for implementing biomarkers in observational and intervention studies. Biomarkers are classified as either prognostic or predictive; the first type is used to identify the likelihood of a patient to develop an endpoint regardless of treatment, whereas the second type is used to determine whether the patient is likely to benefit from a specific treatment. Many single assays and complex biomarkers were shown to improve the prediction of cardiovascular and kidney outcomes in CKD patients on top of the traditional risk factors. Biomarkers were also shown to improve clinical trial designs. Understanding the correct ways to validate and implement novel biomarkers in CKD will help to mitigate the global burden of CKD and to improve the individual prognosis of these high-risk patients.
Collapse
Affiliation(s)
- Michele Provenzano
- Renal Unit, Department of Health Sciences, “Magna Graecia” University of Catanzaro, I-88100 Catanzaro, Italy; (I.G.); (A.M.)
| | - Salvatore Rotundo
- Department of Health Sciences, “Magna Graecia” University of Catanzaro, I-88100 Catanzaro, Italy; (S.R.); (D.F.)
| | - Paolo Chiodini
- Medical Statistics Unit, University of Campania Luigi Vanvitelli, I-80138 Naples, Italy;
| | - Ida Gagliardi
- Renal Unit, Department of Health Sciences, “Magna Graecia” University of Catanzaro, I-88100 Catanzaro, Italy; (I.G.); (A.M.)
| | - Ashour Michael
- Renal Unit, Department of Health Sciences, “Magna Graecia” University of Catanzaro, I-88100 Catanzaro, Italy; (I.G.); (A.M.)
| | - Elvira Angotti
- Clinical Biochemistry Unit, Azienda Ospedaliera Universitaria Mater Domini Hospital, I-88100 Catanzaro, Italy;
| | - Silvio Borrelli
- Renal Unit, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy;
| | - Raffaele Serra
- Interuniversity Center of Phlebolymphology (CIFL), “Magna Graecia” University of Catanzaro, I-88100 Catanzaro, Italy;
| | - Daniela Foti
- Department of Health Sciences, “Magna Graecia” University of Catanzaro, I-88100 Catanzaro, Italy; (S.R.); (D.F.)
| | - Giovambattista De Sarro
- Pharmacology Unit, Department of Health Sciences, School of Medicine, “Magna Graecia” University of Catanzaro, I-88100 Catanzaro, Italy;
| | - Michele Andreucci
- Renal Unit, Department of Health Sciences, “Magna Graecia” University of Catanzaro, I-88100 Catanzaro, Italy; (I.G.); (A.M.)
| |
Collapse
|
72
|
Provenzano M, Chiodini P, Minutolo R, Zoccali C, Bellizzi V, Conte G, Locatelli F, Tripepi G, Del Vecchio L, Mallamaci F, Di Micco L, Russo D, Heerspink HJL, De Nicola L. Reclassification of chronic kidney disease patients for end-stage renal disease risk by proteinuria indexed to estimated glomerular filtration rate: multicentre prospective study in nephrology clinics. Nephrol Dial Transplant 2020; 35:138-147. [PMID: 30053127 DOI: 10.1093/ndt/gfy217] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/09/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND In non-dialysis chronic kidney disease (CKD), absolute proteinuria (Uprot) depends on the extent of kidney damage and residual glomerular filtration rate (GFR). We therefore evaluated, as compared with Uprot, the strength of association of proteinuria indexed to estimated GFR (eGFR) with end-stage renal disease (ESRD) risk. METHODS In a multi-cohort prospective study in 3957 CKD patients of Stages G3-G5 referred to nephrology clinics, we tested two multivariable Cox models for ESRD risk, with either Uprot (g/24 h) or filtration-adjusted proteinuria (F-Uprot) calculated as Uprot/eGFR ×100. RESULTS Mean ± SD age was 67 ± 14 years, males 60%, diabetics 29%, cardiovascular disease (CVD) 34%, eGFR 32 ± 13 mL/min/1.73 m2, median (interquartile range) Uprot 0.41 (0.12-1.29) g/24 h and F-Uprot 1.41 (0.36-4.93) g/24 h per 100 mL/min/1.73 m2 eGFR. Over a median follow-up of 44 months, 862 patients reached ESRD. At competing risk analysis, ESRD risk progressively increased when F-Uprot was 1.0-4.9 and ≥5.0 versus <1.0 g/24 h per 100 mL/min/1.73 m2 eGFR in Stages G3a-G4 (P < 0.001) and Stage G5 (P = 0.002). Multivariable Cox analysis showed that Uprot predicts ESRD in Stages G3a-G4 while in G5 the effect was not significant; conversely, F-Uprot significantly predicted ESRD at all stages. The F-Uprot model allowed a significantly better prediction versus the Uprot model according to Akaike information criterion. Net reclassification improvement was 12.2% (95% confidence interval 4.2-21.1), with higher reclassification in elderly, diabetes and CVD, as well as in diabetic nephropathy and glomerulonephritis, and in CKD Stages G4 and G5. CONCLUSIONS In patients referred to nephrology clinics, F-Uprot predicts ESRD at all stages of overt CKD and improves, as compared with Uprot, reclassification of patients for renal risk, especially in more advanced and complicated disease.
Collapse
Affiliation(s)
| | - Paolo Chiodini
- Medical Statistics Unit, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Roberto Minutolo
- Nephrology Unit, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Carmine Zoccali
- Nephrology Center of National Research Institute of Biomedicine and Molecular Immunology, Reggio Calabria, Italy
| | - Vincenzo Bellizzi
- Division of Nephrology, Dialysis and Transplantation, Salerno Medical School, University Hospital San Giovanni di Dio e Ruggi d'Aragona Unit-University, Salerno, Italy
| | - Giuseppe Conte
- Nephrology Unit, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Giovanni Tripepi
- Nephrology Center of National Research Institute of Biomedicine and Molecular Immunology, Reggio Calabria, Italy
| | - Lucia Del Vecchio
- Department of Nephrology and Dialysis, A. Manzoni Hospital, Lecco, Italy
| | - Francesca Mallamaci
- Nephrology Center of National Research Institute of Biomedicine and Molecular Immunology, Reggio Calabria, Italy
| | - Lucia Di Micco
- Division of Nephrology, A. Landolfi Hospital, Solofra, Avellino, Italy
| | - Domenico Russo
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Luca De Nicola
- Nephrology Unit, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
73
|
Klein J, Buffin-Meyer B, Boizard F, Moussaoui N, Lescat O, Breuil B, Fedou C, Feuillet G, Casemayou A, Neau E, Hindryckx A, Decatte L, Levtchenko E, Raaijmakers A, Vayssière C, Goua V, Lucas C, Perrotin F, Cloarec S, Benachi A, Manca-Pellissier MC, Delmas HL, Bessenay L, Le Vaillant C, Allain-Launay E, Gondry J, Boudailliez B, Simon E, Prieur F, Lavocat MP, Saliou AH, De Parscau L, Bidat L, Noel C, Floch C, Bourdat-Michel G, Favre R, Weingertner AS, Oury JF, Baudouin V, Bory JP, Pietrement C, Fiorenza M, Massardier J, Kessler S, Lounis N, Auriol FC, Marcorelles P, Collardeau-Frachon S, Zürbig P, Mischak H, Magalhães P, Batut J, Blader P, Saulnier Blache JS, Bascands JL, Schaefer F, Decramer S, Schanstra JP. Amniotic fluid peptides predict postnatal kidney survival in developmental kidney disease. Kidney Int 2020; 99:737-749. [PMID: 32750455 DOI: 10.1016/j.kint.2020.06.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/10/2020] [Accepted: 06/18/2020] [Indexed: 12/16/2022]
Abstract
Although a rare disease, bilateral congenital anomalies of the kidney and urinary tract (CAKUT) are the leading cause of end stage kidney disease in children. Ultrasound-based prenatal prediction of postnatal kidney survival in CAKUT pregnancies is far from accurate. To improve prediction, we conducted a prospective multicenter peptidome analysis of amniotic fluid spanning 140 evaluable fetuses with CAKUT. We identified a signature of 98 endogenous amniotic fluid peptides, mainly composed of fragments from extracellular matrix proteins and from the actin binding protein thymosin-β4. The peptide signature predicted postnatal kidney outcome with an area under the curve of 0.96 in the holdout validation set of patients with CAKUT with definite endpoint data. Additionally, this peptide signature was validated in a geographically independent sub-cohort of 12 patients (area under the curve 1.00) and displayed high specificity in non-CAKUT pregnancies (82 and 94% in 22 healthy fetuses and in 47 fetuses with congenital cytomegalovirus infection respectively). Change in amniotic fluid thymosin-β4 abundance was confirmed with ELISA. Knockout of thymosin-β4 in zebrafish altered proximal and distal tubule pronephros growth suggesting a possible role of thymosin β4 in fetal kidney development. Thus, recognition of the 98-peptide signature in amniotic fluid during diagnostic workup of prenatally detected fetuses with CAKUT can provide a long-sought evidence base for accurate management of the CAKUT disorder that is currently unavailable.
Collapse
Affiliation(s)
- Julie Klein
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Bénédicte Buffin-Meyer
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Franck Boizard
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Nabila Moussaoui
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Ophélie Lescat
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Benjamin Breuil
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Camille Fedou
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Guylène Feuillet
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Audrey Casemayou
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Eric Neau
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; Université Toulouse III Paul-Sabatier, Toulouse, France
| | - An Hindryckx
- Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven, Belgium
| | - Luc Decatte
- Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven, Belgium
| | - Elena Levtchenko
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Anke Raaijmakers
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Christophe Vayssière
- Université Toulouse III Paul-Sabatier, Toulouse, France; Department of Obstetrics and Gynecology, Paule de Viguier Hospital, CHU Toulouse, Toulouse, France; INSERM, UMR1027, Toulouse, France
| | - Valérie Goua
- Prenatal Diagnosis Unit, Poitiers University Hospital, Poitiers, France
| | | | - Franck Perrotin
- Department of Obstetrics, Gynecology and Fetal Medicine, University Hospital of Tours, Tours, France; INSERM, U1253, "Imaging and Brain," François-Rabelais University of Tours, Tours, France
| | - Sylvie Cloarec
- Reference Center for Rare Kidney Diseases, Pediatric Nephrology Service, CHRU Clocheville, Tours, France
| | - Alexandra Benachi
- Gynecology-Obstetric Service, AP-HP, Hôpital Antoine Béclère, Université Paris-Sud, Clamart, France
| | - Marie-Christine Manca-Pellissier
- Center for Prenatal Diagnosis, Timone Children's Hospital, Assistance Publique Hopitaux de Marseille, Aix-Marseille Université, Marseille, France
| | | | - Lucie Bessenay
- Pediatric Service, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | | | - Emma Allain-Launay
- Pediatric Nephrology Service, Hôpital Mère-Enfants, CHU Nantes, Nantes, France
| | - Jean Gondry
- Department of Obstetrics and Gynecology, University Hospital of Amiens, Amiens, France; INSERM, U1105, Picardie Jules Verne University, CHU Amiens, Amiens, France
| | | | - Elisabeth Simon
- Prenatal Diagnosis, Fondation Lenval, CHU de Nice, Nice, France
| | - Fabienne Prieur
- Clinical Genetics Service, CHU de Saint-Etienne, Saint-Etienne, France
| | - Marie-Pierre Lavocat
- Department of Pediatrics, Hôpital Nord, CHU de Saint Etienne, Saint Etienne, France
| | - Anne-Hélène Saliou
- Multidisciplinary Center for Prenatal Diagnosis, CHRU de Brest, Brest, France
| | - Loic De Parscau
- Department of Pediatrics and Medical Genetics, CHRU Morvan, Brest, France
| | - Laurent Bidat
- Gynecology-Obstetrics Service, Centre Hospitalier René Dubos, Pontoise, France
| | - Catherine Noel
- Gynecology-Obstetrics Service, Centre Hospitalier René Dubos, Pontoise, France
| | - Corinne Floch
- Pediatric Service, Hôpital Louis Mourier, Colombes, France
| | | | - Romain Favre
- Ultrasound and Foetal Medicine Service of the Department of Gynecology and Obstetrics, Hôpitaux Universitaires de Strasbourg, CMCO, Schiltigheim, France
| | - Anne-Sophie Weingertner
- Ultrasound and Foetal Medicine Service of the Department of Gynecology and Obstetrics, Hôpitaux Universitaires de Strasbourg, CMCO, Schiltigheim, France
| | - Jean-François Oury
- Gynecology-Obstetrics Service, Hôpital Universitaire Robert Debré, APHP, Paris, France
| | - Véronique Baudouin
- Pediatric Nephrology Service, Hôpital Universitaire Robert-Debré, APHP, Paris, France
| | - Jean-Paul Bory
- Service de Gynécologie-Obstétrique, Maternité Alix-de-Champagne, CHU de Reims, Reims, France
| | | | - Maryse Fiorenza
- Gynecology-Obstetrics Service, l'Hôpital Mère Enfant de Limoges, Limoges, France
| | - Jérôme Massardier
- Gynecology-Obstetrics Service, Hospices Civils de Lyon, Hôpital Femme Mère Enfant, Bron, France
| | | | - Nadia Lounis
- Pediatric Clinical Research Unit, Clinical Research Center Toulouse, Hôpital des Enfants, Toulouse, France
| | - Françoise Conte Auriol
- Pediatric Clinical Research Unit, Clinical Research Center Toulouse, Hôpital des Enfants, Toulouse, France
| | - Pascale Marcorelles
- Department of Pathology, EA 4685, Neuronal Epithelium Interaction Laboratory, Université de Bretagne Occidentale Brest, France
| | - Sophie Collardeau-Frachon
- Department of Pathology, Children and Mother's Hospital, Groupement Hospitalier Est, CHU de Lyon-Bron, France
| | - Petra Zürbig
- Mosaiques Diagnostics and Therapeutics, Hannover, Germany
| | - Harald Mischak
- Mosaiques Diagnostics and Therapeutics, Hannover, Germany; Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Pedro Magalhães
- Mosaiques Diagnostics and Therapeutics, Hannover, Germany; Department of Pediatric Nephrology, Hannover Medical School, Hannover, Germany
| | - Julie Batut
- Center for Developmental Biology (UMR5547) and Center for Integrative Biology (FR 3743), Université de Toulouse, CNRS, UPS, 31062, Toulouse, France
| | - Patrick Blader
- Center for Developmental Biology (UMR5547) and Center for Integrative Biology (FR 3743), Université de Toulouse, CNRS, UPS, 31062, Toulouse, France
| | - Jean-Sebastien Saulnier Blache
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; Université Toulouse III Paul-Sabatier, Toulouse, France
| | | | - Franz Schaefer
- Division of Pediatric Nephrology, Heidelberg University Center for Pediatrics and Adolescent Medicine, Heidelberg, Germany
| | - Stéphane Decramer
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; Université Toulouse III Paul-Sabatier, Toulouse, France; Pediatric Nephrology Service, Hôpital des Enfants, CHU Toulouse, Toulouse, France; Reference Center for Rare Renal Diseases of the Southwest (SORARE), Toulouse, France.
| | - Joost P Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; Université Toulouse III Paul-Sabatier, Toulouse, France.
| |
Collapse
|
74
|
Yang L, Chu TK, Lian J, Lo CW, Zhao S, He D, Qin J, Liang J. Individualised risk prediction model for new-onset, progression and regression of chronic kidney disease in a retrospective cohort of patients with type 2 diabetes under primary care in Hong Kong. BMJ Open 2020; 10:e035308. [PMID: 32641324 PMCID: PMC7348646 DOI: 10.1136/bmjopen-2019-035308] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES This study is aimed to develop and validate a prediction model for multistate transitions across different stages of chronic kidney disease (CKD) in patients with type 2 diabetes mellitus under primary care. SETTING We retrieved the anonymised electronic health records of a population-based retrospective cohort in Hong Kong. PARTICIPANTS A total of 26 197 patients were included in the analysis. PRIMARY AND SECONDARY OUTCOME MEASURES The new-onset, progression and regression of CKD were defined by the transitions of four stages that were classified by combining glomerular filtration rate and urine albumin-to-creatinine ratio. We applied a multiscale multistate Poisson regression model to estimate the rates of the stage transitions by integrating the baseline demographic characteristics, routine laboratory test results and clinical data from electronic health records. RESULTS During the mean follow-up time of 1.8 years, there were 2632 patients newly diagnosed with CKD, 1746 progressed to the next stage and 1971 regressed into an earlier stage. The models achieved the best performance in predicting the new-onset and progression with the predictors of sex, age, body mass index, systolic blood pressure, diastolic blood pressure, serum creatinine, haemoglobin A1c, total cholesterol, low-density lipoprotein, high-density lipoprotein, triglycerides and drug prescriptions. CONCLUSIONS This study demonstrated that individual risks of new-onset and progression of CKD can be predicted from the routine physical and laboratory test results. The individualised prediction curves developed from this study could potentially be applied to routine clinical practices, to facilitate clinical decision making, risk communications with patients and early interventions.
Collapse
Affiliation(s)
- Lin Yang
- School of Nursing, Hong Kong Polytechnic University, Hong Kong, Hong Kong
| | - Tsun Kit Chu
- Department of Family Medicine and Primary Healthcare, New Territory West Cluster, Hospital Authority, Hong Kong, Hong Kong
| | - Jinxiao Lian
- School of Optometry, Hong Kong Polytechnic University, Hong Kong, Hong Kong
| | - Cheuk Wai Lo
- Department of Family Medicine and Primary Healthcare, New Territory West Cluster, Hospital Authority, Hong Kong, Hong Kong
| | - Shi Zhao
- School of Nursing, Hong Kong Polytechnic University, Hong Kong, Hong Kong
- JC School of Public Health and Primary Care, Chinese University of Hong Kong, Hong Kong, China
- Shenzhen Research Institute of Chinese University of Hong Kong, Shenzhen, China
| | - Daihai He
- Department of Applied Mathematics, Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Jing Qin
- School of Nursing, Hong Kong Polytechnic University, Hong Kong, Hong Kong
| | - Jun Liang
- Department of Family Medicine and Primary Healthcare, New Territory West Cluster, Hospital Authority, Hong Kong, Hong Kong
| |
Collapse
|
75
|
Glazyrin YE, Veprintsev DV, Ler IA, Rossovskaya ML, Varygina SA, Glizer SL, Zamay TN, Petrova MM, Minic Z, Berezovski MV, Kichkailo AS. Proteomics-Based Machine Learning Approach as an Alternative to Conventional Biomarkers for Differential Diagnosis of Chronic Kidney Diseases. Int J Mol Sci 2020; 21:ijms21134802. [PMID: 32645927 PMCID: PMC7369970 DOI: 10.3390/ijms21134802] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 11/16/2022] Open
Abstract
Diabetic nephropathy, hypertension, and glomerulonephritis are the most common causes of chronic kidney diseases (CKD). Since CKD of various origins may not become apparent until kidney function is significantly impaired, a differential diagnosis and an appropriate treatment are needed at the very early stages. Conventional biomarkers may not have sufficient separation capabilities, while a full-proteomic approach may be used for these purposes. In the current study, several machine learning algorithms were examined for the differential diagnosis of CKD of three origins. The tested dataset was based on whole proteomic data obtained after the mass spectrometric analysis of plasma and urine samples of 34 CKD patients and the use of label-free quantification approach. The k-nearest-neighbors algorithm showed the possibility of separation of a healthy group from renal patients in general by proteomics data of plasma with high confidence (97.8%). This algorithm has also be proven to be the best of the three tested for distinguishing the groups of patients with diabetic nephropathy and glomerulonephritis according to proteomics data of plasma (96.3% of correct decisions). The group of hypertensive nephropathy could not be reliably separated according to plasma data, whereas analysis of entire proteomics data of urine did not allow differentiating the three diseases. Nevertheless, the group of hypertensive nephropathy was reliably separated from all other renal patients using the k-nearest-neighbors classifier “one against all” with 100% of accuracy by urine proteome data. The tested algorithms show good abilities to differentiate the various groups across proteomic data sets, which may help to avoid invasive intervention for the verification of the glomerulonephritis subtypes, as well as to differentiate hypertensive and diabetic nephropathy in the early stages based not on individual biomarkers, but on the whole proteomic composition of urine and blood.
Collapse
Affiliation(s)
- Yury E. Glazyrin
- Laboratory for Biomolecular and Medical Technologies, Krasnoyarsk State Medical University Named after Prof. V.F. Voyno-Yasenetsky, 660022 Krasnoyarsk, Russia; (T.N.Z.); (A.S.K.)
- Laboratory for Digital Controlled Drugs and Theranostics, Federal Research Center “Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Science”, 660036 Krasnoyarsk, Russia;
- Correspondence:
| | - Dmitry V. Veprintsev
- Laboratory for Digital Controlled Drugs and Theranostics, Federal Research Center “Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Science”, 660036 Krasnoyarsk, Russia;
| | - Irina A. Ler
- Department of Nephrology, Krasnoyarsk Interdistrict Clinical Hospital of Emergency Medical Care Named after N.S. Karpovich, 660062 Krasnoyarsk, Russia; (I.A.L.); (M.L.R.); (S.A.V.); (S.L.G.)
| | - Maria L. Rossovskaya
- Department of Nephrology, Krasnoyarsk Interdistrict Clinical Hospital of Emergency Medical Care Named after N.S. Karpovich, 660062 Krasnoyarsk, Russia; (I.A.L.); (M.L.R.); (S.A.V.); (S.L.G.)
| | - Svetlana A. Varygina
- Department of Nephrology, Krasnoyarsk Interdistrict Clinical Hospital of Emergency Medical Care Named after N.S. Karpovich, 660062 Krasnoyarsk, Russia; (I.A.L.); (M.L.R.); (S.A.V.); (S.L.G.)
| | - Sofia L. Glizer
- Department of Nephrology, Krasnoyarsk Interdistrict Clinical Hospital of Emergency Medical Care Named after N.S. Karpovich, 660062 Krasnoyarsk, Russia; (I.A.L.); (M.L.R.); (S.A.V.); (S.L.G.)
- Faculty of Medicine, Krasnoyarsk State Medical University Named after Prof. V.F. Voyno-Yasenetsky, 660022 Krasnoyarsk, Russia;
| | - Tatiana N. Zamay
- Laboratory for Biomolecular and Medical Technologies, Krasnoyarsk State Medical University Named after Prof. V.F. Voyno-Yasenetsky, 660022 Krasnoyarsk, Russia; (T.N.Z.); (A.S.K.)
| | - Marina M. Petrova
- Faculty of Medicine, Krasnoyarsk State Medical University Named after Prof. V.F. Voyno-Yasenetsky, 660022 Krasnoyarsk, Russia;
| | - Zoran Minic
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N6N5, Canada; (Z.M.); (M.V.B.)
| | - Maxim V. Berezovski
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N6N5, Canada; (Z.M.); (M.V.B.)
| | - Anna S. Kichkailo
- Laboratory for Biomolecular and Medical Technologies, Krasnoyarsk State Medical University Named after Prof. V.F. Voyno-Yasenetsky, 660022 Krasnoyarsk, Russia; (T.N.Z.); (A.S.K.)
- Laboratory for Digital Controlled Drugs and Theranostics, Federal Research Center “Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Science”, 660036 Krasnoyarsk, Russia;
| |
Collapse
|
76
|
Wang D, Yang J, Fan J, Chen W, Nikolic‐Paterson DJ, Li J. Omics technologies for kidney disease research. Anat Rec (Hoboken) 2020; 303:2729-2742. [PMID: 32592293 DOI: 10.1002/ar.24413] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/31/2019] [Accepted: 02/17/2020] [Indexed: 12/24/2022]
Affiliation(s)
- Dan Wang
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen University Guangzhou China
- Key Laboratory of Nephrology, National Health Commission and Guangdong Province Guangzhou China
| | - Jiayi Yang
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen University Guangzhou China
- Key Laboratory of Nephrology, National Health Commission and Guangdong Province Guangzhou China
| | - Jinjin Fan
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen University Guangzhou China
- Key Laboratory of Nephrology, National Health Commission and Guangdong Province Guangzhou China
| | - Wei Chen
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen University Guangzhou China
- Key Laboratory of Nephrology, National Health Commission and Guangdong Province Guangzhou China
| | | | - Jinhua Li
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen University Guangzhou China
- Key Laboratory of Nephrology, National Health Commission and Guangdong Province Guangzhou China
- Shunde Women and Children Hospital, Guangdong Medical University Shunde Guangdong China
- The Second Clinical College, Guangdong Medical University Dongguan Guangdong China
- Department of Anatomy and Developmental BiologyMonash Biomedicine Discovery Institute, Monash University Clayton Victoria Australia
| |
Collapse
|
77
|
Chen C, Yang L, Li H, Chen F, Chen C, Gao R, Lv XY, Tang J. Raman spectroscopy combined with multiple algorithms for analysis and rapid screening of chronic renal failure. Photodiagnosis Photodyn Ther 2020; 30:101792. [DOI: 10.1016/j.pdpdt.2020.101792] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 10/24/2022]
|
78
|
Omics research in diabetic kidney disease: new biomarker dimensions and new understandings? J Nephrol 2020; 33:931-948. [DOI: 10.1007/s40620-020-00759-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/23/2020] [Indexed: 12/14/2022]
|
79
|
Predictive Value of Precision-Cut Kidney Slices as an Ex Vivo Screening Platform for Therapeutics in Human Renal Fibrosis. Pharmaceutics 2020; 12:pharmaceutics12050459. [PMID: 32443499 PMCID: PMC7285118 DOI: 10.3390/pharmaceutics12050459] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/10/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
Animal models are a valuable tool in preclinical research. However, limited predictivity of human biological responses in the conventional models has stimulated the search for reliable preclinical tools that show translational robustness. Here, we used precision-cut kidney slices (PCKS) as a model of renal fibrosis and investigated its predictive capacity for screening the effects of anti-fibrotics. Murine and human PCKS were exposed to TGFβ or PDGF pathway inhibitors with established anti-fibrotic efficacy. For each treatment modality, we evaluated whether it affected: (1) culture-induced collagen type I gene expression and interstitial accumulation; (2) expression of markers of TGFβ and PDGF signaling; and (3) expression of inflammatory markers. We summarized the outcomes of published in vivo animal and human studies testing the three inhibitors in renal fibrosis, and drew a parallel to the PCKS data. We showed that the responses of murine PCKS to anti-fibrotics highly corresponded with the known in vivo responses observed in various animal models of renal fibrosis. Moreover, our results suggested that human PCKS can be used to predict drug efficacy in clinical trials. In conclusion, our study demonstrated that the PCKS model is a powerful predictive tool for ex vivo screening of putative drugs for renal fibrosis.
Collapse
|
80
|
Huang QF, Zhang ZY, Van Keer J, Trenson S, Nkuipou-Kenfack E, Yang WY, Thijs L, Vanhaecke J, Van Aelst LNL, Van Cleemput J, Janssens S, Verhamme P, Mischak H, Staessen JA. Urinary peptidomic biomarkers of renal function in heart transplant recipients. Nephrol Dial Transplant 2020; 34:1336-1343. [PMID: 29982668 PMCID: PMC6680096 DOI: 10.1093/ndt/gfy185] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 04/19/2018] [Indexed: 12/15/2022] Open
Abstract
Background Chronic kidney disease (CKD) is common in patients after heart transplantation (HTx). We assessed whether in HTx recipients the proteomic urinary classifier CKD273 or sequenced urinary peptides revealing the parental proteins correlated with the estimated glomerular filtration rate (eGFR). Methods In 368 HTx patients, we measured the urinary peptidome and analysed CKD273 and 48 urinary peptides with a detectable signal in >95% of participants. After 9.1 months (median), eGFR and the urinary biomarkers were reassessed. Results In multivariable Bonferroni-corrected analyses of the baseline data, a 1-SD increase in CKD273 was associated with a 11.4 [95% confidence interval (CI) 7.25–15.5] mL/min/1.73 m2 lower eGFR and an odds ratio of 2.63 (1.56–4.46) for having eGFR <60 mL/min/1.73 m2. While relating eGFR category at follow-up to baseline urinary biomarkers, CKD273 had higher (P = 0.007) area under the curve (0.75; 95% CI 0.70–0.80) than 24-h proteinuria (0.64; 95% CI 0.58–0.69), but additional adjustment for baseline eGFR removed significance of both biomarkers. In partial least squares analysis, the strongest correlates of the multivariable-adjusted baseline eGFR were fragments of collagen I (positive) and the mucin-1 subunit α (inverse). Associations between the changes in eGFR and the urinary markers were inverse for CKD273 and mucin-1 and positive for urinary collagen I. Conclusions With the exception of baseline eGFR, CKD273 was more closer associated with imminent renal dysfunction than 24-h proteinuria. Fragments of collagen I and mucin-1—respectively, positively and inversely associated with eGFR and change in eGFR—are single-peptide markers associated with renal dysfunction.
Collapse
Affiliation(s)
- Qi-Fang Huang
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium.,Center for Epidemiological Studies and Clinical Trials and Center for Vascular Evaluations, Shanghai Institute of Hypertension, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhen-Yu Zhang
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium.,Institut universitaire de médicine sociale et préventive, University of Lausanne, Lausanne, Switzerland
| | - Jan Van Keer
- Division of Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Sander Trenson
- Division of Cardiology, University Hospitals Leuven, Leuven, Belgium
| | | | - Wen-Yi Yang
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Lutgarde Thijs
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Johan Vanhaecke
- Division of Cardiology, University Hospitals Leuven, Leuven, Belgium
| | | | | | - Stefan Janssens
- Division of Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Peter Verhamme
- Centre for Molecular and Vascular Biology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Harald Mischak
- Mosaiques-Diagnostics AG, Hannover, Germany.,BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Jan A Staessen
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium.,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
81
|
Sun Q, Baues M, Klinkhammer BM, Ehling J, Djudjaj S, Drude NI, Daniel C, Amann K, Kramann R, Kim H, Saez-Rodriguez J, Weiskirchen R, Onthank DC, Botnar RM, Kiessling F, Floege J, Lammers T, Boor P. Elastin imaging enables noninvasive staging and treatment monitoring of kidney fibrosis. Sci Transl Med 2020; 11:11/486/eaat4865. [PMID: 30944168 DOI: 10.1126/scitranslmed.aat4865] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 11/28/2018] [Accepted: 03/11/2019] [Indexed: 12/13/2022]
Abstract
Fibrosis is the common endpoint and currently the best predictor of progression of chronic kidney diseases (CKDs). Despite several drawbacks, biopsies remain the only available means to specifically assess the extent of renal fibrosis. Here, we show that molecular imaging of the extracellular matrix protein elastin allows for noninvasive staging and longitudinal monitoring of renal fibrosis. Elastin was hardly expressed in healthy mouse, rat, and human kidneys, whereas it was highly up-regulated in cortical, medullar, and perivascular regions in progressive CKD. Compared to a clinically relevant control contrast agent, the elastin-specific magnetic resonance imaging agent ESMA specifically detected elastin expression in multiple mouse models of renal fibrosis and also in fibrotic human kidneys. Elastin imaging allowed for repetitive and reproducible assessment of renal fibrosis, and it enabled longitudinal monitoring of therapeutic interventions, accurately capturing anti-fibrotic therapy effects. Last, in a model of reversible renal injury, elastin imaging detected ensuing fibrosis not identifiable via routine assessment of kidney function. Elastin imaging thus has the potential to become a noninvasive, specific imaging method to assess renal fibrosis.
Collapse
Affiliation(s)
- Qinxue Sun
- Institute of Pathology, RWTH Aachen University Hospital, 52074 Aachen, Germany.,Department of Radiology, Ningbo Medical Center Li Huili Hospital, 315040 Ningbo, China
| | - Maike Baues
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Barbara M Klinkhammer
- Institute of Pathology, RWTH Aachen University Hospital, 52074 Aachen, Germany.,Department of Nephrology and Immunology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Josef Ehling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Sonja Djudjaj
- Institute of Pathology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Natascha I Drude
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany.,Department for Nuclear Medicine, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Christoph Daniel
- Institute of Pathology and Department of Nephropathology, University Erlangen, 91054 Erlangen, Germany
| | - Kerstin Amann
- Institute of Pathology and Department of Nephropathology, University Erlangen, 91054 Erlangen, Germany
| | - Rafael Kramann
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Hyojin Kim
- Joint Research Center for Computational Biomedicine, RWTH Aachen University Hospital, 52074 Aachen, Germany.,Institute of Computational Biomedicine, Heidelberg University, 69120 Heidelberg, Germany
| | - Julio Saez-Rodriguez
- Joint Research Center for Computational Biomedicine, RWTH Aachen University Hospital, 52074 Aachen, Germany.,Institute of Computational Biomedicine, Heidelberg University, 69120 Heidelberg, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | | | - Rene M Botnar
- School of Biomedical Engineering and Imaging Sciences, King's College London, WC2R 2LS London, UK
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Jürgen Floege
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany. .,Department of Targeted Therapeutics, University of Twente, 7522 NB Enschede, Netherlands
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, 52074 Aachen, Germany. .,Department of Nephrology and Immunology, RWTH Aachen University Hospital, 52074 Aachen, Germany.,Electron Microscopy Facility, RWTH Aachen University Hospital, 52074 Aachen, Germany.,Institute of Molecular Biomedicine, Comenius University, 81972 Bratislava, Slovakia
| |
Collapse
|
82
|
Wendt R, He T, Latosinska A, Siwy J, Mischak H, Beige J. Proteomic characterization of obesity-related nephropathy. Clin Kidney J 2020; 13:684-692. [PMID: 32905225 PMCID: PMC7467596 DOI: 10.1093/ckj/sfaa016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 01/27/2020] [Indexed: 12/13/2022] Open
Abstract
Background Nephropathy related to obesity lacks a pathophysiological understanding and definite diagnostic pathways by biomarkers. Methods In this study we investigated the association between urinary peptides and body mass index (BMI) and renal function in proteome data sets from 4015 individuals. Results A total of 365 urinary peptides were identified to be significantly associated with BMI. The majority of these peptides were collagen fragments. In addition, most of the peptides also demonstrated a significant concordant association with estimated glomerular filtration rate (eGFR) in the investigated cohort, with the presence of diabetes exhibiting no significant association. A new classifier was developed, based on 150 urinary peptides, that enabled the distinction of non-obese subjects with preserved kidney function from obese, non-diabetic subjects with eGFR >45 mL/min/1.73 m2 in an independent cohort, with an area under the curve of 0.93. Conclusions On a molecular level, the data strongly suggest a link between obesity and fibrosis, which may be a major cause of obesity-related nephropathy.
Collapse
Affiliation(s)
- Ralph Wendt
- Department of Nephrology and Kuratorium for Dialysis and Transplantation Renal Unit, Hospital St Georg, Leipzig, Germany
| | - Tianlin He
- Mosaiques Diagnostics, Hannover, Germany
| | | | | | | | - Joachim Beige
- Department of Nephrology and Kuratorium for Dialysis and Transplantation Renal Unit, Hospital St Georg, Leipzig, Germany.,Department of Nephrology, Martin-Luther-University Halle/Wittenberg, Halle, Germany
| |
Collapse
|
83
|
Tofte N, Lindhardt M, Adamova K, Bakker SJL, Beige J, Beulens JWJ, Birkenfeld AL, Currie G, Delles C, Dimos I, Francová L, Frimodt-Møller M, Girman P, Göke R, Havrdova T, Heerspink HJL, Kooy A, Laverman GD, Mischak H, Navis G, Nijpels G, Noutsou M, Ortiz A, Parvanova A, Persson F, Petrie JR, Ruggenenti PL, Rutters F, Rychlík I, Siwy J, Spasovski G, Speeckaert M, Trillini M, Zürbig P, von der Leyen H, Rossing P. Early detection of diabetic kidney disease by urinary proteomics and subsequent intervention with spironolactone to delay progression (PRIORITY): a prospective observational study and embedded randomised placebo-controlled trial. Lancet Diabetes Endocrinol 2020; 8:301-312. [PMID: 32135136 DOI: 10.1016/s2213-8587(20)30026-7] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/07/2020] [Accepted: 01/16/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Microalbuminuria is an early sign of kidney disease in people with diabetes and indicates increased risk of cardiovascular disease. We tested whether a urinary proteomic risk classifier (CKD273) score was associated with development of microalbuminuria and whether progression to microalbuminuria could be prevented with the mineralocorticoid receptor antagonist spironolactone. METHODS In this multicentre, prospective, observational study with embedded randomised controlled trial (PRIORITY), we recruited people with type 2 diabetes, normal urinary albumin excretion, and preserved renal function from 15 specialist centres in ten European countries. All participants (observational cohort) were tested with the CKD273 classifier and classified as high risk (CKD273 classifier score >0·154) or low risk (≤0·154). Participants who were classified as high risk were entered into a randomised controlled trial and randomly assigned (1:1), by use of an interactive web-response system, to receive spironolactone 25 mg once daily or matched placebo (trial cohort). The primary endpoint was development of confirmed microalbuminuria in all individuals with available data (observational cohort). Secondary endpoints included reduction in incidence of microalbuminuria with spironolactone (trial cohort, intention-to-treat population) and association between CKD273 risk score and measures of impaired renal function based on estimated glomerular filtration rate (eGFR; observational cohort). Adverse events (particularly gynaecomastia and hyperkalaemia) and serious adverse events were recorded for the intention-to-treat population (trial cohort). This study is registered with the EU Clinical Trials Register (EudraCT 20120-004523-4) and ClinicalTrials.gov (NCT02040441) and is completed. FINDINGS Between March 25, 2014, and Sept 30, 2018, we enrolled and followed-up 1775 participants (observational cohort), 1559 (88%) of 1775 participants had a low-risk urinary proteomic pattern and 216 (12%) had a high-risk pattern, of whom 209 were included in the trial cohort and assigned to spironolactone (n=102) or placebo (n=107). The overall median follow-up time was 2·51 years (IQR 2·0-3·0). Progression to microalbuminuria was seen in 61 (28%) of 216 high-risk participants and 139 (9%) of 1559 low-risk participants (hazard ratio [HR] 2·48, 95% CI 1·80-3·42; p<0·0001, after adjustment for baseline variables of age, sex, HbA1c, systolic blood pressure, retinopathy, urine albumin-to-creatinine ratio [UACR], and eGFR). Development of impaired renal function (eGFR <60 mL/min per 1·73 m2) was seen in 48 (26%) of 184 high-risk participants and 119 (8%) of 1423 low-risk participants (HR 3·50; 95% CI 2·50-4·90, after adjustment for baseline variables). A 30% decrease in eGFR from baseline (post-hoc endpoint) was seen in 42 (19%) of 216 high-risk participants and 62 (4%) of 1559 low-risk participants (HR 5·15, 95% CI 3·41-7·76; p<0·0001, after adjustment for basline eGFR and UACR). In the intention-to-treat trial cohort, development of microalbuminuria was seen in 35 (33%) of 107 in the placebo group and 26 (25%) of 102 in the spironolactone group (HR 0·81, 95% CI 0·49-1·34; p=0·41). In the safety analysis (intention-to-treat trial cohort), events of plasma potassium concentrations of more than 5·5 mmol/L were seen in 13 (13%) of 102 participants in the spironolactone group and four (4%) of 107 participants in the placebo group, and gynaecomastia was seen in three (3%) participants in the spironolactone group and none in the placebo group. One patient died in the placebo group due to a cardiac event (considered possibly related to study drug) and one patient died in the spironolactone group due to cancer, deemed unrelated to study drug. INTERPRETATION In people with type 2 diabetes and normoalbuminuria, a high-risk score from the urinary proteomic classifier CKD273 was associated with an increased risk of progression to microalbuminuria over a median of 2·5 years, independent of clinical characteristics. However, spironolactone did not prevent progression to microalbuminuria in high-risk patients. FUNDING European Union Seventh Framework Programme.
Collapse
Affiliation(s)
- Nete Tofte
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | | | - Katarina Adamova
- University Clinic of Endocrinology, Diabetes and Metabolic Disorders, Skopje, Macedonia
| | - Stephan J L Bakker
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Joachim Beige
- Division of Nephrology and KfH Renal Unit, Hospital St Georg, Leipzig, Germany; Martin-Luther University Halle, Wittenberg, Germany
| | - Joline W J Beulens
- Amsterdam Public Health Research Institute, VU University Medical Center, Amsterdam, Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Andreas L Birkenfeld
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich at Eberhard Karls University of Tübingen, Tübingen, Germany; German Center for Diabetes Research, Neuherberg, Germany
| | - Gemma Currie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | | | - Lidmila Francová
- 1st Department, Charles University, Third Faculty of Medicine, Prague, Czech Republic
| | | | - Peter Girman
- Diabetes Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Rüdiger Göke
- Diabetologische Schwerpunktpraxis, Diabetologen Hessen, Marburg, Germany
| | - Tereza Havrdova
- Diabetes Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adriaan Kooy
- Bethesda Diabetes Research Center, Hoogeveen, Netherlands; Diabetes Vascular Research Foundation (DVRF), Hoogeveen, Netherlands; University Medical Center Groningen, Groningen, Netherlands
| | - Gozewijn D Laverman
- Department of Internal Medicine/Nephrology, Ziekenhuisgroep Twente Hospital, Almelo, Netherlands
| | | | - Gerjan Navis
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Giel Nijpels
- Department General Practice and Elderly Care, Amsterdam, Netherlands
| | - Marina Noutsou
- Diabetes Center, 2nd Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Hippokratio General Hospital, Athens, Greece
| | - Alberto Ortiz
- Instituto de Investigacion Sanitaria de la Fundacion Jiménez Díaz UAM, Madrid, Spain
| | - Aneliya Parvanova
- Department of Renal Medicine, Clinical Research Centre for Rare Diseases "Aldo e CeleDaccò": Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Ranica, Bergamo, Italy
| | | | - John R Petrie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Piero L Ruggenenti
- Department of Renal Medicine, Clinical Research Centre for Rare Diseases "Aldo e CeleDaccò": Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Ranica, Bergamo, Italy
| | - Femke Rutters
- Amsterdam Public Health Research Institute, VU University Medical Center, Amsterdam, Netherlands
| | - Ivan Rychlík
- 1st Department, Charles University, Third Faculty of Medicine, Prague, Czech Republic; Faculty Hospital Královské Vinohrady, Prague, Czech Republic
| | | | - Goce Spasovski
- Department of Nephrology, Cyril and Methodius University in Skopje, Skopje, North Macedonia
| | | | - Matias Trillini
- Department of Renal Medicine, Clinical Research Centre for Rare Diseases "Aldo e CeleDaccò": Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Ranica, Bergamo, Italy
| | | | | | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark; University of Copenhagen, Copenhagen, Denmark.
| | | |
Collapse
|
84
|
Buffin-Meyer B, Tkaczyk M, Stańczyk M, Breuil B, Siwy J, Szaflik K, Talar T, Wojtera J, Krzeszowski W, Decramer S, Klein J, Schanstra JP. A single-center study to evaluate the efficacy of a fetal urine peptide signature predicting postnatal renal outcome in fetuses with posterior urethral valves. Pediatr Nephrol 2020; 35:469-475. [PMID: 31701236 DOI: 10.1007/s00467-019-04390-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/22/2019] [Accepted: 09/30/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Posterior urethral valves (PUVs) account for 17% of pediatric renal failure. The management of pregnancies involving fetuses with PUV is hampered by the fact that current clinical parameters obtained from fetal ultrasound and/or fetal urine biochemistry are insufficient to predict postnatal renal function. We previously have developed a fetal urine peptide signature (12PUV) that predicted with high precision postnatal renal failure at 2 years of age in fetuses with PUV. Here, we evaluated the accuracy of this signature to predict postnatal renal outcome in fetuses with PUV in an independent single-center study. METHODS Thirty-three women carrying fetuses with suspected PUV were included. Twenty-five fetuses received vesicoamniotic shunts during pregnancy. PUV was confirmed postnatally in 23 patients. Of those 23 fetuses, 2 were lost in follow-up. Four and 3 patients died in the pre- and perinatal periods, respectively. Follow-up renal function at 6 months of age was obtained for the remaining 14 patients. The primary outcome was early renal failure, defined by an eGFR < 60 mL/min/1.73 m2 before 6 months of age or pre- or perinatal death. RESULTS The peptide signature predicted postnatal renal outcome in postnatally confirmed PUV fetuses with an AUC of 0.94 (95%CI 0.74-1.0) and an accuracy of 90% (95%CI 78-100). The signature predicted postnatal renal outcome for the suspected PUV cases with an AUC of 0.89 (95%CI 0.72-0.97) and an accuracy of 84% (95%CI 71-97). CONCLUSIONS This single-center study confirms the predictive power of the previously identified 12PUV fetal urinary peptide signature.
Collapse
Affiliation(s)
- Bénédicte Buffin-Meyer
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Marcin Tkaczyk
- Department of Pediatrics, Immunology and Nephrology, Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Małgorzata Stańczyk
- Department of Pediatrics, Immunology and Nephrology, Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Benjamin Breuil
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | | | - Krzysztof Szaflik
- Department of Genecology, Fertility and Fetal Therapy, Polish Mother's Memorial Hospital - Research Institute, Lodz, Poland
| | - Tomasz Talar
- Department of Neonatology, Polish Mother's Memorial Hospital - Research Institute, Lodz, Poland
| | - Justyna Wojtera
- Department of Genecology, Fertility and Fetal Therapy, Polish Mother's Memorial Hospital - Research Institute, Lodz, Poland
| | - Waldemar Krzeszowski
- Department of Genecology, Fertility and Fetal Therapy, Polish Mother's Memorial Hospital - Research Institute, Lodz, Poland
| | - Stéphane Decramer
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
- Service de Néphrologie Pédiatrique, Hôpital des Enfants, CHU Toulouse, Toulouse, France
- Centre de Référence des Maladies Rénales Rares du Sud-Ouest (SORARE), Toulouse, France
| | - Julie Klein
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Joost P Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.
- Université Toulouse III Paul-Sabatier, Toulouse, France.
| |
Collapse
|
85
|
Epidemiology research to foster improvement in chronic kidney disease care. Kidney Int 2020; 97:477-486. [DOI: 10.1016/j.kint.2019.11.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/12/2019] [Accepted: 11/15/2019] [Indexed: 11/24/2022]
|
86
|
Urinary peptidomics and bioinformatics for the detection of diabetic kidney disease. Sci Rep 2020; 10:1242. [PMID: 31988353 PMCID: PMC6985249 DOI: 10.1038/s41598-020-58067-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 01/07/2020] [Indexed: 01/15/2023] Open
Abstract
The aim of this study was to establish a peptidomic profile based on LC-MS/MS and random forest (RF) algorithm to distinguish the urinary peptidomic scenario of type 2 diabetes mellitus (T2DM) patients with different stages of diabetic kidney disease (DKD). Urine from 60 T2DM patients was collected: 22 normal (stage A1), 18 moderately increased (stage A2) and 20 severely increased (stage A3) albuminuria. A total of 1080 naturally occurring peptides were detected, which resulted in the identification of a total of 100 proteins, irrespective of the patients’ renal status. The classification accuracy showed that the most severe DKD (A3) presented a distinct urinary peptidomic pattern. Estimates for peptide importance assessed during RF model training included multiple fragments of collagen and alpha-1 antitrypsin, previously associated to DKD. Proteasix tool predicted 48 proteases potentially involved in the generation of the 60 most important peptides identified in the urine of DM patients, including metallopeptidases, cathepsins, and calpains. Collectively, our study lightened some biomarkers possibly involved in the pathogenic mechanisms of DKD, suggesting that peptidomics is a valuable tool for identifying the molecular mechanisms underpinning the disease and thus novel therapeutic targets.
Collapse
|
87
|
Genovese F, Rasmussen DGK, Karsdal MA, Jesky M, Ferro C, Fenton A, Cockwell P. Imbalanced turnover of collagen type III is associated with disease progression and mortality in high-risk chronic kidney disease patients. Clin Kidney J 2020; 14:593-601. [PMID: 33623684 PMCID: PMC7886548 DOI: 10.1093/ckj/sfz174] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/05/2019] [Indexed: 12/16/2022] Open
Abstract
Background Tubulointerstitial fibrosis is a major pathological feature in chronic kidney disease (CKD) and collagen type III (COL3) is a major component of the renal fibrotic scar. We hypothesized that a dysregulated turnover of COL3 is an important determinant of CKD progression. We assessed the relationship between fragments reflecting active formation (PRO-C3) and degradation (C3M) of COL3 and CKD disease progression and mortality in a prospective cohort of CKD patients. Methods We measured PRO-C3 and C3M in urine (uPRO-C3 and uC3M) and serum (sPRO-C3 and sC3M) of 500 patients from the Renal Impairment in Secondary Care study. Disease progression was defined as a decline in estimated glomerular filtration rate >30% or the start of renal replacement therapy within 12 and 30 months. Results Levels of uC3M/creatinine decreased, whereas levels of uPRO-C3/creatinine and sPRO-C3 increased with increasing CKD stage. uC3M/creatinine was inversely and independently associated with disease progression by 12 months {odds ratio [OR] 0.39 [95% confidence interval (CI) 0.18-0.83]; P = 0.01 per doubling of uC3M/creatinine} with development of end-stage renal disease [hazard ratio (HR) 0.70 (95% CI 0.50-0.97); P = 0.03 per doubling of uC3M/creatinine]. sPRO-C3 at baseline was independently associated with increased mortality [HR 1.93 (95% CI 1.21-3.1); P = 0.006 per doubling of sPRO-C3] and disease progression by 30 months [OR 2.16 (95% CI 1.21-3.84); P = 0.009 per doubling of sPRO-C3]. Conclusions Dynamic products of COL3 formation and degradation were independently associated with CKD progression and mortality and may represent an opportunity to link pathological processes with targeted treatments against fibrosis.
Collapse
Affiliation(s)
| | | | | | - Mark Jesky
- Department of Nephrology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Charles Ferro
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,Department of Renal Medicine, Queen Elizabeth Hospital, Birmingham, UK
| | - Anthony Fenton
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,Department of Renal Medicine, Queen Elizabeth Hospital, Birmingham, UK
| | - Paul Cockwell
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,Department of Renal Medicine, Queen Elizabeth Hospital, Birmingham, UK
| |
Collapse
|
88
|
Sirolli V, Pieroni L, Di Liberato L, Urbani A, Bonomini M. Urinary Peptidomic Biomarkers in Kidney Diseases. Int J Mol Sci 2019; 21:E96. [PMID: 31877774 PMCID: PMC6982248 DOI: 10.3390/ijms21010096] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 12/20/2022] Open
Abstract
In order to effectively develop personalized medicine for kidney diseases we urgently need to develop highly accurate biomarkers for use in the clinic, since current biomarkers of kidney damage (changes in serum creatinine and/or urine albumin excretion) apply to a later stage of disease, lack accuracy, and are not connected with molecular pathophysiology. Analysis of urine peptide content (urinary peptidomics) has emerged as one of the most attractive areas in disease biomarker discovery. Urinary peptidome analysis allows the detection of short and long-term physiological or pathological changes occurring within the kidney. Urinary peptidomics has been applied extensively for several years now in renal patients, and may greatly improve kidney disease management by supporting earlier and more accurate detection, prognostic assessment, and prediction of response to treatment. It also promises better understanding of kidney disease pathophysiology, and has been proposed as a "liquid biopsy" to discriminate various types of renal disorders. Furthermore, proteins being the major drug targets, peptidome analysis may allow one to evaluate the effects of therapies at the protein signaling pathway level. We here review the most recent findings on urinary peptidomics in the setting of the most common kidney diseases.
Collapse
Affiliation(s)
- Vittorio Sirolli
- Nephrology and Dialysis Unit, Department of Medicine, G. d’Annunzio University, Chieti-Pescara, SS.Annunziata Hospital, Via dei Vestini, 66013 Chieti, Italy; (V.S.); (L.D.L.)
| | - Luisa Pieroni
- Proteomics and Metabonomics Unit, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy;
| | - Lorenzo Di Liberato
- Nephrology and Dialysis Unit, Department of Medicine, G. d’Annunzio University, Chieti-Pescara, SS.Annunziata Hospital, Via dei Vestini, 66013 Chieti, Italy; (V.S.); (L.D.L.)
| | - Andrea Urbani
- Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Department of Laboratory Diagnostic and Infectious Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Mario Bonomini
- Nephrology and Dialysis Unit, Department of Medicine, G. d’Annunzio University, Chieti-Pescara, SS.Annunziata Hospital, Via dei Vestini, 66013 Chieti, Italy; (V.S.); (L.D.L.)
| |
Collapse
|
89
|
Verbeke F, Siwy J, Van Biesen W, Mischak H, Pletinck A, Schepers E, Neirynck N, Magalhães P, Pejchinovski M, Pontillo C, Lichtinghagen R, Brand K, Vlahou A, De Bacquer D, Glorieux G. The urinary proteomics classifier chronic kidney disease 273 predicts cardiovascular outcome in patients with chronic kidney disease. Nephrol Dial Transplant 2019; 36:811-818. [DOI: 10.1093/ndt/gfz242] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Indexed: 12/11/2022] Open
Abstract
Abstract
Background
The urinary proteomic classifier chronic kidney disease 273 (CKD273) is predictive for the development and progression of chronic kidney disease (CKD) and/or albuminuria in type 2 diabetes. This study evaluates its role in the prediction of cardiovascular (CV) events in patients with CKD Stages G1–G5.
Methods
We applied the CKD273 classifier in a cohort of 451 patients with CKD Stages G1–G5 followed prospectively for a median of 5.5 years. Primary endpoints were all-cause mortality, CV mortality and the composite of non-fatal and fatal CV events (CVEs).
Results
In multivariate Cox regression models adjusting for age, sex, prevalent diabetes and CV history, the CKD273 classifier at baseline was significantly associated with total mortality and time to fatal or non-fatal CVE, but not CV mortality. Because of a significant interaction between CKD273 and CV history (P = 0.018) and CKD stages (P = 0.002), a stratified analysis was performed. In the fully adjusted models, CKD273 classifier was a strong and independent predictor of fatal or non-fatal CVE only in the subgroup of patients with CKD Stages G1–G3b and without a history of CV disease. In those patients, the highest tertile of CKD273 was associated with a >10-fold increased risk as compared with the lowest tertile.
Conclusions
The urinary CKD273 classifier provides additional independent information regarding the CV risk in patients with early CKD stage and a blank CV history. Determination of CKD273 scores on a random urine sample may improve the efficacy of intensified surveillance and preventive strategies by selecting patients who potentially will benefit most from early risk management.
Collapse
Affiliation(s)
- Francis Verbeke
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | | | - Wim Van Biesen
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | | | - Anneleen Pletinck
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Eva Schepers
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Nathalie Neirynck
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | | | | | | | - Ralf Lichtinghagen
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, Germany
| | - Korbinian Brand
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, Germany
| | - Antonia Vlahou
- Biotechnology Division, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Dirk De Bacquer
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| | - Griet Glorieux
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
90
|
Dubin RF, Rhee EP. Proteomics and Metabolomics in Kidney Disease, including Insights into Etiology, Treatment, and Prevention. Clin J Am Soc Nephrol 2019; 15:404-411. [PMID: 31636087 PMCID: PMC7057308 DOI: 10.2215/cjn.07420619] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In this review of the application of proteomics and metabolomics to kidney disease research, we review key concepts, highlight illustrative examples, and outline future directions. The proteome and metabolome reflect the influence of environmental exposures in addition to genetic coding. Circulating levels of proteins and metabolites are dynamic and modifiable, and thus amenable to therapeutic targeting. Design and analytic considerations in proteomics and metabolomics studies should be tailored to the investigator's goals. For the identification of clinical biomarkers, adjustment for all potential confounding variables, particularly GFR, and strict significance thresholds are warranted. However, this approach has the potential to obscure biologic signals and can be overly conservative given the high degree of intercorrelation within the proteome and metabolome. Mass spectrometry, often coupled to up-front chromatographic separation techniques, is a major workhorse in both proteomics and metabolomics. High-throughput antibody- and aptamer-based proteomic platforms have emerged as additional, powerful approaches to assay the proteome. As the breadth of coverage for these methodologies continues to expand, machine learning tools and pathway analyses can help select the molecules of greatest interest and categorize them in distinct biologic themes. Studies to date have already made a substantial effect, for example elucidating target antigens in membranous nephropathy, identifying a signature of urinary peptides that adds prognostic information to urinary albumin in CKD, implicating circulating inflammatory proteins as potential mediators of diabetic nephropathy, demonstrating the key role of the microbiome in the uremic milieu, and highlighting kidney bioenergetics as a modifiable factor in AKI. Additional studies are required to replicate and expand on these findings in independent cohorts. Further, more work is needed to understand the longitudinal trajectory of select protein and metabolite markers, perform transomics analyses within merged datasets, and incorporate more kidney tissue-based investigation.
Collapse
Affiliation(s)
- Ruth F Dubin
- Division of Nephrology, San Francisco Veterans Affairs Medical Center, University of California, San Francisco, California; and
| | - Eugene P Rhee
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
91
|
Tajima S, Yamamoto N, Masuda S. Clinical prospects of biomarkers for the early detection and/or prediction of organ injury associated with pharmacotherapy. Biochem Pharmacol 2019; 170:113664. [PMID: 31606409 DOI: 10.1016/j.bcp.2019.113664] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/08/2019] [Indexed: 12/31/2022]
Abstract
Several biomarkers are used to monitor organ damage caused by drug toxicity. Traditional markers of kidney function, such as serum creatinine and blood urea nitrogen are commonly used to estimate glomerular filtration rate. However, these markers have several limitations including poor specificity and sensitivity. A number of serum and urine biomarkers have recently been described to detect kidney damage caused by drugs such as cisplatin, gentamicin, vancomycin, and tacrolimus. Neutrophil gelatinase-associated lipocalin (NGAL), liver-type fatty acid-binding protein (L-FABP), kidney injury molecule-1 (KIM-1), monocyte chemotactic protein-1 (MCP-1), and cystatin C have been identified as biomarkers for early kidney damage. Hy's Law is widely used as to predict a high risk of severe drug-induced liver injury caused by drugs such as acetaminophen. Recent reports have indicated that glutamate dehydrogenase (GLDH), high-mobility group box 1 (HMGB-1), Keratin-18 (k18), MicroRNA-122 and ornithine carbamoyltransferase (OCT) are more sensitive markers of hepatotoxicity compared to the traditional markers including the blood levels of amiotransferases and total bilirubin. Additionally, the rapid development of proteomic technologies in biofluids and tissue provides a new multi-marker panel, leading to the discovery of more sensitive biomarkers. In this review, an update topics of biomarkers for the detection of kidney or liver injury associated with pharmacotherapy.
Collapse
Affiliation(s)
- Soichiro Tajima
- Department of Pharmacy, Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Nanae Yamamoto
- Department of Pharmacy, Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Satohiro Masuda
- Department of Pharmacy, Kyushu University Hospital, Fukuoka 812-8582, Japan; Department of Clinical Pharmacology and Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Department of Pharmacy, International University of Health and Welfare Narita Hospital, Japan; Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, International University of Health and Welfare Narita Hospital, Japan.
| |
Collapse
|
92
|
Siwy J, Mischak H, Zürbig P. Proteomics and personalized medicine: a focus on kidney disease. Expert Rev Proteomics 2019; 16:773-782. [DOI: 10.1080/14789450.2019.1659138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Justyna Siwy
- R & D, Mosaiques Diagnostics GmbH, Hannover, Germany
| | - Harald Mischak
- R & D, Mosaiques Diagnostics GmbH, Hannover, Germany
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Petra Zürbig
- R & D, Mosaiques Diagnostics GmbH, Hannover, Germany
| |
Collapse
|
93
|
Mwenda V, Githuku J, Gathecha G, Wambugu BM, Roka ZG, Ong'or WO. Prevalence and factors associated with chronic kidney disease among medical inpatients at the Kenyatta National Hospital, Kenya, 2018: a cross-sectional study. Pan Afr Med J 2019; 33:321. [PMID: 31692795 PMCID: PMC6815467 DOI: 10.11604/pamj.2019.33.321.18114] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 05/02/2019] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION The burden of chronic kidney disease (CKD) is increasing worldwide. Few studies in low and low-middle income countries have estimated the prevalence of CKD. We aimed to estimate prevalence and factors associated with CKD among medical inpatients at the largest referral hospital in Kenya. METHODS We conducted a cross-sectional study among medical inpatients at the Kenyatta National Hospital. We used systematic sampling and collected demographic information, behavioural risk factors, medical history, underlying conditions, laboratory and imaging workup using a structured questionnaire. We estimated glomerular filtration rate (GFR) in ml/min/1.73m2 classified into 5 stages; G1 (≥ 90), G2 (60-89), G3a (45-59), G3b (30-44), G4 (15-29) and G5 (<15, or treated by dialysis/renal transplant). Ethical approval was obtained from Kenyatta National Hospital-University of Nairobi Ethics and Research Committee (KNH-UoN ERC), approval number P510/09/2017. We estimated prevalence of CKD and used logistic regression to determine factors independently associated with CKD diagnosis. RESULTS We interviewed 306 inpatients; median age 40.0 years (IQR 24.0), 162 (52.9%) were male, 155 (50.7%) rural residents. CKD prevalence was 118 patients (38.6%, 95% CI 33.3-44.1); median age 42.5 years (IQR 28.0), 74 (62.7%) were male, 64 (54.2%) rural residents. Respondents with CKD were older than those without (difference 4.4 years, 95% CI 3.7-8.4 years, P = 0.032). Fifty-six (47.5%) of the patients had either stage G1 or G2, 17 (14.4%) had end-stage renal disease; 64 (54.2%) had haemoglobin below 10g/dl while 33 (28.0%) had sodium levels below 135 mmol/l. ). History of unexplained anaemia (aOR 1.80, 95% CI 1.02-3.19), proteinuria (aOR 5.16, 95% CI 2.09-12.74), hematuria (aOR 7.68, 95% CI 2.37-24.86); hypertension (aOR 2.71, 95% CI 1.53-4.80) and herbal medications use (aOR 1.97, 95% CI 1.07-3.64) were independently associated with CKD. CONCLUSION Burden of CKD was high among this inpatient population. Haematuria and proteinuria can aid CKD diagnosis. Public awareness on health hazards of herbal medication use is necessary.
Collapse
Affiliation(s)
- Valerian Mwenda
- Field Epidemiology and Laboratory Training Programme, Ministry of Health, Nairobi, Kenya
- Division of Non-communicable Diseases, Ministry of Health, Nairobi Kenya
| | - Jane Githuku
- Field Epidemiology and Laboratory Training Programme, Ministry of Health, Nairobi, Kenya
| | - Gladwell Gathecha
- Division of Non-communicable Diseases, Ministry of Health, Nairobi Kenya
| | | | - Zeinab Gura Roka
- Field Epidemiology and Laboratory Training Programme, Ministry of Health, Nairobi, Kenya
| | | |
Collapse
|
94
|
Zürbig P, Siwy J, Mischak H. Emerging urine-based proteomic biomarkers as valuable tools in the management of chronic kidney disease. Expert Rev Mol Diagn 2019; 19:853-856. [DOI: 10.1080/14737159.2019.1657406] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
| | | | - Harald Mischak
- Mosaiques Diagnostics GmbH, Hannover, Germany
- Institute of Cardiovascular and Medical Sciences University of Glasgow, Glasgow, UK
| |
Collapse
|
95
|
Lindhardt M, Persson F, Oxlund C, Jacobsen IA, Zürbig P, Mischak H, Rossing P, Heerspink HJL. Predicting albuminuria response to spironolactone treatment with urinary proteomics in patients with type 2 diabetes and hypertension. Nephrol Dial Transplant 2019; 33:296-303. [PMID: 28064163 DOI: 10.1093/ndt/gfw406] [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: 06/26/2016] [Accepted: 10/24/2016] [Indexed: 01/07/2023] Open
Abstract
Background The mineralocorticoid receptor antagonist spironolactone significantly reduces albuminuria in patients with diabetes. Prior studies have shown large between-patient variability in albuminuria treatment response. We previously developed and validated a urinary proteomic classifier that predicts onset and progression of chronic kidney disease. Here, we tested whether the proteomic classifier based on 273 urinary peptides (CKD273) predicts albuminuria response to spironolactone treatment. Methods We performed a post hoc analysis in a double-blind randomized clinical trial with allocation to either spironolactone 12.5-50 mg/day (n = 57) or placebo (n = 54) for 16 weeks. Patients were diagnosed with type 2 diabetes and resistant hypertension. Treatment was an adjunct to renin-angiotensin system inhibition. Primary endpoint was the percentage change in urine albumin to creatinine ratio (UACR). Capillary electrophoresis mass spectrometry was used to quantify urinary peptides at baseline. The previously validated combination of 273 known urinary peptides was used as proteomic classifier. Results Spironolactone reduced UACR relative to placebo by 50%, although with a large between-patient variability in UACR response (5th to 95th percentile, 7 to 312%). An interaction was detected between CKD273 and treatment assignment (β = -1.09, P = 0.026). Higher values of CKD273 at baseline were associated with a larger reduction in UACR in the spironolactone group (β = -0.70, P = 0.049), but not in the placebo group (β = 0.39, P = 0.25). Stratified in tertiles of baseline CKD273, reduction in UACR was greater in the highest tertile, 63% (95% confidence interval: 35-79%), as compared with the two other tertiles combined, 16% (-17 to 40%) (P = 0.011). Conclusions A urinary proteomics classifier can be used to identify individuals with type 2 diabetes who are more likely to show an albuminuria-lowering response to spironolactone treatment. These results suggest that urinary proteomics may be a valuable tool to tailor therapy, but confirmation in a larger clinical trial is required.
Collapse
Affiliation(s)
| | | | - Christina Oxlund
- University of Southern Denmark, Research Unit for Cardiovascular and renal protection, Odense, Denmark
| | - Ib A Jacobsen
- University of Southern Denmark, Research Unit for Cardiovascular and renal protection, Odense, Denmark
| | | | | | - Peter Rossing
- Faculty of Heath, University of Aarhus, Aarhus, Denmark.,The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
96
|
Shan D, Wang H, Khatri P, Niu Y, Song W, Zhao S, Jiang Y, Ma Q, Liu X, Zhang R, Wang W, Yin C. The Urinary Peptidome as a Noninvasive Biomarker Development Strategy for Prenatal Screening of Down's Syndrome. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 23:439-447. [PMID: 31381471 DOI: 10.1089/omi.2019.0098] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Prenatal screening for Down's syndrome based on maternal age, ultrasound measures, and maternal serum biomarkers is recommended worldwide, but the false-positive rate and poor diagnostic performance of these screening tests remain problematic. Genetic analysis of cell-free DNA in maternal blood has been developed as a new prenatal screening for Down's syndrome, but it has a number of limitations, including turnaround time and cost. Prenatal screening diagnostic innovation calls for new tests that are noninvasive, accurate, and affordable. We report original observations on potential peptide biomarkers in maternal urine for screening of fetal Down's syndrome. The peptidome of urine samples from 23 pregnant women carrying Down's syndrome fetuses and 30 pregnant women carrying fetuses with normal karyotype was fractionated by weak cation exchange magnetic beads and analyzed by MALDI-TOF mass spectrometry. Levels of six peptides (m/z 1022.1, 1032.1, 1099.5, 1155.9, 1306.6, and 2365.6) were significantly altered between the case and control groups after controlling for maternal and gestational age. A classification model was constructed based on these candidate peptides that could differentiate fetuses with Down's syndrome from controls with a sensitivity of 95.7%, a specificity of 70.0%, and an area under receiver operating characteristic curves of 0.909 (95% confidence interval, 0.835-0.984). Peptide peaks at m/z 1099.5 and 1155.9 were identified as the partial sequences of alpha-1-antitrypsin and heat shock protein beta-1, respectively. These new findings support the new idea that maternal urinary peptidome offers prospects for noninvasive biomarker discovery and development for the prenatal screening of fetal Down's syndrome.
Collapse
Affiliation(s)
- Dan Shan
- Department of Perinatal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Hao Wang
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China.,School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| | - Prekshya Khatri
- Department of Perinatal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Yue Niu
- Department of Perinatal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Wei Song
- Department of Perinatal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Shenglong Zhao
- Department of Perinatal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Yan Jiang
- Department of Perinatal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Qingwei Ma
- Bioyong Technology Co., Ltd., Beijing, China
| | - Xinchao Liu
- Bioyong Technology Co., Ltd., Beijing, China
| | - Rong Zhang
- Bioyong Technology Co., Ltd., Beijing, China
| | - Wei Wang
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China.,School of Medical and Health Sciences, Edith Cowan University, Perth, Australia.,School of Public Health, Shandong First Medical University, Taian, China
| | - Chenghong Yin
- Department of Perinatal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
97
|
Abstract
Proteome analysis has been applied in multiple studies in the context of chronic kidney disease, aiming at improving our knowledge on the molecular pathophysiology of the disease. The approach is generally based on the hypothesis that proteins are key in maintaining kidney function, and disease is a clinical consequence of a significant change of the protein level. Knowledge on critical proteins and their alteration in disease should in turn enable identification of ideal biomarkers that could guide patient management. In addition, all drugs currently employed target proteins. Hence, proteome analysis also promises to enable identifying the best suited therapeutic target, and, in combination with biomarkers, could be used as the rationale basis for personalized intervention. To assess the current status of proteome analysis in the context of CKD, we present the results of a systematic review, of up-to-date scientific research, and give an outlook on the developments that can be expected in near future. Based on the current literature, proteome analysis has already seen implementation in the management of CKD patients, and it is expected that this approach, also supported by the positive results generated to date, will see advanced high-throughput application.
Collapse
|
98
|
Scurt FG, Menne J, Brandt S, Bernhardt A, Mertens PR, Haller H, Chatzikyrkou C. Systemic Inflammation Precedes Microalbuminuria in Diabetes. Kidney Int Rep 2019; 4:1373-1386. [PMID: 31701047 PMCID: PMC6829192 DOI: 10.1016/j.ekir.2019.06.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/22/2019] [Accepted: 06/03/2019] [Indexed: 12/13/2022] Open
Abstract
Aim The aim of the case-control study was to investigate if serum biomarkers indicative of vascular inflammation and endothelial dysfunction can predict the development of microalbuminuria in patients with diabetes mellitus type 2. Methods Among participants enrolled in the ROADMAP (Randomized Olmesartan And Diabetes MicroAlbuminuria Prevention) and observational follow-up (OFU) studies, a panel of 15 serum biomarkers was quantified from samples obtained at initiation of the study and tested for associations with the development of new-onset microalbuminuria during follow-up. A case-control study was conducted with inclusion of 172 patients with microalbuminuria and 188 matched controls. Nonparametric inferential, nonlinear regression, mediation, and bootstrapping statistical methods were used for the analysis. Results The median follow-up time was 37 months. At baseline, mean concentrations of C-X-C motif chemokine ligand 16 (CXCL-16), transforming growth factor (TGF)–β1 and angiopoietin-2 were higher in patients with subsequent microalbuminuria. In the multivariate analysis, after adjustment for age, sex, body mass index, glycated hemoglobin, duration of diabetes, low-density lipoprotein (LDL), smoking status, blood pressure, baseline urine albumin-to-creatinine ratio (UACR), estimated glomerular filtration rate (eGFR), time of follow-up and cardiovascular disease, CXCL-16 (odds ratio [OR] 2.60, 95% confidence interval [CI] 1.71–3.96), angiopoietin-2 (OR 1.50, 95% CI 1.14–1.98) and TGF-β1 (OR 1.03, 95% CI 1.01–1.04) remained significant predictors of new-onset microalbuminuria (P < 0.001). Inclusion of these biomarkers in conventional clinical risk models for prediction of microalbuminuria increased the area under the curve (AUC) from 0.638 to 0.760 (P < 0.001). Conclusion In patients with type 2 diabetes, elevated plasma levels of CXCL-16, angiopoietin-2, and TGF-β1 are independently predictive of microalbuminuria. Thus, these serum markers improve renal risk models beyond established clinical risk factors.
Collapse
Affiliation(s)
- Florian G Scurt
- Clinic of Nephrology, Hypertension, Diabetes and Endocrinology, Health Campus Immunology, Infectiology, and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Jan Menne
- Nephrology Section, Hanover Medical School, Hanover, Germany
| | - Sabine Brandt
- Clinic of Nephrology, Hypertension, Diabetes and Endocrinology, Health Campus Immunology, Infectiology, and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Anja Bernhardt
- Clinic of Nephrology, Hypertension, Diabetes and Endocrinology, Health Campus Immunology, Infectiology, and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter R Mertens
- Clinic of Nephrology, Hypertension, Diabetes and Endocrinology, Health Campus Immunology, Infectiology, and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Hermann Haller
- Nephrology Section, Hanover Medical School, Hanover, Germany
| | - Christos Chatzikyrkou
- Clinic of Nephrology, Hypertension, Diabetes and Endocrinology, Health Campus Immunology, Infectiology, and Inflammation, Otto-von-Guericke University, Magdeburg, Germany.,Nephrology Section, Hanover Medical School, Hanover, Germany
| | | |
Collapse
|
99
|
Pelander L, Brunchault V, Buffin-Meyer B, Klein J, Breuil B, Zürbig P, Magalhães P, Mullen W, Elliott J, Syme H, Schanstra JP, Häggström J, Ljungvall I. Urinary peptidome analyses for the diagnosis of chronic kidney disease in dogs. Vet J 2019; 249:73-79. [PMID: 31239169 DOI: 10.1016/j.tvjl.2019.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 12/17/2022]
Abstract
Chronic kidney disease (CKD) is clinically important in canine medicine. Current diagnostic tools lack sensitivity for detection of subclinical CKD. The aim of the present study was to evaluate urinary peptidome analysis for diagnosis of CKD in dogs. Capillary electrophoresis coupled to mass spectrometry analysis demonstrated presence of approximately 5400 peptides in dog urine. Comparison of urinary peptide abundance of dogs with and without CKD led to the identification of 133 differentially excreted peptides (adjusted P for each peptide <0.05). Sequence information was obtained for 35 of these peptides. This 35 peptide subset and the total group of 133 peptides were used to construct two predictive models of CKD which were subsequently validated by researchers masked to results in an independent cohort of 20 dogs. Both models diagnosed CKD with an area under the receiver operating characteristic (ROC) curve of 0.88 (95% confidence intervals [CI], 0.72-1.0). Most differentially excreted peptides represented fragments of collagen I, indicating possible association with fibrotic processes in CKD (similar to the equivalent human urinary peptide CKD model, CKD273). This first study of the urinary peptidome in dogs identified peptides that were associated with presence of CKD. Future studies are needed to validate the utility of this model for diagnosis and prediction of progression of canine CKD in a clinical setting.
Collapse
Affiliation(s)
- L Pelander
- Department of Clinical Sciences, University of Agricultural Sciences, Ulls väg 12, 750 07 Uppsala, Sweden.
| | - V Brunchault
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Equipe 12, 1 Avenue Jean Poulhès, BP 84225, 31432 Toulouse Cedex 4, France; Université Toulouse III Paul-Sabatier Toulouse, France
| | - B Buffin-Meyer
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Equipe 12, 1 Avenue Jean Poulhès, BP 84225, 31432 Toulouse Cedex 4, France; Université Toulouse III Paul-Sabatier Toulouse, France
| | - J Klein
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Equipe 12, 1 Avenue Jean Poulhès, BP 84225, 31432 Toulouse Cedex 4, France; Université Toulouse III Paul-Sabatier Toulouse, France
| | - B Breuil
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Equipe 12, 1 Avenue Jean Poulhès, BP 84225, 31432 Toulouse Cedex 4, France; Université Toulouse III Paul-Sabatier Toulouse, France
| | - P Zürbig
- Department of Pediatric Nephrology, Hannover Medical School, Hannover, Germany; Mosaiques Diagnostics GmbH, Hannover, Germany
| | - P Magalhães
- Department of Pediatric Nephrology, Hannover Medical School, Hannover, Germany; Mosaiques Diagnostics GmbH, Hannover, Germany
| | - W Mullen
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - J Elliott
- Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - H Syme
- Clinical Science and Services, Royal Veterinary College, North Mymms, UK
| | - J P Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Equipe 12, 1 Avenue Jean Poulhès, BP 84225, 31432 Toulouse Cedex 4, France; Université Toulouse III Paul-Sabatier Toulouse, France
| | - J Häggström
- Department of Clinical Sciences, University of Agricultural Sciences, Ulls väg 12, 750 07 Uppsala, Sweden
| | - I Ljungvall
- Department of Clinical Sciences, University of Agricultural Sciences, Ulls väg 12, 750 07 Uppsala, Sweden
| |
Collapse
|
100
|
Bülow RD, Boor P. Extracellular Matrix in Kidney Fibrosis: More Than Just a Scaffold. J Histochem Cytochem 2019; 67:643-661. [PMID: 31116062 DOI: 10.1369/0022155419849388] [Citation(s) in RCA: 227] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Kidney fibrosis is the common histological end-point of progressive, chronic kidney diseases (CKDs) regardless of the underlying etiology. The hallmark of renal fibrosis, similar to all other organs, is pathological deposition of extracellular matrix (ECM). Renal ECM is a complex network of collagens, elastin, and several glycoproteins and proteoglycans forming basal membranes and interstitial space. Several ECM functions beyond providing a scaffold and organ stability are being increasingly recognized, for example, in inflammation. ECM composition is determined by the function of each of the histological compartments of the kidney, that is, glomeruli, tubulo-interstitium, and vessels. Renal ECM is a dynamic structure undergoing remodeling, particularly during fibrosis. From a clinical perspective, ECM proteins are directly involved in several rare renal diseases and indirectly in CKD progression during renal fibrosis. ECM proteins could serve as specific non-invasive biomarkers of fibrosis and scaffolds in regenerative medicine. The gold standard and currently only specific means to measure renal fibrosis is renal biopsy, but new diagnostic approaches are appearing. Here, we discuss the localization, function, and remodeling of major renal ECM components in healthy and diseased, fibrotic kidneys and the potential use of ECM in diagnostics of renal fibrosis and in tissue engineering.
Collapse
Affiliation(s)
- Roman David Bülow
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany.,Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|