1
|
Kaszyńska A, Kępska-Dzilińska M, Karakulska-Prystupiuk E, Tomaszewska A, Basak GW, Żórawski M, Jakubowska Z, Małyszko J. Markers of Kidney Injury: Proenkephalin A and Uromodulin, but Not Dickkopf-3, Are Elevated in Patients After Hematopoietic Stem Cell Transplantation. Int J Mol Sci 2025; 26:3581. [PMID: 40332103 PMCID: PMC12027477 DOI: 10.3390/ijms26083581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/23/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Kidney injury encompasses a broad spectrum of structural and functional abnormalities, directly associated with stem cell transplantation. Acute kidney injury and chronic kidney disease represent perilous complications of hematopoietic stem cell transplantation (HSCT), with an elevated risk of mortality and progression to end-stage renal disease. The early detection of these complications is, therefore, paramount, and research is increasingly focused on the identification of novel biomarkers of kidney damage. Recently, proenkephalin (PENK), a monomeric peptide that is freely filtered by the glomerulus and thus reflects glomerular filtration very well, has been shown to be an additional useful predictor of the occurrence of acute kidney injury and heart failure. Dickkopf-3 (DKK3) is a glycoprotein secreted by the renal tubular epithelium in response to stress and has been implicated in the development of interstitial fibrosis. It has therefore been evaluated primarily as a marker of fibrosis in chronic kidney disease (CKD), but may also help predict the development of acute kiney injury. Uromodulin is regarded as a renal marker. Previous studies have examined the potential of PENK, DKK-3 and uromodulin as a biomarker in individuals with preserved renal function. However, the urinary levels of PENK, DKK-3 and uromodulin in patients following HSCT have not yet been established. The objective of the present study was to assess urinary PENK, DKK-3, and uromodulin concentrations in patients who had been under ambulatory care of the Hematology, Transplantation and Internal Medicine Department for a minimum of three months following HSCT, and to investigate their correlations with kidney function, as reflected by serum creatinine and eGFR. The study population comprised 80 patients who had undergone allogeneic HSCT for various reasons, primarily hematological malignancies such as acute leukemias and lymphomas. In addition, 32 healthy volunteers were included in order to establish normal ranges for the biomarkers of interest. Urine concentrations of proenkephalin, DKK-3, and uromodulin were evaluated using a commercially available sandwich ELISA immunoassay. Demographic and clinical data were retrieved from the patients' records. Statistical analyses were conducted using XLSLAT 2022 (Lumivero, Denver, CO, USA) and STATISTICAv13.0 (StatSoft, Tulsa, OH, USA). The results showed that PENK and DKK-3 levels were significantly higher in patients after HSCT compared to healthy volunteers. Furthermore, when patients were divided according to kidney function (below and over 60 mL/min/1.72 m2), it was found that the concentration of PENK and DKK-3 were significantly higher in 23 patients with CKD stage 3 relative to patients with eGFR over 60 mL min 1.72 m2. In univariate correlations, PENK demonstrated an inverse relationship with eGFR (r: -0.21, p < 0.05), while DKK-3 exhibited no significant correlation with creatinine or eGFR.Patients following allogeneic HSCT, despite having normal or near-normal kidney function, exhibited evidence of kidney injury. However, further research is necessary to ascertain the clinical utility of the novel biomarker.
Collapse
Affiliation(s)
- Aleksandra Kaszyńska
- Department of Nephrology, Dialysis and Internal Medicine, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; (A.K.); (M.K.-D.); (Z.J.)
| | - Małgorzata Kępska-Dzilińska
- Department of Nephrology, Dialysis and Internal Medicine, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; (A.K.); (M.K.-D.); (Z.J.)
| | - Ewa Karakulska-Prystupiuk
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; (E.K.-P.); (A.T.); (G.W.B.)
| | - Agnieszka Tomaszewska
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; (E.K.-P.); (A.T.); (G.W.B.)
| | - Grzegorz Władysław Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; (E.K.-P.); (A.T.); (G.W.B.)
| | - Marcin Żórawski
- Department of Clinical Medicine, Medical University, Szpitalna 37, 15-254 Bialystok, Poland;
| | - Zuzanna Jakubowska
- Department of Nephrology, Dialysis and Internal Medicine, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; (A.K.); (M.K.-D.); (Z.J.)
| | - Jolanta Małyszko
- Department of Nephrology, Dialysis and Internal Medicine, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; (A.K.); (M.K.-D.); (Z.J.)
| |
Collapse
|
2
|
Sun Y, Xiao Z, Zhao H, An Y. Serum Dickkopf-3 as a biomarker for predicting acute kidney injury in postoperative intensive care patients. Minerva Anestesiol 2025; 91:306-313. [PMID: 39912573 DOI: 10.23736/s0375-9393.24.18677-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
BACKGROUND Acute kidney injury (AKI) is a common and significant complication in the Intensive Care Unit (ICU), affecting more than half of all patients admitted. This condition is associated with increased morbidity and mortality, underscoring the urgent need for accurate and specific biomarkers to enable early diagnosis and intervention. Dickkopf-3 (DKK3) has emerged as a promising candidate biomarker for renal injury. METHODS We conducted a single-center, prospective cohort study from March 1 to July 1, 2023, enrolling 166 non-cardiac postoperative patients admitted to the ICU. Serum and urinary DKK3 levels were quantified using enzyme-linked immunosorbent assay (ELISA) kits. A multifactorial logistic regression model was constructed, incorporating changes in serum creatinine (ΔScr), cystatin C (CysC), serum DKK3 levels, and the serum DKK3 to urine DKK3 ratio. RESULTS Elevated serum DKK3 levels were significantly associated with an increased incidence of AKI and a composite outcome of adverse events (AKI or death). The multifactorial logistic regression model exhibited excellent performance, with an area under the receiver operating characteristic curve (AUC) of 0.98. Decision curve analysis (DCA) demonstrated a net clinical benefit of utilizing serum DKK3 levels to guide treatment decisions, particularly at higher risk thresholds. CONCLUSIONS Serum DKK3 is a robust diagnostic biomarker for AKI, effectively stratifying patients based on protein levels. The predictive model that incorporates DKK3 provides a valuable tool for clinical decision-making in the ICU setting. Further validation in larger and more diverse populations is warranted.
Collapse
Affiliation(s)
- Yao Sun
- Department of Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Zengli Xiao
- Department of Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Huiying Zhao
- Department of Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Youzhong An
- Department of Critical Care Medicine, Peking University People's Hospital, Beijing, China -
| |
Collapse
|
3
|
Sun Y, Xiao Z, Zhao H, An Y. Urinary dickkopf-3 as a predictor for postoperative acute kidney injury in the intensive care unit. Am J Med Sci 2025; 369:434-442. [PMID: 39561890 DOI: 10.1016/j.amjms.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND As a life-threatening complication in patients undergoing surgery, acute kidney injury (AKI) is strongly associated with a worse prognosis. Urinary dickkopf-related protein 3 (DKK3) has been identified as a biomarker for predicting postoperative AKI in patients undergoing cardiac surgery. OBJECTIVE To investigate the predictive value of urinary DKK3 on postoperative AKI and develop a clinical model based on the predictor for predicting the development of AKI within seven days for patients undergoing noncardiac surgery. METHODS All patients who were admitted to the Intensive Care Unit (ICU) after noncardiac surgery from March 2023 to June 2023 were included in this study. The patients' baseline data on demographic characteristics, lifestyle risk factors, medical history, clinical features, and laboratory tests before surgery were collected at the time of admission. Besides, the blood samples for cystatin C and routine laboratory tests and the urine samples for DKK3 tests were simultaneously collected at the time of admission to the ICU. In addition, the independent predictors of postoperative AKI were identified by univariate, multivariate, and LASSO analyses. Moreover, a nomogram for predicting postoperative AKI was developed based on these independent predictors. Finally, the nomogram was evaluated through calibration and decision curve analyses. RESULTS A total of 166 patients with a median age of 67 years old were included in this study, including 94 (56.63 %) males. Among these patients, 47 patients (28.3 %) developed postoperative AKI. Additionally, 7 independent risk factors, including preoperative serum creatinine, preoperative eGFR, preoperative serum albumin, preoperative serum potassium ion, cystatin C, uDKK3/uCr, and SOFA score, were selected by univariate and multivariate regression analyses. Eventually, 4 independent risk factors (including preoperative eGFR, cystatin C, uDKK3/uCr, and SOFA score) identified in this study by LASSO analyses were used to establish the nomogram. The area under the receiver operating characteristic (ROC) curve (AUC) for the prediction model was 0.868. The calibration curve and decision curve analysis results demonstrated that the nomogram had good prediction performance. CONCLUSIONS Urinary DKK3/creatinine was independently associated with postoperative AKI for patients in the ICU after noncardiac surgery. The nomogram constructed based on uDKK3/uCr, preoperative eGFR, cystatin C, and SOFA score showed a higher accuracy in predicting postoperative AKI.
Collapse
Affiliation(s)
- Yao Sun
- Department of Critical Care Medicine, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing 100044, PR China
| | - Zengli Xiao
- Department of Critical Care Medicine, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing 100044, PR China
| | - Huiying Zhao
- Department of Critical Care Medicine, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing 100044, PR China.
| | - Youzhong An
- Department of Critical Care Medicine, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing 100044, PR China.
| |
Collapse
|
4
|
González Rodríguez J, Valdivielso JM, Jatem Escalante E, Borràs Sans M, García Carrasco A, Del Carpio Salas J, Muijsenberg Alcalá A, Pinyol Ribas M, Ostos Roldán E, Segarra Medrano A, Martín Conde ML. Development of a multiple urinary biomarker model to predict the tubulointerstitial fibrosis area in patients with primary IgA Nephropathy. BMC Nephrol 2025; 26:141. [PMID: 40114119 PMCID: PMC11927370 DOI: 10.1186/s12882-025-04049-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/26/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Previous studies highlighted the utility of individual urinary biomarkers in the prediction of interstitial fibrosis in IgA Nephropathy patients. However, it´s uncertain which biomarker or combination of biomarkers provides a more accurate estimation of renal interstitial fibrosis Surface. Herein, we measured the urinary excretion of a set of seven tubular injury biomarkers in a group of patients with primary IgA Nephropathy and analyzed their utility as non-invasive estimators of interstitial fibrosis area found on kidney biopsy. METHODS Two hundred forty-seven adults with primary IgA Nephropathy diagnosed by kidney biopsy and a control group of 50 healthy control were included. The urinary excretion of EGF, MCP-1, NGAL, KIM-1, L-FABP, β2-microglobulin and DKK-3 was measured in urine samples collected at the day of the renal biopsy. Estimated glomerular filtration rate was measured by the CKD-EPI formula. Interstitial fibrosis area was quantified using a quantitative morphometric procedure and graded according to Oxford Classification. Predictive multivariate models were developed to predict the interstitial fibrosis surface. RESULTS Patients with primary IgA Nephropathy showed significantly higher urinary levels of DKK-3, L-FABP and β2-microglobulin, and lower EGF levels than healthy controls. Interstitial fibrosis was negatively correlated with urinary EGF levels and positively with age, proteinuria, eGFR and urinary DKK-3, L-FABP and β2-microglobulin. The best model to predict interstitial fibrosis area accounted for > 60% of its variability and included age, eGFR, proteinuria, DKK-3, EGF, L-FABP and β2-microglobulin. CONCLUSIONS Our study provides a model to estimate the IFS in IgA Nephropathy which could be useful to monitor the progression of chronic kidney injury.
Collapse
Affiliation(s)
- Jorge González Rodríguez
- Nephrology Department, Arnau de Vilanova University Hospital, Avda. Alcalde Rovira Roure, 80, 25198, Lleida, Spain.
- Biomedical Research Institute August Pi I Sunyer, Lleida, Spain.
- University of Lleida - Universitat de Lleida (UdL), Faculty of Medicine, Nephrology, Lleida, Spain.
| | - Jose Manuel Valdivielso
- Nephrology Department, Arnau de Vilanova University Hospital, Avda. Alcalde Rovira Roure, 80, 25198, Lleida, Spain
- Biomedical Research Institute August Pi I Sunyer, Lleida, Spain
- University of Lleida - Universitat de Lleida (UdL), Faculty of Medicine, Nephrology, Lleida, Spain
| | - Elías Jatem Escalante
- Nephrology Department, Arnau de Vilanova University Hospital, Avda. Alcalde Rovira Roure, 80, 25198, Lleida, Spain
- Biomedical Research Institute August Pi I Sunyer, Lleida, Spain
- University of Lleida - Universitat de Lleida (UdL), Faculty of Medicine, Nephrology, Lleida, Spain
| | - Mercè Borràs Sans
- Nephrology Department, Arnau de Vilanova University Hospital, Avda. Alcalde Rovira Roure, 80, 25198, Lleida, Spain
- Biomedical Research Institute August Pi I Sunyer, Lleida, Spain
- University of Lleida - Universitat de Lleida (UdL), Faculty of Medicine, Nephrology, Lleida, Spain
| | - Alicia García Carrasco
- Biomedical Research Institute August Pi I Sunyer, Lleida, Spain
- University of Lleida - Universitat de Lleida (UdL), Faculty of Medicine, Nephrology, Lleida, Spain
| | - Jacqueline Del Carpio Salas
- Nephrology Department, Arnau de Vilanova University Hospital, Avda. Alcalde Rovira Roure, 80, 25198, Lleida, Spain
- University of Lleida - Universitat de Lleida (UdL), Faculty of Medicine, Nephrology, Lleida, Spain
| | - Andrea Muijsenberg Alcalá
- Nephrology Department, Arnau de Vilanova University Hospital, Avda. Alcalde Rovira Roure, 80, 25198, Lleida, Spain
- Biomedical Research Institute August Pi I Sunyer, Lleida, Spain
- University of Lleida - Universitat de Lleida (UdL), Faculty of Medicine, Nephrology, Lleida, Spain
| | - Miquel Pinyol Ribas
- Pathology Department, Arnau de Vilanova University Hospital, Lleida, Spain
- University of Lleida - Universitat de Lleida (UdL), Faculty of Medicine, Nephrology, Lleida, Spain
| | - Elena Ostos Roldán
- Vall d'Hebrón Research Institute, Barcelona, Spain
- University of Lleida - Universitat de Lleida (UdL), Faculty of Medicine, Nephrology, Lleida, Spain
| | - Alfons Segarra Medrano
- Nephrology Department, Arnau de Vilanova University Hospital, Avda. Alcalde Rovira Roure, 80, 25198, Lleida, Spain
- Vall d'Hebrón Research Institute, Barcelona, Spain
- Biomedical Research Institute August Pi I Sunyer, Lleida, Spain
- Nephrology Department, University Hospital Vall d'Hebrón, Barcelona, Spain
- University of Lleida - Universitat de Lleida (UdL), Faculty of Medicine, Nephrology, Lleida, Spain
| | - Maria Luisa Martín Conde
- Nephrology Department, Arnau de Vilanova University Hospital, Avda. Alcalde Rovira Roure, 80, 25198, Lleida, Spain
- Biomedical Research Institute August Pi I Sunyer, Lleida, Spain
- University of Lleida - Universitat de Lleida (UdL), Faculty of Medicine, Nephrology, Lleida, Spain
| |
Collapse
|
5
|
Fallois JD, Günzel A, Daniel C, Stumpf J, Busch M, Pein U, Paliege A, Amann K, Wiech T, Hantmann E, Wolf G, Pfeifer F, Girndt M, Lindner TH, Weimann A, Seehofer D, Bachmann A, Budde K, Biemann R, Isermann B, Engel C, Dittrich K, Hugo C, Halbritter J. Deceased donor urinary Dickkopf-3 associates with future allograft function following kidney transplantation. Am J Transplant 2025; 25:516-530. [PMID: 39303796 DOI: 10.1016/j.ajt.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
Predicting future kidney allograft function is challenging. Novel biomarkers, such as urinary Dickkopf-3 (uDKK3), may help guide donor selection and improve allograft outcomes. In this prospective multicenter pilot trial, we investigated whether donor uDKK3 reflects organ quality and is associated with future allograft function. We measured uDKK3/crea ratios (uDKK3/crea) from 95 deceased and 46 living kidney donors. Prenephrectomy uDKK3/crea levels were 100× higher in deceased than in living donors (9888 pg/mg vs 113 pg/mg; P < .001). Among deceased donor transplantations, recipients were stratified by their corresponding uDKK3/crea donor levels ranging below (group A, n = 68) or above (group B, n = 65) median. The primary end point of best estimated glomerular filtration rate (eGFR) within the first 3 months after kidney transplantation was superior in group A (56.3 mL/min/1.73 m2) than that in group B (44.2 mL/min/1.73 m2; P = .0139). Second, the composite clinical end point consisting of death, allograft failure or eGFR decline >50% occurred less frequent in group A. By mixed linear regression modeling, donor uDKK3/crea remained an independent predictor of eGFR after transplantation, with a slope of -4.282 mL/min/1.73 m2 per logarithmic increase in donor uDKK3/crea. In summary, uDKK3 may serve as a noninvasive, donor-dependent biomarker for assessing organ quality and future allograft function.
Collapse
Affiliation(s)
- Jonathan de Fallois
- Division of Nephrology, Department of Internal Medicine, University Medical Center Leipzig, Leipzig, Germany.
| | - Anna Günzel
- Division of Nephrology, Department of Internal Medicine, University Medical Center Leipzig, Leipzig, Germany
| | - Christoph Daniel
- Department of Nephropathology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Julian Stumpf
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Martin Busch
- Department of Internal Medicine III, University Hospital Jena, Jena, Germany
| | - Ulrich Pein
- Department of Internal Medicine II, University Hospital Halle (Saale), Halle (Saale), Germany
| | - Alexander Paliege
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Kerstin Amann
- Department of Nephropathology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Thorsten Wiech
- Nephropathology Section, Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elena Hantmann
- Division of Nephrology, Department of Internal Medicine, University Medical Center Leipzig, Leipzig, Germany; Department of Nephrology and Medical Intensive Care, Charité Berlin, Berlin, Germany
| | - Gunter Wolf
- Department of Internal Medicine III, University Hospital Jena, Jena, Germany
| | - Felix Pfeifer
- German Organ Procurement Organization (DSO), Region East, Leipzig, Germany
| | - Matthias Girndt
- Department of Internal Medicine II, University Hospital Halle (Saale), Halle (Saale), Germany
| | - Tom H Lindner
- Division of Nephrology, Department of Internal Medicine, University Medical Center Leipzig, Leipzig, Germany
| | - Antje Weimann
- Division of Visceral Surgery and Transplantation Medicine, University Medical Center Leipzig, Leipzig, Germany
| | - Daniel Seehofer
- Division of Visceral Surgery and Transplantation Medicine, University Medical Center Leipzig, Leipzig, Germany
| | - Anette Bachmann
- Division of Nephrology, Department of Internal Medicine, University Medical Center Leipzig, Leipzig, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité Berlin, Berlin, Germany
| | - Ronald Biemann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Christoph Engel
- Institute for Medical Informatics, Statistics, and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Katalin Dittrich
- German Organ Procurement Organization (DSO), Region East, Leipzig, Germany; Division of Pediatric Nephrology and Transplantation, Department of Pediatrics, University Medical Center Leipzig, Leipzig, Germany
| | - Christian Hugo
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Jan Halbritter
- Division of Nephrology, Department of Internal Medicine, University Medical Center Leipzig, Leipzig, Germany; Department of Nephrology and Medical Intensive Care, Charité Berlin, Berlin, Germany.
| |
Collapse
|
6
|
Sun Y, Xiao Z, Yang S, Hao C, Zhao H, An Y. Advances and insights for DKK3 in non-cancerous diseases: a systematic review. PeerJ 2025; 13:e18935. [PMID: 39959827 PMCID: PMC11830365 DOI: 10.7717/peerj.18935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/14/2025] [Indexed: 02/18/2025] Open
Abstract
This review delves into the role of Dickkopf-3 (DKK3), a secreted glycoprotein and member of the Dickkopf family, in non-malignant diseases. DKK3 is particularly known for its regulatory effects on the Wnt signaling pathway, a critical mediator in various biological processes including cell proliferation, differentiation, and migration. Our review highlights DKK3's influence in disorders of the cardiovascular, respiratory, renal, and muscular systems, where it contributes to disease progression by modulating these key biological processes. As an emerging biomarker, DKK3's levels have been found to correlate with various disease states, underscoring its potential diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Yao Sun
- Intensive Care Unit, Peking University People’s Hospital, Beijing, China
| | - Zengli Xiao
- Intensive Care Unit, Peking University People’s Hospital, Beijing, China
| | - Shuguang Yang
- Intensive Care Unit, Peking University People’s Hospital, Beijing, China
| | - Chenxiao Hao
- Intensive Care Unit, Peking University People’s Hospital, Beijing, China
| | - Huiying Zhao
- Intensive Care Unit, Peking University People’s Hospital, Beijing, China
| | - Youzhong An
- Intensive Care Unit, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
7
|
Boeckhaus J, Tönshoff B, Weber LT, Pape L, Latta K, Fehrenbach H, Lange-Sperandio B, Kettwig M, König S, John-Kroegel U, Gellermann J, Galiano M, Jami S, Pieper D, Dihazi GH, Hafke A, Kohl S, Liebau MC, König J, Haffner D, Gross O, Wallbach M. Urinary Dickkopf-related protein 3 as a novel biomarker for kidney function decline in children with Alport syndrome. Pediatr Nephrol 2025:10.1007/s00467-025-06696-3. [PMID: 39904897 DOI: 10.1007/s00467-025-06696-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Chronic kidney disease (CKD) seriously affects the well-being and shortens the life expectancy of children and adolescents, but its progression is challenging to predict. Therefore, there is an urgent need for biomarkers that can identify children at risk of faster CKD progression. Alport syndrome (AS) is the most common monogenetic glomerular kidney disease. Urinary Dickkopf-related protein 3 (DKK3) is associated with a decline in estimated glomerular filtration rate (eGFR) in adults and children with advanced CKD. However, its potential for early detection of CKD and its prognostic value in children with AS remain unknown. METHODS Urine samples from 49 children enrolled in the EARLY PRO-TECT Alport trial were analyzed to evaluate whether DKK3 could identify children with AS to be at risk for faster CKD progression. RESULTS DKK3 levels in patients with AS were higher than those of healthy individuals reported in the literature. DKK3 levels were more elevated in patients with later stages of AS. Furthermore, children who were not treated with renin angiotensin system inhibitors (RASi) had higher DKK3 levels than treated children. Children with above-average DKK3 levels were more likely to have increased albuminuria after 2 years of follow-up than children with below-average DKK3 levels. CONCLUSION Urinary DKK3 is significantly elevated in children at early stages of AS. There was a potential association between higher DKK3 levels, worsening albuminuria, and a decline in kidney function. These findings suggest that DKK3 may be a prognostic marker for predicting the risk of kidney damage in children with AS.
Collapse
Affiliation(s)
- Jan Boeckhaus
- Clinic for Nephrology and Rheumatology, University Medical Center Göttingen, Goettingen, Germany.
| | - Burkhard Tönshoff
- Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Lutz T Weber
- Pediatric Nephrology, Children's and Adolescents' Hospital, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Lars Pape
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
- Department of Pediatrics II, University Children's Hospital, University of Duisburg-Essen, Essen, Germany
| | - Kay Latta
- Clementine Kinderhospital Frankfurt, Frankfurt, Germany
| | | | | | - Matthias Kettwig
- Department of Pediatrics and Adolescent Medicine, University Medical Center Göttingen, Göttingen, Germany
| | - Sabine König
- University Children's Hospital Münster, Münster, Germany
| | - Ulrike John-Kroegel
- Division of Pediatric Nephrology, University Children's Hospital, Jena, Germany
| | - Jutta Gellermann
- Pediatric Nephrology, Charité Children's Hospital, Berlin, Germany
| | - Matthias Galiano
- Department of Pediatrics and Adolescent Medicine, University Hospital, Friedrich-Alexander-University Erlangen, Erlangen, Germany
| | - Sima Jami
- Clinic for Nephrology and Rheumatology, University Medical Center Göttingen, Goettingen, Germany
| | - Dennis Pieper
- Clinic for Nephrology and Rheumatology, University Medical Center Göttingen, Goettingen, Germany
| | - Gry Helene Dihazi
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Angelika Hafke
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Stefan Kohl
- Pediatric Nephrology, Children's and Adolescents' Hospital, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Max C Liebau
- Department of Pediatrics, Center for Family Health, Center for Molecular Medicine and Center for Rare Diseases, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Jens König
- University Children's Hospital Münster, Münster, Germany
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Oliver Gross
- Clinic for Nephrology and Rheumatology, University Medical Center Göttingen, Goettingen, Germany
| | - Manuel Wallbach
- Clinic for Nephrology and Rheumatology, University Medical Center Göttingen, Goettingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| |
Collapse
|
8
|
Dahmer-Heath M, Gerß J, Fliser D, Liebau MC, Speer T, Telgmann AK, Burgmaier K, Pennekamp P, Pape L, Schaefer F, Konrad M, König JC. Urinary Dickkopf-3 Reflects Disease Severity and Predicts Short-Term Kidney Function Decline in Renal Ciliopathies. Kidney Int Rep 2025; 10:197-208. [PMID: 39810774 PMCID: PMC11725807 DOI: 10.1016/j.ekir.2024.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/17/2024] [Accepted: 09/30/2024] [Indexed: 01/16/2025] Open
Abstract
Introduction Phenotypic heterogeneity and unpredictability of individual disease progression present enormous challenges in ultrarare renal ciliopathies. The tubular-derived glycoprotein, Dickkopf-related protein 3 (DKK3) is a promising biomarker for kidney fibrosis and prediction of kidney function decline. Here, we measured urinary DKK3 (uDKK3) levels in 195 pediatric patients with renal ciliopathy to assess its potential as a discriminative and prediction marker. Methods uDKK3 concentration was measured in 357 spot urine samples from 247 individuals, including 52 healthy age-matched controls. Disease entities comprised nephronophthisis (NPH) (n = 37), autosomal recessive polycystic kidney disease (ARPKD) (n = 61), Bardet Biedl syndrome (BBS) (n = 57), and hepatocyte nuclear factor 1 beta (HNF1B)-nephropathy (n = 40). The results were correlated with chronic kidney disease (CKD) stage and annual estimated glomerular filtration rate (eGFR) decline. Results Median uDKK3-to-creatinine ratios (uDKK3/crea) in all disease entities were significantly higher compared with healthy controls (11pg/mg uDKK3/crea, P < 0.001): NPH, 1.219 pg/mg; HNF1B, 731 pg/mg; BBS, 541 pg/mg; and ARPKD, 437 pg/mg. A significant correlation of CKD stage with uDKK3 levels was observed for all disease entities (P < 0.0001) with no other clinical parameter having a relevant impact. In our cohort, uDKK3 values >4.700 pg/mg were associated with a significantly greater annual eGFR loss independently of diagnosis and eGFR (P = 0.0029). Although we observed a trend toward lower uDKK3 levels in glomerulopathies compared to renal ciliopathies, there was no discriminative difference between individual ciliopathy entities (P = 0.2637). Conclusion In renal ciliopathies, uDKK3 is a marker to assess disease severity and estimate short-term kidney function decline.
Collapse
Affiliation(s)
- Mareike Dahmer-Heath
- Department of General Pediatrics, University Children's Hospital Münster, Münster, Germany
| | - Joachim Gerß
- Institute of Biostatistics and Clinical Research, University of Münster, Münster, Germany
| | - Danilo Fliser
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Medical Center, Homburg/ Saar, Germany
| | - Max Christoph Liebau
- Department of Pediatrics, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Center for Family Health and Center for Rare Disease, University Hospital Cologne and Medical Faculty, University of Cologne, Cologne, Germany
| | - Thimoteus Speer
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Medical Center, Homburg/ Saar, Germany
- Else Kroener Fresenius Center for Nephrological Research, University Hospital Frankfurt, Frankfurt, Germany
| | | | - Kathrin Burgmaier
- Department of Pediatrics, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
- Faculty of Applied Healthcare Science, Deggendorf Institute of Technology, Deggendorf, Germany
| | - Petra Pennekamp
- Department of General Pediatrics, University Children's Hospital Münster, Münster, Germany
| | - Lars Pape
- Department of Pediatrics II, University Hospital of Essen, Essen, Germany
| | - Franz Schaefer
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Heidelberg, Germany
| | - Martin Konrad
- Department of General Pediatrics, University Children's Hospital Münster, Münster, Germany
| | - Jens Christian König
- Department of General Pediatrics, University Children's Hospital Münster, Münster, Germany
| | - NEOCYST Consortium10
- Department of General Pediatrics, University Children's Hospital Münster, Münster, Germany
- Institute of Biostatistics and Clinical Research, University of Münster, Münster, Germany
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Medical Center, Homburg/ Saar, Germany
- Department of Pediatrics, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Center for Family Health and Center for Rare Disease, University Hospital Cologne and Medical Faculty, University of Cologne, Cologne, Germany
- Else Kroener Fresenius Center for Nephrological Research, University Hospital Frankfurt, Frankfurt, Germany
- Faculty of Applied Healthcare Science, Deggendorf Institute of Technology, Deggendorf, Germany
- Department of Pediatrics II, University Hospital of Essen, Essen, Germany
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Heidelberg, Germany
| |
Collapse
|
9
|
Schuster A, Steines L, Banas B, Bergler T. Dickkopf 3 as a New Monitoring Tool for Kidney Function After Living Kidney Donation. J Clin Med 2024; 13:7454. [PMID: 39685912 DOI: 10.3390/jcm13237454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/21/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Even today, a non-invasive biomarker to identify donors with enhanced risk for renal impairment is missing. Dickkopf 3 (DKK3) can cause tubulointerstitial fibrosis and is associated with rapid eGFR loss. The aim of our work was to analyze whether DKK3 can be used as a non-invasive alert marker for an increased risk of loss of kidney function in living kidney donors (LKDs). Methods: All donors who were examined between July 2022 and June 2023 (n = 117) were included. DKK3 was measured in the urine. The collected patient-related data were compared with parameters before donation. The study cohort was stratified by DKK3 values (≥200). Results: In the follow-up, 89 donors had a DKK3 value < 200 (group 1) and 28 donors had a DKK3 value ≥200 (group 2). During post-donation follow-up, renal function in group 1 was significantly better than that in group 2 (p = 0.01), although no difference in renal function before donation was detected (p = 0.84). Group 2 showed also a greater eGFR loss over time than group 1. Conclusions: LKDs with elevated DKK3 levels in the FU had impaired kidney function without evidence of increased risk factors pre-donation. DKK3 can represent a possible monitoring tool for kidney function in LKDs.
Collapse
Affiliation(s)
- Antonia Schuster
- Department of Nephrology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Louisa Steines
- Department of Nephrology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Bernhard Banas
- Department of Nephrology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Tobias Bergler
- Department of Nephrology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
- Medical Clinic III-Nephrology, Hospital Ingolstadt, Krumenauerstr. 25, 85049 Ingolstadt, Germany
| |
Collapse
|
10
|
Song J, Chen Y, Chen Y, Qiu M, Xiang W, Ke B, Fang X. DKK3 promotes renal fibrosis by increasing MFF-mediated mitochondrial dysfunction in Wnt/β-catenin pathway-dependent manner. Ren Fail 2024; 46:2343817. [PMID: 38682264 PMCID: PMC11060011 DOI: 10.1080/0886022x.2024.2343817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 04/11/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) lacks effective treatments and renal fibrosis (RF) is one of CKD's outcomes. Dickkopf 3 (DKK3) has been identified as an agonist in CKD. However, the underlying mechanisms of DKK3 in CKD are not fully understood. METHODS H2O2-treated HK-2 cells and ureteric obstruction (UUO) mice were used as RF models. Biomarkers, Masson staining, PAS staining, and TUNEL were used to assess kidney function and apoptosis. Oxidative stress and mitochondria function were also evaluated. CCK-8 and flow cytometry were utilized to assess cell viability and apoptosis. Western blotting, IHC, and qRT-PCR were performed to detect molecular expression levels. Immunofluorescence was applied to determine the subcellular localization. Dual luciferase assay, MeRIP, RIP, and ChIP were used to validate the m6A level and the molecule interaction. RESULTS DKK3 was upregulated in UUO mouse kidney tissue and H2O2-treated HK-2 cells. Knockdown of DKK3 inhibited oxidative stress, maintained mitochondrial homeostasis, and alleviated kidney damage and RF in UUO mice. Furthermore, DKK3 silencing suppressed HK-2 cell apoptosis, oxidative stress, and mitochondria fission. Mechanistically, DKK3 upregulation was related to the high m6A level regulated by METTL3. DKK3 activated TCF4/β-catenin and enhanced MFF transcriptional expression by binding to its promoter. Overexpression of MFF reversed in the inhibitory effect of DKK3 knockdown on cell damage. CONCLUSION Upregulation of DKK3 caused by m6A modification activated the Wnt/β-catenin pathway to increase MFF transcriptional expression, leading to mitochondrial dysfunction and oxidative stress, thereby promoting RF progression.
Collapse
Affiliation(s)
- Jianling Song
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Yanxia Chen
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Yan Chen
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Minzi Qiu
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Wenliu Xiang
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Ben Ke
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Xiangdong Fang
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, P.R. China
| |
Collapse
|
11
|
Pieper D, Sandek A, Schäfer AK, Dihazi H, Dihazi GH, Leha A, Zeisberg M, Lüders S, Koziolek M, Wallbach M. Urinary Dickkopf-3 as a Potential Marker for Estimated Glomerular Filtration Rate Decline in Patients With Heart Failure. J Am Heart Assoc 2024:e036637. [PMID: 39604022 DOI: 10.1161/jaha.124.036637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 10/09/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Patients with chronic heart failure (HF) show an increased risk for the occurrence of chronic kidney disease and cardiorenal syndrome. Urinary Dickkopf-3 (uDKK3), a stress-induced, tubular profibrotic glycoprotein, may be elevated in HF as early as in New York Heart Association class I HF and may indicate subsequent decline in estimated glomerular filtration rate (eGFR). METHODS AND RESULTS uDKK3 levels in patients with HF and controls were measured by enzyme-linked immunosorbent assay. eGFR was determined up to 5 years in HF. Change in eGFR was assessed with respect to baseline uDKK3 using (mixed-effect) linear and logistic regression models. A total of 488 patients with chronic HF and 45 control patients were included. Patients with HF showed higher median uDKK3 levels than controls (259.6 pg/mg creatinine [interquartile range (IQR), 119.2-509.4 pg/mg creatinine] versus 107.5 pg/mg creatinine [IQR, 60.5-181.2 pg/mg creatinine], P<0.001). Regression models demonstrated a significant association between log uDKK3 and the decline in eGFR during a median of 13 months (IQR, 12-59 months) (estimated higher eGFR loss, 0.8039 mL/min per 1.73 m2/year [95% CI, 0.002-1.606 mL/min per 1.73 m2/year], P=0.049; odds ratio, 1.345 [95% CI, 1.049-1.741], P=0.021). uDKK3 levels ≥354 pg/mg creatinine were associated with a significantly higher risk for eGFR decline at 1-year follow-up (estimated higher eGFR loss, 4.538 mL/min per 1.73 m2 [95% CI, 1.482-9.593 mL/min per 1.73 m2]), P=0.004). Even patients with HF without chronic kidney disease (n=334) had higher uDKK3 levels compared with controls (233.4 [IQR, 109.0-436.9 pg/mg creatinine] versus 107.5 [IQR, 60.5-181.2 pg/mg creatinine], P<0.001). CONCLUSIONS The present findings indicate that uDKK3 is a promising prognostic biomarker for subsequent eGFR decline in patients with HF, irrespective of the presence of chronic kidney disease and even in the early stages of HF. This potential allows for early intervention to mitigate the deterioration of kidney function. Further investigation is warranted to validate its clinical utility.
Collapse
Affiliation(s)
- Dennis Pieper
- Department of Nephrology and Rheumatology University Medical Center Göttingen, Georg-August-University Göttingen Göttingen Germany
| | - Anja Sandek
- German Center for Cardiovascular Research (DZHK) Partner Site Göttingen Göttingen Germany
- Department of Cardiology and Pneumology University Medical Center Göttingen Göttingen Germany
| | - Ann-Kathrin Schäfer
- Department of Nephrology and Rheumatology University Medical Center Göttingen, Georg-August-University Göttingen Göttingen Germany
| | - Hassan Dihazi
- Department of Nephrology and Rheumatology University Medical Center Göttingen, Georg-August-University Göttingen Göttingen Germany
| | - Gry Helene Dihazi
- Institute for Clinical Chemistry University Medical Center Göttingen Göttingen Germany
| | - Andreas Leha
- Department of Medical Statistics University Medical Center Göttingen Göttingen Germany
| | - Michael Zeisberg
- Department of Nephrology and Rheumatology University Medical Center Göttingen, Georg-August-University Göttingen Göttingen Germany
| | - Stephan Lüders
- Department of Nephrology and Rheumatology University Medical Center Göttingen, Georg-August-University Göttingen Göttingen Germany
- Department of Nephrology St.-Josefs-Hospital Cloppenburg Germany
| | - Michael Koziolek
- Department of Nephrology and Rheumatology University Medical Center Göttingen, Georg-August-University Göttingen Göttingen Germany
- German Center for Cardiovascular Research (DZHK) Partner Site Göttingen Göttingen Germany
| | - Manuel Wallbach
- Department of Nephrology and Rheumatology University Medical Center Göttingen, Georg-August-University Göttingen Göttingen Germany
- German Center for Cardiovascular Research (DZHK) Partner Site Göttingen Göttingen Germany
| |
Collapse
|
12
|
Rroji M, Spasovski G. Omics Studies in CKD: Diagnostic Opportunities and Therapeutic Potential. Proteomics 2024:e202400151. [PMID: 39523931 DOI: 10.1002/pmic.202400151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Omics technologies have significantly advanced the prediction and therapeutic approaches for chronic kidney disease (CKD) by providing comprehensive molecular insights. This is a review of the current state and future prospects of integrating biomarkers into the clinical practice for CKD, aiming to improve patient outcomes by targeted therapeutic interventions. In fact, the integration of genomic, transcriptomic, proteomic, and metabolomic data has enhanced our understanding of CKD pathogenesis and identified novel biomarkers for an early diagnosis and targeted treatment. Advanced computational methods and artificial intelligence (AI) have further refined multi-omics data analysis, leading to more accurate prediction models for disease progression and therapeutic responses. These developments highlight the potential to improve CKD patient care with a precise and individualized treatment plan .
Collapse
Affiliation(s)
- Merita Rroji
- Faculty of Medicine, Department of Nephrology, University of Medicine Tirana, Tirana, Albania
| | - Goce Spasovski
- Medical Faculty, Department of Nephrology, University of Skopje, Skopje, North Macedonia
| |
Collapse
|
13
|
Weyrich M, Cremer S, Gerster M, Sarakpi T, Rasper T, Zewinger S, Patyna SR, Leistner DM, Heine GH, Wanner C, März W, Fliser D, Dimmeler S, Zeiher AM, Speer T. Loss of Y Chromosome and Cardiovascular Events in Chronic Kidney Disease. Circulation 2024; 150:746-757. [PMID: 39005209 PMCID: PMC11361358 DOI: 10.1161/circulationaha.124.069139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Chronic kidney disease represents one of the strongest risk factors for cardiovascular diseases, and particularly for heart failure. Despite improved pharmaceutical treatments, mortality remains high. Recently, experimental studies demonstrated that mosaic loss of Y chromosome (LOY) associates with cardiac fibrosis in male mice. Since diffuse cardiac fibrosis is the common denominator for progression of all forms of heart failure, we determined the association of LOY on mortality and cardiovascular disease outcomes in patients with chronic kidney disease. METHODS LOY was quantified in men with stable chronic kidney disease (CARE for HOMe study, n=279) and dialysis patients (4D study, n=544). The association between LOY and mortality, combined cardiovascular and heart failure-specific end points, and echocardiographic measures was assessed. RESULTS In CARE for HOMe, the frequency of LOY increased with age. LOY >17% was associated with increased mortality (heart rate, 2.58 [95% CI, 1.33-5.03]) and risk for cardiac decompensation or death (heart rate, 2.30 [95% CI, 1.23-4.27]). Patients with LOY >17% showed a significant decline of ejection fraction and an increase of E/E' within 5 years. Consistently, in the 4D study, LOY >17% was significantly associated with increased mortality (heart rate, 2.76 [95% CI, 1.83-4.16]), higher risk of death due to heart failure and sudden cardiac death (heart rate, 4.11 [95% CI, 2.09-8.08]), but not atherosclerotic events. Patients with LOY >17% showed significantly higher plasma levels of soluble interleukin 1 receptor-like 1, a biomarker for myocardial fibrosis. Mechanistically, intermediate monocytes from patients with LOY >17% showed significantly higher C-C chemokine receptor type 2 expression and higher plasma levels of the C-C chemokine receptor type 2 chemokine (C-C motif) ligand 2, which may have contributed to increased heart failure events. CONCLUSIONS LOY identifies male patients with chronic kidney disease at high risk for mortality and heart failure events.
Collapse
Affiliation(s)
- Michael Weyrich
- 1Goethe University Frankfurt’s University Hospital, Department of Internal Medicine 4, Nephrology (T.S., S.R.P., M.G., T.S., M.W.)
| | - Sebastian Cremer
- University Hospital, Department of Medicine, Cardiology (S.C., D.M.L.)
- German Center for Cardiovascular Research DZHK, Berlin, Germany (S.C., D.M.L., S.D., A.M.Z.)
| | - Martin Gerster
- Else Kroener-Fresenius Center for Nephrological Research (T.S., S.R.P., M.G., T.S., M.W.)
| | - Tamim Sarakpi
- 1Goethe University Frankfurt’s University Hospital, Department of Internal Medicine 4, Nephrology (T.S., S.R.P., M.G., T.S., M.W.)
- Else Kroener-Fresenius Center for Nephrological Research (T.S., S.R.P., M.G., T.S., M.W.)
| | - Tina Rasper
- Institute for Cardiovascular Regeneration (T.R., S.D., A.M.Z.), Germany
| | - Stephen Zewinger
- Hôpital Robert Schumann, Hôpital Kirchberg, Luxembourg City, Luxembourg (S.Z.)
- Saarland University, Department of Internal Medicine 4, Homburg/Saar, Germany (D.F., G.H.H., S.Z.)
| | - Sammy R. Patyna
- 1Goethe University Frankfurt’s University Hospital, Department of Internal Medicine 4, Nephrology (T.S., S.R.P., M.G., T.S., M.W.)
- Else Kroener-Fresenius Center for Nephrological Research (T.S., S.R.P., M.G., T.S., M.W.)
| | - David M. Leistner
- University Hospital, Department of Medicine, Cardiology (S.C., D.M.L.)
- German Center for Cardiovascular Research DZHK, Berlin, Germany (S.C., D.M.L., S.D., A.M.Z.)
| | - Gunnar H. Heine
- Saarland University, Department of Internal Medicine 4, Homburg/Saar, Germany (D.F., G.H.H., S.Z.)
| | - Christoph Wanner
- University of Wuerzburg, University Hospital, Department of Clinical Research and Epidemiology, Comprehensive Heart Failure Center, Germany (C.W.)
| | - Winfried März
- Else Kroener-Fresenius Center for Nephrological Research (T.S., S.R.P., M.G., T.S., M.W.)
- University of Heidelberg, University Medical Center, Medical Faculty Mannheim, Vth Department of Medicine, Germany (W.M.)
- Medical University of Graz, Clinical Institute of Medical and Chemical Laboratory Diagnostics, Austria (W.M.)
- SYNLAB Holding Deutschland GmbH, SYNLAB Academy, Mannheim, Germany (W.M.)
| | - Danilo Fliser
- Saarland University, Department of Internal Medicine 4, Homburg/Saar, Germany (D.F., G.H.H., S.Z.)
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration (T.R., S.D., A.M.Z.), Germany
- German Center for Cardiovascular Research DZHK, Berlin, Germany (S.C., D.M.L., S.D., A.M.Z.)
| | - Andreas M. Zeiher
- Institute for Cardiovascular Regeneration (T.R., S.D., A.M.Z.), Germany
- German Center for Cardiovascular Research DZHK, Berlin, Germany (S.C., D.M.L., S.D., A.M.Z.)
| | - Thimoteus Speer
- 1Goethe University Frankfurt’s University Hospital, Department of Internal Medicine 4, Nephrology (T.S., S.R.P., M.G., T.S., M.W.)
| |
Collapse
|
14
|
Ehleiter H, Miranda J, Boes D, Scheidt U, von Vietinghoff S, Schwab S. Peritoneal and renal DKK3 clearance in peritoneal dialysis. BMC Nephrol 2024; 25:268. [PMID: 39179976 PMCID: PMC11342714 DOI: 10.1186/s12882-024-03715-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Urinary Dickkopf 3 (DKK3) excretion is a recently established biomarker of renal functional development. Its excretion into the peritoneal cavity has not been reported. We here studied DKK3 in peritoneal dialysis. METHODS DKK3 was assessed in serum, urine and dialysate in a prevalent adult peritoneal dialysis cohort and its concentration analyzed in relation to creatinine and clinical characteristics. RESULTS Highest DKK3 concentrations were found in serum, followed by urine. Dialysate concentrations were significantly lower. Dialysate DKK3 correlated with both other compartments. Serum, dialysate and urine values were stable during three months of follow-up. Continuous ambulatory dialysis (CAPD) but not cycler-assisted peritoneal dialysis (CCPD) volume-dependently increased peritoneal DKK3 in relation to creatinine. RAAS blockade significantly decreased urinary, but not serum or peritoneal DKK3. CONCLUSION Our data provide a detailed characterization of DKK3 in peritoneal dialysis. They support the notion that the RAAS system is essential for renal DKK3 handling.
Collapse
Affiliation(s)
- Hagen Ehleiter
- Nephrology Section, Medical Clinic 1, University Hospital Bonn, Rheinische Friedrich- Wilhelms Universität Bonn, Venusberg Campus 1, D-53127, Bonn, Germany
| | - Julia Miranda
- Nephrology Section, Medical Clinic 1, University Hospital Bonn, Rheinische Friedrich- Wilhelms Universität Bonn, Venusberg Campus 1, D-53127, Bonn, Germany
| | - Dominik Boes
- Kuratorium for Dialysis, KfH Renal Center, Bonn, Germany
| | - Uta Scheidt
- Nephrology Section, Medical Clinic 1, University Hospital Bonn, Rheinische Friedrich- Wilhelms Universität Bonn, Venusberg Campus 1, D-53127, Bonn, Germany
| | - Sibylle von Vietinghoff
- Nephrology Section, Medical Clinic 1, University Hospital Bonn, Rheinische Friedrich- Wilhelms Universität Bonn, Venusberg Campus 1, D-53127, Bonn, Germany
| | - Sebastian Schwab
- Nephrology Section, Medical Clinic 1, University Hospital Bonn, Rheinische Friedrich- Wilhelms Universität Bonn, Venusberg Campus 1, D-53127, Bonn, Germany.
| |
Collapse
|
15
|
Duan ZY, Zhang C, Chen XM, Cai GY. Blood and urine biomarkers of disease progression in IgA nephropathy. Biomark Res 2024; 12:72. [PMID: 39075557 PMCID: PMC11287988 DOI: 10.1186/s40364-024-00619-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/12/2024] [Indexed: 07/31/2024] Open
Abstract
The prognosis of patients with IgA nephropathy (IgAN) is variable but overall not good. Almost all patients with IgAN are at risk of developing end-stage renal disease within their expected lifetime. The models presently available for prediction of the risk of progression of IgAN, including the International IgA Nephropathy Prediction Tool, consist of traditional clinical, pathological, and therapeutic indicators. Finding biomarkers to improve the existing risk prediction models or replace pathological indicators is important for clinical practice. Many studies have attempted to identify biomarkers for prediction of progression of IgAN, such as galactose-deficient IgA1, complement, a spectrum of protein biomarkers, non-coding RNA, and shedding cells. This article reviews the biomarkers of progression of IgAN identified in recent years, with a focus on those with clinical value, in particular the combination of multiple biomarkers into a biomarker spectrum. Future research should focus on establishing a model based primarily on biomarkers that can predict progression of IgAN and testing it in various patient cohorts.
Collapse
Affiliation(s)
- Zhi-Yu Duan
- Department of Nephrology, State Key Laboratory of Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, First Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Kidney Diseases, Beijing, 100853, China
| | - Chun Zhang
- Department of Nephrology, State Key Laboratory of Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, First Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Kidney Diseases, Beijing, 100853, China
| | - Xiang-Mei Chen
- Department of Nephrology, State Key Laboratory of Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, First Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Kidney Diseases, Beijing, 100853, China
| | - Guang-Yan Cai
- Department of Nephrology, State Key Laboratory of Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, First Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Kidney Diseases, Beijing, 100853, China.
| |
Collapse
|
16
|
López Iglesias A, Blanco Pardo M, Rodríguez Magariños C, Pértega S, Sierra Castro D, García Falcón T, Rodríguez-Carmona A, Pérez Fontán M. Association of urinary excretion rates of uric acid with biomarkers of kidney injury in patients with advanced chronic kidney disease. PLoS One 2024; 19:e0304105. [PMID: 38861521 PMCID: PMC11166352 DOI: 10.1371/journal.pone.0304105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 05/06/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND The potential influence of hyperuricemia on the genesis and progression of chronic kidney disease (CKD) remains controversial. In general, the correlation between blood levels of uric acid (UA) and the rate of progression of CKD is considered to be modest, if any, and the results of relevant trials oriented to disclose the effect of urate-lowering therapies on this outcome have been disappointing. Urinary excretion rates of UA could reflect more accurately the potential consequences of urate-related kidney injury. METHOD Using a cross-sectional design, we investigated the correlation between different estimators of the rates of urinary excretion of UA (total 24-hour excretion, mean urinary concentration, renal clearance and fractional excretion)(main study variables), on one side, and urinary levels of selected biomarkers of kidney injury and CKD progression (DKK3, KIM1, NGAL, interleukin 1b and MCP)(main outcome variables), in 120 patients with advanced CKD (mean glomerular filtration rate 21.5 mL/minute). We took into consideration essential demographic, clinical and analytic variables with a potential confounding effect on the explored correlations (control variables). Spearman's rho correlation and nonlinear generalized additive regression models (GAM) with p-splines smoothers were used for statistical analysis. MAIN RESULTS Multivariate analysis disclosed independent correlations between urinary UA concentrations, clearances and fractional excretion rates (but not plasma UA or total 24-hour excretion rates of UA), on one side, and the scrutinized markers. These correlations were more consistent for DKK3 and NGAL than for the other biomarkers. Glomerular filtration rate, proteinuria and treatment with statins or RAA axis antagonists were other independent correlates of the main outcome variables. CONCLUSIONS Our results support the hypothesis that urinary excretion rates of UA may represent a more accurate marker of UA-related kidney injury than plasma levels of this metabolite, in patients with advanced stages of CKD. Further, longitudinal studies will be necessary, to disclose the clinical significance of these findings.
Collapse
Affiliation(s)
| | | | | | - Sonia Pértega
- Rheumatology and Health Research Group, Faculty of Health Sciences, A Coruña University, A Coruña, Spain
- Nursing and Health Care Research Group, A Coruña Institute of Biomedical Reasearch (INIBIC), A Coruña, Spain
| | | | | | | | - Miguel Pérez Fontán
- División of Nephrology, A Coruña University Hospital, A Coruña, Spain
- Department of Medicine, Faculty of Health Sciences, A Coruña University, A Coruña, Spain
| |
Collapse
|
17
|
Scurt FG, Bose K, Mertens PR, Chatzikyrkou C, Herzog C. Cardiac Surgery-Associated Acute Kidney Injury. KIDNEY360 2024; 5:909-926. [PMID: 38689404 PMCID: PMC11219121 DOI: 10.34067/kid.0000000000000466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
AKI is a common and serious complication of cardiac surgery that has a significant impact on patient morbidity and mortality. The Kidney Disease Improving Global Outcomes definition of AKI is widely used to classify and identify AKI associated with cardiac surgery (cardiac surgery-associated AKI [CSA-AKI]) on the basis of changes in serum creatinine and/or urine output. There are various preoperative, intraoperative, and postoperative risk factors for the development of CSA-AKI which should be recognized and addressed as early as possible to expedite its diagnosis, reduce its occurrence, and prevent or ameliorate its devastating complications. Crucial issues are the inaccuracy of serum creatinine as a surrogate parameter of kidney function in the perioperative setting of cardiothoracic surgery and the necessity to discover more representative markers of the pathophysiology of AKI. However, except for the tissue inhibitor of metalloproteinase-2 and insulin-like growth factor binding protein 7 ratio, other diagnostic biomarkers with an acceptable sensitivity and specificity are still lacking. This article provides a comprehensive review of various aspects of CSA-AKI, including pathogenesis, risk factors, diagnosis, biomarkers, classification, prevention, and treatment management.
Collapse
Affiliation(s)
- Florian G. Scurt
- Clinic of Nephrology, Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Katrin Bose
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Magdeburg, Magdeburg, Germany
| | - Peter R. Mertens
- Clinic of Nephrology, Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Christos Chatzikyrkou
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Carolin Herzog
- Clinic of Nephrology, Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
18
|
Carbayo J, Verdalles Ú, Díaz-Crespo F, Lázaro A, González-Nicolás M, Arroyo D, Blanco D, Redondo VC, García-Gámiz M, Goicoechea M. Tubular biomarkers and histology in lupus nephritis. Int J Rheum Dis 2024; 27:e15210. [PMID: 38837302 DOI: 10.1111/1756-185x.15210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 05/06/2024] [Accepted: 05/19/2024] [Indexed: 06/07/2024]
Abstract
INTRODUCTION The relevance of tubulo-interstitial involvement for kidney prognosis has recently been emphasized, but validated biomarkers for predicting histology are still lacking. The aim of our study was to evaluate different serum and urinary markers of tubular damage in patients with lupus nephritis (LN) and to correlate them with kidney histopathology. METHODS A single-center retrospective study was conducted from January 2016 to December 2021. Serum and urine samples were collected on the same day of kidney biopsy and correlated with histologic data from a cohort of 15 LN patients. We analyzed the following urinary markers, adjusted for urine creatinine: beta 2-microglobulin, alpha 1-microglobulin, NGAL, uKIM-1, MCP-1, uDKK-3, and uUMOD. The serum markers sKIM-1 and sUMOD were also analyzed. RESULTS A positive and strong correlation was observed between the degree of interstitial fibrosis (rho = 0.785, p = .001) and tubular atrophy (rho = 0.781, p = .001) and the levels of uDKK3. uUMOD also showed an inverse and moderate correlation with interstitial fibrosis (rho = -0.562, p = .037) and tubular atrophy (rho = -0.694, p = .006). Patients with >10% cortical interstitial inflammation had higher levels of uKIM-1 [4.9 (3.9, 5.5) vs. 0.8 (0.6, 1.5) mcg/mg, p = .001], MCP-1 [3.8 (2. 3, 4.2) vs. 0.7 (0.3, 1.2) mcg/mg, p = .001], sKIM-1 [9.2 (5.9, 32.7) vs. 1.4 (0, 3.5) pg/mL, p = .001], and lower sUMOD [8.7 (0, 39.7) vs. 46.1 (35.7, 53) ng/mL, p = .028]. CONCLUSION The use of specific urinary and serum biomarkers of tubular dysfunction or injury may help to predict certain histologic parameters in LN patients.
Collapse
Affiliation(s)
- Javier Carbayo
- Department of Nephrology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Úrsula Verdalles
- Department of Nephrology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Francisco Díaz-Crespo
- Department of Pathology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Alberto Lázaro
- Renal Pathophysiology Laboratory, Instituto Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Marian González-Nicolás
- Renal Pathophysiology Laboratory, Instituto Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - David Arroyo
- Department of Nephrology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - David Blanco
- Renal Pathophysiology Laboratory, Instituto Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | | | - Mercedes García-Gámiz
- Clinical Biochemistry Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Marian Goicoechea
- Department of Nephrology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| |
Collapse
|
19
|
Jehn U, Altuner U, Henkel L, Menke AF, Strauss M, Pavenstädt H, Reuter S. Urinary Dickkopf 3 Is Not an Independent Risk Factor in a Cohort of Kidney Transplant Recipients and Living Donors. Int J Mol Sci 2024; 25:5376. [PMID: 38791413 PMCID: PMC11121870 DOI: 10.3390/ijms25105376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/23/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Urinary dickkopf 3 (uDKK3) is a marker released by kidney tubular epithelial cells that is associated with the progression of chronic kidney disease (CKD) and may cause interstitial fibrosis and tubular atrophy. Recent evidence suggests that uDKK3 can also predict the loss of kidney function in CKD patients and kidney transplant recipients, regardless of their current renal function. We conducted a prospective study on 181 kidney transplant (KTx) recipients who underwent allograft biopsy to determine the cause, analyzing the relationship between uDKK3 levels in urine, histological findings, and future allograft function progression. Additionally, we studied 82 living kidney donors before unilateral nephrectomy (Nx), 1-3 days after surgery, and 1 year post-surgery to observe the effects of rapid kidney function loss. In living donors, the uDKK3/creatinine ratio significantly increased 5.3-fold 1-3 days after Nx. However, it decreased significantly to a median level of 620 pg/mg after one year, despite the absence of underlying primary kidney pathology. The estimated glomerular filtration rate (eGFR) decreased by an average of 29.3% to approximately 66.5 (±13.5) mL/min/1.73 m2 after one year, with no further decline in the subsequent years. uDKK3 levels increased in line with eGFR loss after Nx, followed by a decrease as the eGFR partially recovered within the following year. However, uDKK3 did not correlate with the eGFR at the single time points in living donors. In KTx recipients, the uDKK3/creatinine ratio was significantly elevated with a median of 1550 pg/mg compared to healthy individuals or donors after Nx. The mean eGFR in the recipient group was 35.5 mL/min/1.73 m2. The uDKK3/creatinine ratio was statistically associated with the eGFR at biopsy but was not independently associated with the eGFR one year after biopsy or allograft loss. In conclusion, uDKK3 correlates with recent and future kidney function and kidney allograft survival in the renal transplant cohort. Nevertheless, our findings indicate that the uDKK3/creatinine ratio has no prognostic influence on future renal outcome in living donors and kidney recipients beyond the eGFR, independent of the presence of acute renal graft pathology, as correlations are GFR-dependent.
Collapse
Affiliation(s)
- Ulrich Jehn
- Department of Medicine D, Division of General Internal Medicine, Nephrology and Rheumatology, University Hospital of Muenster, 48149 Muenster, Germany; (U.A.); (L.H.); (A.F.M.); (S.R.)
| | - Ugur Altuner
- Department of Medicine D, Division of General Internal Medicine, Nephrology and Rheumatology, University Hospital of Muenster, 48149 Muenster, Germany; (U.A.); (L.H.); (A.F.M.); (S.R.)
| | - Lino Henkel
- Department of Medicine D, Division of General Internal Medicine, Nephrology and Rheumatology, University Hospital of Muenster, 48149 Muenster, Germany; (U.A.); (L.H.); (A.F.M.); (S.R.)
| | - Amélie Friederike Menke
- Department of Medicine D, Division of General Internal Medicine, Nephrology and Rheumatology, University Hospital of Muenster, 48149 Muenster, Germany; (U.A.); (L.H.); (A.F.M.); (S.R.)
| | - Markus Strauss
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure Medicine, University Hospital Muenster, 48149 Muenster, Germany;
| | - Hermann Pavenstädt
- Department of Medicine D, Division of General Internal Medicine, Nephrology and Rheumatology, University Hospital of Muenster, 48149 Muenster, Germany; (U.A.); (L.H.); (A.F.M.); (S.R.)
| | - Stefan Reuter
- Department of Medicine D, Division of General Internal Medicine, Nephrology and Rheumatology, University Hospital of Muenster, 48149 Muenster, Germany; (U.A.); (L.H.); (A.F.M.); (S.R.)
| |
Collapse
|
20
|
Peschard VG, Scherzer R, Katz R, Chen TK, Bullen AL, Campos K, Estrella MM, Ix JH, Shlipak MG. Association of Urinary Dickkopf-3 Levels with Cardiovascular Events and Kidney Disease Progression in Systolic Blood Pressure Intervention Trial. KIDNEY360 2024; 5:690-697. [PMID: 38472135 PMCID: PMC11146650 DOI: 10.34067/kid.0000000000000413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024]
Abstract
Key Points In unadjusted analyses, elevated urinary Dickkopf-3 levels were strongly associated with higher risks of cardiovascular disease, ESKD, AKI, and mortality. However, associations were substantially weakened after adjustment for eGFR and albuminuria, suggesting limited prognostic value. Background Urinary Dickkopf-3 (uDKK3) is a tubular epithelial-derived profibrotic protein secreted into the urine under tubular stress. It is associated with kidney disease progression in persons with CKD and diabetes and postoperative and contrast-associated AKI. We explored associations of uDKK3 with cardiovascular disease (CVD), kidney, and mortality outcomes within the subset of Systolic Blood Pressure Intervention Trial participants with nondiabetic CKD. Methods We included 2344 participants with eGFR <60 ml/min per 1.73 m2 at baseline. We used Cox proportional hazards models to evaluate associations of uDKK3 with CVD (acute decompensated heart failure, myocardial infarction, acute coronary syndrome, stroke, or CVD death), kidney outcomes (incident ESKD, incident AKI, and eGFR decline ≥30%), and all-cause mortality. We used linear mixed models to examine the association of uDKK3 with annual percentage change in eGFR. Models were adjusted for demographic and clinical characteristics, eGFR, and albuminuria. Results Over a median follow-up of 3.5 years, there were 292 CVD, 73 ESKD, 183 AKI, 471 eGFR decline, and 228 mortality events. In multivariable models without adjustment for eGFR and albuminuria, uDKK3 was strongly associated with CVD, ESKD, AKI, eGFR decline ≥30%, and mortality. However, after further adjustment for eGFR and albuminuria, uDKK3 was no longer associated with risks for composite CVD (hazard ratio, 1.07; 95% confidence interval, 0.92 to 1.23), ESKD (0.80; 0.62 to 1.02), AKI (1.01; 0.85 to 1.21), eGFR decline ≥30% (0.88; 0.79 to 0.99), or mortality (1.02; 0.87 to 1.20). For the linear eGFR change outcome, higher uDKK3 also had no association in the fully adjusted model (−0.03; −0.41 to 0.36). Conclusions Among individuals with hypertension and nondiabetic CKD, higher uDKK3 appeared to have associations with a greater risk of CVD events, incident ESKD, incident AKI, eGFR decline ≥30%, and mortality but these associations were not independent of eGFR and albuminuria.
Collapse
Affiliation(s)
- Vanessa-Giselle Peschard
- University of California, San Francisco, California
- Kidney Health Research Collaborative, San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - Rebecca Scherzer
- University of California, San Francisco, California
- Kidney Health Research Collaborative, San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - Ronit Katz
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Teresa K. Chen
- University of California, San Francisco, California
- Kidney Health Research Collaborative, San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - Alexander L. Bullen
- Division of Nephrology-Hypertension, Department of Medicine, University of California, San Diego, California
| | - Kasey Campos
- Kidney Health Research Collaborative, San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - Michelle M. Estrella
- University of California, San Francisco, California
- Kidney Health Research Collaborative, San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - Joachim H. Ix
- Division of Nephrology-Hypertension, Department of Medicine, University of California, San Diego, California
- Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Michael G. Shlipak
- University of California, San Francisco, California
- Kidney Health Research Collaborative, San Francisco Veterans Affairs Medical Center, San Francisco, California
| |
Collapse
|
21
|
Carbayo J, Verdalles Ú, Díaz-Crespo F, Lázaro A, González-Nicolás M, Arroyo D, Blanco D, García-Gámiz M, Goicoechea M. Tubular biomarkers in proteinuric kidney disease: histology correlation and kidney prognosis of tubular biomarkers. Clin Kidney J 2024; 17:sfae146. [PMID: 38803396 PMCID: PMC11129590 DOI: 10.1093/ckj/sfae146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Indexed: 05/29/2024] Open
Abstract
Background Proteinuria is not only a biomarker of chronic kidney disease (CKD) but also a driver of CKD progression. The aim of this study was to evaluate serum and urinary tubular biomarkers in patients with biopsied proteinuric kidney disease and to correlate them with histology and kidney outcomes. Methods A single-center retrospective study was conducted on a cohort of 156 patients from January 2016 to December 2021. The following urinary and serum biomarkers were analyzed on the day of kidney biopsy: beta 2 microglobulin (β2-mcg), alpha 1 microglobulin (α1-mcg), neutrophil gelatinase-associated lipocalin (NGAL), urinary kidney injury molecule-1 (uKIM-1), monocyte chemoattractant protein-1 (MCP-1), urinary Dickkopf-3 (uDKK3), uromodulin (urinary uUMOD), serum kidney injury molecule-1 (sKIM-1) and serum uromodulin (sUMOD). A composite outcome of kidney progression or death was recorded during a median follow-up period of 26 months. Results Multivariate regression analysis identified sUMOD (β-0.357, P < .001) and uDKK3 (β 0.483, P < .001) as independent predictors of interstitial fibrosis, adjusted for age, estimated glomerular filtration rate (eGFR) and log proteinuria. Elevated levels of MCP-1 [odds ratio 15.61, 95% confidence interval (CI) 3.52-69.20] were associated with a higher risk of cortical interstitial inflammation >10% adjusted for eGFR, log proteinuria and microhematuria. Upper tertiles of uDKK3 were associated with greater eGFR decline during follow-up. Although not a predictor of the composite outcome, doubling of uDKK3 was a predictor of kidney events (hazard ratio 2.26, 95% CI 1.04-4.94) after adjustment for interstitial fibrosis, eGFR and proteinuria. Conclusions Tubular markers may have prognostic value in proteinuric kidney disease, correlating with specific histologic parameters and identifying cases at higher risk of CKD progression.
Collapse
Affiliation(s)
- Javier Carbayo
- Department of Nephrology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Úrsula Verdalles
- Department of Nephrology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Francisco Díaz-Crespo
- Department of Pathology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Alberto Lázaro
- Renal Pathophysiology Laboratory, Instituto Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Marian González-Nicolás
- Renal Pathophysiology Laboratory, Instituto Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - David Arroyo
- Department of Nephrology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - David Blanco
- Renal Pathophysiology Laboratory, Instituto Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Mercedes García-Gámiz
- Department of Biochemistry, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Marian Goicoechea
- Department of Nephrology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| |
Collapse
|
22
|
Song J, Chen Y, Chen Y, Qiu M, Xiang W, Ke B, Fang X. DKK3 promotes oxidative stress injury and fibrosis in HK-2 cells by activating NOX4 via β-catenin/TCF4 signaling. Mol Cell Biochem 2024; 479:1231-1241. [PMID: 37368156 DOI: 10.1007/s11010-023-04789-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023]
Abstract
Oxidative stress and fibrosis may accelerate the progression of chronic kidney disease (CKD). DKK3 is related to regulating renal fibrosis and CKD. However, the molecular mechanism of DKK3 in regulating oxidative stress and fibrosis during CKD development has not been clarified, which deserves to be investigated. Human proximal tubule epithelial cells (HK-2 cells) were treated with H2O2 to establish a cell model of renal fibrosis. The mRNA and protein expressions were analyzed using qRT-PCR and western blot, respectively. Cell viability and apoptosis were evaluated using MTT assay and flow cytometry, respectively. ROS production was estimated using DCFH-DA. The interactions among TCF4, β-catenin and NOX4 were validated using luciferase activity assay, ChIP and Co-IP. Herein, our results revealed that DKK3 was highly expressed in HK-2 cells treated with H2O2. DKK3 depletion increased H2O2-treated HK-2 cell viability and reduced cell apoptosis, oxidative stress, and fibrosis. Mechanically, DKK3 promoted formation of the β-catenin/TCF4 complex, and activated NOX4 transcription. Upregulation of NOX4 or TCF4 weakened the inhibitory effect of DKK3 knockdown on oxidative stress and fibrosis in H2O2-stimulated HK-2 cells. All our results suggested that DKK3 accelerated oxidative stress and fibrosis through promoting β-catenin/TCF4 complex-mediated activation of NOX4 transcription, which could lead to novel molecules and therapeutic targets for CKD.
Collapse
Affiliation(s)
- Jianling Song
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Yanxia Chen
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Yan Chen
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Minzi Qiu
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Wenliu Xiang
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Ben Ke
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, People's Republic of China.
| | - Xiangdong Fang
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, People's Republic of China.
| |
Collapse
|
23
|
Rivedal M, Mikkelsen H, Marti HP, Liu L, Kiryluk K, Knoop T, Bjørneklett R, Haaskjold YL, Furriol J, Leh S, Paunas F, Bábíčková J, Scherer A, Serre C, Eikrem O, Strauss P. Glomerular transcriptomics predicts long term outcome and identifies therapeutic strategies for patients with assumed benign IgA nephropathy. Kidney Int 2024; 105:717-730. [PMID: 38154557 DOI: 10.1016/j.kint.2023.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/17/2023] [Accepted: 12/08/2023] [Indexed: 12/30/2023]
Abstract
Some patients diagnosed with benign IgA nephropathy (IgAN) develop a progressive clinical course, not predictable by known clinical or histopathological parameters. To assess if gene expression can differentiate between progressors and non-progressors with assumed benign IgAN, we tested microdissected glomeruli from archival kidney biopsy sections from adult patients with stable clinical remission (21 non-progressors) or from 15 patients that had undergone clinical progression within a 25-year time frame. Based on 1 240 differentially expressed genes from patients with suitable sequencing results, we identified eight IgAN progressor and nine non-progressor genes using a two-component classifier. These genes, including APOL5 and ZXDC, predicted disease progression with 88% accuracy, 75% sensitivity and 100% specificity on average 21.6 years before progressive disease was clinically documented. APOL lipoproteins are associated with inflammation, autophagy and kidney disease while ZXDC is a zinc-finger transcription factor modulating adaptive immunity. Ten genes from our transcriptomics data overlapped with an external genome wide association study dataset, although the gene set enrichment test was not statistically significant. We also identified 45 drug targets in the DrugBank database, including angiotensinogen, a target of sparsentan (dual antagonist of the endothelin type A receptor and the angiotensin II type 1 receptor) currently investigated for IgAN treatment. Two validation cohorts were used for substantiating key results, one by immunohistochemistry and the other by nCounter technology. Thus, glomerular mRNA sequencing from diagnostic kidney biopsies from patients with assumed benign IgAN can differentiate between future progressors and non-progressors at the time of diagnosis.
Collapse
Affiliation(s)
- Mariell Rivedal
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Håvard Mikkelsen
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Hans-Peter Marti
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Lili Liu
- Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, New York, USA
| | - Krzysztof Kiryluk
- Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, New York, USA; Institute for Genomic Medicine, Columbia University, New York, New York, USA
| | - Thomas Knoop
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Rune Bjørneklett
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; Emergency Care Clinic, Haukeland University Hospital, Bergen, Norway
| | - Yngvar Lunde Haaskjold
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Jessica Furriol
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Sabine Leh
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Flavia Paunas
- Department of Medicine, Haugesund Hospital, Haugesund, Norway
| | - Janka Bábíčková
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Andreas Scherer
- Spheromics, Kontiolahti, Finland; Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Camille Serre
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Oystein Eikrem
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Philipp Strauss
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| |
Collapse
|
24
|
Rupprecht H, Catanese L, Amann K, Hengel FE, Huber TB, Latosinska A, Lindenmeyer MT, Mischak H, Siwy J, Wendt R, Beige J. Assessment and Risk Prediction of Chronic Kidney Disease and Kidney Fibrosis Using Non-Invasive Biomarkers. Int J Mol Sci 2024; 25:3678. [PMID: 38612488 PMCID: PMC11011737 DOI: 10.3390/ijms25073678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Effective management of chronic kidney disease (CKD), a major health problem worldwide, requires accurate and timely diagnosis, prognosis of progression, assessment of therapeutic efficacy, and, ideally, prediction of drug response. Multiple biomarkers and algorithms for evaluating specific aspects of CKD have been proposed in the literature, many of which are based on a small number of samples. Based on the evidence presented in relevant studies, a comprehensive overview of the different biomarkers applicable for clinical implementation is lacking. This review aims to compile information on the non-invasive diagnostic, prognostic, and predictive biomarkers currently available for the management of CKD and provide guidance on the application of these biomarkers. We specifically focus on biomarkers that have demonstrated added value in prospective studies or those based on prospectively collected samples including at least 100 subjects. Published data demonstrate that several valid non-invasive biomarkers of potential value in the management of CKD are currently available.
Collapse
Affiliation(s)
- Harald Rupprecht
- Department of Nephrology, Angiology and Rheumatology, Klinikum Bayreuth GmbH, 95445 Bayreuth, Germany; (H.R.); (L.C.)
- Department of Nephrology, Medizincampus Oberfranken, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Kuratorium for Dialysis and Transplantation (KfH) Bayreuth, 95445 Bayreuth, Germany
| | - Lorenzo Catanese
- Department of Nephrology, Angiology and Rheumatology, Klinikum Bayreuth GmbH, 95445 Bayreuth, Germany; (H.R.); (L.C.)
- Department of Nephrology, Medizincampus Oberfranken, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Kuratorium for Dialysis and Transplantation (KfH) Bayreuth, 95445 Bayreuth, Germany
| | - Kerstin Amann
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Felicitas E. Hengel
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.E.H.); (T.B.H.); (M.T.L.)
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany
| | - Tobias B. Huber
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.E.H.); (T.B.H.); (M.T.L.)
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany
| | | | - Maja T. Lindenmeyer
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.E.H.); (T.B.H.); (M.T.L.)
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany
| | - Harald Mischak
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (A.L.); (H.M.); (J.S.)
| | - Justyna Siwy
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (A.L.); (H.M.); (J.S.)
| | - Ralph Wendt
- Department of Nephrology, Hospital St. Georg, 04129 Leipzig, Germany;
| | - Joachim Beige
- Department of Nephrology, Hospital St. Georg, 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
| |
Collapse
|
25
|
Catanese L, Rupprecht H, Huber TB, Lindenmeyer MT, Hengel FE, Amann K, Wendt R, Siwy J, Mischak H, Beige J. Non-Invasive Biomarkers for Diagnosis, Risk Prediction, and Therapy Guidance of Glomerular Kidney Diseases: A Comprehensive Review. Int J Mol Sci 2024; 25:3519. [PMID: 38542491 PMCID: PMC10970781 DOI: 10.3390/ijms25063519] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 04/09/2025] Open
Abstract
Effective management of glomerular kidney disease, one of the main categories of chronic kidney disease (CKD), requires accurate diagnosis, prognosis of progression, assessment of therapeutic efficacy, and, ideally, prediction of drug response. Multiple biomarkers and algorithms for the assessment of specific aspects of glomerular diseases have been reported in the literature. Though, the vast majority of these have not been implemented in clinical practice or are not available on a global scale due to limited access, missing medical infrastructure, or economical as well as political reasons. The aim of this review is to compile all currently available information on the diagnostic, prognostic, and predictive biomarkers currently available for the management of glomerular diseases, and provide guidance on the application of these biomarkers. As a result of the compiled evidence for the different biomarkers available, we present a decision tree for a non-invasive, biomarker-guided diagnostic path. The data currently available demonstrate that for the large majority of patients with glomerular diseases, valid biomarkers are available. However, despite the obvious disadvantages of kidney biopsy, being invasive and not applicable for monitoring, especially in the context of rare CKD etiologies, kidney biopsy still cannot be replaced by non-invasive strategies.
Collapse
Affiliation(s)
- Lorenzo Catanese
- Department of Nephrology, Angiology and Rheumatology, Klinikum Bayreuth GmbH, 95445 Bayreuth, Germany; (L.C.); (H.R.)
- Kuratorium for Dialysis and Transplantation (KfH) Bayreuth, 95445 Bayreuth, Germany
- Department of Nephrology, Medizincampus Oberfranken, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Harald Rupprecht
- Department of Nephrology, Angiology and Rheumatology, Klinikum Bayreuth GmbH, 95445 Bayreuth, Germany; (L.C.); (H.R.)
- Kuratorium for Dialysis and Transplantation (KfH) Bayreuth, 95445 Bayreuth, Germany
- Department of Nephrology, Medizincampus Oberfranken, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Tobias B. Huber
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (T.B.H.); (M.T.L.); (F.E.H.)
| | - Maja T. Lindenmeyer
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (T.B.H.); (M.T.L.); (F.E.H.)
| | - Felicitas E. Hengel
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (T.B.H.); (M.T.L.); (F.E.H.)
| | - Kerstin Amann
- Department of Nephropathology, Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Ralph Wendt
- Division of Nephrology, St. Georg Hospital, 04129 Leipzig, Germany;
| | - Justyna Siwy
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (J.S.); (H.M.)
| | - Harald Mischak
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (J.S.); (H.M.)
| | - Joachim Beige
- Division of Nephrology, St. Georg Hospital, 04129 Leipzig, Germany;
- Department of Internal Medicine II, Martin-Luther-University Halle-Wittenberg, 06108 Halle, Germany
- Kuratorium for Dialysis and Transplantation (KfH) Leipzig, 04129 Leipzig, Germany
| |
Collapse
|
26
|
Singh R, Watchorn JC, Zarbock A, Forni LG. Prognostic Biomarkers and AKI: Potential to Enhance the Identification of Post-Operative Patients at Risk of Loss of Renal Function. Res Rep Urol 2024; 16:65-78. [PMID: 38476861 PMCID: PMC10928916 DOI: 10.2147/rru.s385856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Acute kidney injury (AKI) is a common complication after surgery and the more complex the surgery, the greater the risk. During surgery, patients are exposed to a combination of factors all of which are associated with the development of AKI. These include hypotension and hypovolaemia, sepsis, systemic inflammation, the use of nephrotoxic agents, tissue injury, the infusion of blood or blood products, ischaemia, oxidative stress and reperfusion injury. Given the risks of AKI, it would seem logical to conclude that early identification of patients at risk of AKI would translate into benefit. The conventional markers of AKI, namely serum creatinine and urine output are the mainstay of defining chronic kidney disease but are less suited to the acute phase. Such concerns are compounded in surgical patients given they often have significantly reduced mobility, suboptimal levels of nutrition and reduced muscle bulk. Many patients may also have misleadingly low serum creatinine and high urine output due to aggressive fluid resuscitation, particularly in intensive care units. Over the last two decades, considerable information has accrued with regard to the performance of what was termed "novel" biomarkers of AKI, and here, we discuss the most examined molecules and performance in surgical settings. We also discuss the application of biomarkers to guide patients' postoperative care.
Collapse
Affiliation(s)
- Rishabh Singh
- Department of Surgery, Royal Surrey Hospital, Guildford, Surrey, UK
| | - James C Watchorn
- Intensive Care Unit, Royal Berkshire NHS Foundation Trust, Reading, Berkshire, UK
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Lui G Forni
- Critical Care Unit, Royal Surrey Hospital, Guildford, Surrey, UK
- School of Medicine, Kate Granger Building, University of Surrey, Guildford, UK
| |
Collapse
|
27
|
Canki E, Kho E, Hoenderop JGJ. Urinary biomarkers in kidney disease. Clin Chim Acta 2024; 555:117798. [PMID: 38280489 DOI: 10.1016/j.cca.2024.117798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/22/2024] [Accepted: 01/22/2024] [Indexed: 01/29/2024]
Abstract
BACKGROUND Chronic kidney disease (CKD) affects many people worldwide and early diagnosis is essential for successful treatment and improved outcome. Unfortunately, current methods are insufficient especially for early disease detection. However, advances in the analytical methods for urinary biomarkers may provide a unique opportunity for diagnosis and management of CKD. This review explores evolving technology and highlights the importance of early marker detection in these patients. APPROACH A search strategy was set up using the terms CKD, biomarkers, and urine. The search included 53 studies comprising 37 biomarkers. The value of these biomarkers for CKD are based on their ability to diagnose CKD, monitor progression, assess mortality and nephrotoxicity. RESULTS KIM-1 was the best marker for diagnosis as it increased with the development of incident CKD. DKK3 increased in patients with declining eGFR, whereas UMOD decreased in those with declining kidney function. Unfortunately, none fulfilled all criteria to adequately assess mortality and nephrotoxicity. CONCLUSION New developments in the field of urinalysis using smart toilets may open several possibilities for urinary biomarkers. This review explored which biomarkers could be used for CKD disease detection and management.
Collapse
Affiliation(s)
- Esra Canki
- Department of Medical BioSciences, Radboudumc, Nijmegen, The Netherlands
| | - Esther Kho
- imec within OnePlanet Research Center, Wageningen, The Netherlands
| | | |
Collapse
|
28
|
Arjune S, Späth MR, Oehm S, Todorova P, Schunk SJ, Lettenmeier K, Chon SH, Bartram MP, Antczak P, Grundmann F, Fliser D, Müller RU. DKK3 as a potential novel biomarker in patients with autosomal polycystic kidney disease. Clin Kidney J 2024; 17:sfad262. [PMID: 38186869 PMCID: PMC10768788 DOI: 10.1093/ckj/sfad262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Indexed: 01/09/2024] Open
Abstract
Backgound Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease, and leads to a steady loss of kidney function in adulthood. The variable course of the disease makes it necessary to identify the patients with rapid disease progression who will benefit the most from targeted therapies and interventions. Currently, magnetic resonance imaging-based volumetry of the kidney is the most commonly used tool for this purpose. Biomarkers that can be easily and quantitatively determined, which allow a prediction of the loss of kidney function, have not yet been established in clinical practice. The glycoprotein Dickkopf 3 (DKK3) which is secreted in the renal tubular epithelium upon stress and contributes to tubulointerstitial fibrosis via the Wnt signaling pathway, was recently described as a biomarker for estimating risk of kidney function loss, but has not been investigated for ADPKD. This study aimed to obtain a first insight into whether DKK3 may indeed improve outcome prediction in ADPKD in the future. Methods In 184 ADPKD patients from the AD(H)PKD registry and 47 healthy controls, the urinary DKK3 (uDKK3) levels were determined using ELISA. Multiple linear regression was used to examine the potential of these values in outcome prediction. Results ADPKD patients showed significantly higher uDKK3 values compared with the controls (mean 1970 ± 5287 vs 112 ± 134.7 pg/mg creatinine). Furthermore, there was a steady increase in uDKK3 with an increase in the Mayo class (A/B 1262 ± 2315 vs class D/E 3104 ± 7627 pg/mg creatinine), the best-established biomarker of progression in ADPKD. uDKK3 also correlated with estimated glomerular filtration rate (eGFR). Patients with PKD1 mutations show higher uDKK3 levels compared with PKD2 patients (PKD1: 2304 ± 5119; PKD2: 506.6 ± 526.8 pg/mg creatinine). Univariate linear regression showed uDKK3 as a significant predictor of future eGFR slope estimation. In multiple linear regression this effect was not significant in models also containing height-adjusted total kidney volume and/or eGFR. However, adding both copeptin levels and the interaction term between copeptin and uDKK3 to the model resulted in a significant predictive value of all these three variables and the highest R2 of all models examined (∼0.5). Conclusion uDKK3 shows a clear correlation with the Mayo classification in patients with ADPKD. uDKK3 levels correlated with kidney function, which could indicate that uDKK3 also predicts a disproportionate loss of renal function in this collective. Interestingly, we found an interaction between copeptin and uDKK3 in our prediction models and the best model containing both variables and their interaction term resulted in a fairly good explanation of variance in eGFR slope compared with previous models. Considering the limited number of patients in these analyses, future studies will be required to confirm the results. Nonetheless, uDKK3 appears to be an attractive candidate to improve outcome prediction of ADPKD in the future.
Collapse
Affiliation(s)
- Sita Arjune
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Center for Rare Diseases Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Martin R Späth
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Simon Oehm
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Polina Todorova
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Stefan J Schunk
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Medical Center, Homburg/Saar, Germany
| | - Katharina Lettenmeier
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Seung-Hun Chon
- Department of General, Visceral, Cancer and Transplant Surgery, University Hospital of Cologne, Cologne, Germany
- Department of Gastroenterology and Hepatology, University Hospital of Cologne, Cologne, Germany
| | - Malte P Bartram
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Philipp Antczak
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Franziska Grundmann
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Danilo Fliser
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Medical Center, Homburg/Saar, Germany
| | - Roman-Ulrich Müller
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Center for Rare Diseases Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| |
Collapse
|
29
|
Azushima K, Kovalik JP, Yamaji T, Ching J, Chng TW, Guo J, Liu JJ, Nguyen M, Sakban RB, George SE, Tan PH, Lim SC, Gurley SB, Coffman TM. Abnormal lactate metabolism is linked to albuminuria and kidney injury in diabetic nephropathy. Kidney Int 2023; 104:1135-1149. [PMID: 37843477 DOI: 10.1016/j.kint.2023.08.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 07/28/2023] [Accepted: 08/04/2023] [Indexed: 10/17/2023]
Abstract
Diabetic nephropathy (DN) is characterized by abnormal kidney energy metabolism, but its causes and contributions to DN pathogenesis are not clear. To examine this issue, we carried out targeted metabolomics profiling in a mouse model of DN that develops kidney disease resembling the human disorder. We found a distinct profile of increased lactate levels and impaired energy metabolism in kidneys of mice with DN, and treatment with an angiotensin-receptor blocker (ARB) reduced albuminuria, attenuated kidney pathology and corrected many metabolic abnormalities, restoring levels of lactate toward normal while increasing kidney ATP content. We also found enhanced expression of lactate dehydrogenase isoforms in DN. Expression of both the LdhA and LdhB isoforms were significantly increased in kidneys of mice, and treatment with ARB significantly reduced their expression. Single-cell sequencing studies showed specific up-regulation of LdhA in the proximal tubule, along with enhanced expression of oxidative stress pathways. There was a significant correlation between albuminuria and lactate in mice, and also in a Southeast Asian patient cohort consisting of individuals with type 2 diabetes and impaired kidney function. In the individuals with diabetes, this association was independent of ARB and angiotensin-converting enzyme inhibitor use. Furthermore, urinary lactate levels predicted the clinical outcomes of doubling of serum creatinine or development of kidney failure, and there was a significant correlation between urinary lactate levels and biomarkers of tubular injury and epithelial stress. Thus, we suggest that kidney metabolic disruptions leading to enhanced generation of lactate contribute to the pathogenesis of DN and increased urinary lactate levels may be a potential biomarker for risk of kidney disease progression.
Collapse
Affiliation(s)
- Kengo Azushima
- Cardiovascular and Metabolic Disorders Signature Research Program, Duke-NUS Medical School, Singapore; Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan; Japan Society for the Promotion of Science, Tokyo, Japan
| | - Jean-Paul Kovalik
- Cardiovascular and Metabolic Disorders Signature Research Program, Duke-NUS Medical School, Singapore
| | - Takahiro Yamaji
- Cardiovascular and Metabolic Disorders Signature Research Program, Duke-NUS Medical School, Singapore; Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Jianhong Ching
- Cardiovascular and Metabolic Disorders Signature Research Program, Duke-NUS Medical School, Singapore
| | - Tze Wei Chng
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Jing Guo
- Cardiovascular and Metabolic Disorders Signature Research Program, Duke-NUS Medical School, Singapore
| | - Jian-Jun Liu
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | - Mien Nguyen
- Cardiovascular and Metabolic Disorders Signature Research Program, Duke-NUS Medical School, Singapore
| | - Rashidah Binte Sakban
- Cardiovascular and Metabolic Disorders Signature Research Program, Duke-NUS Medical School, Singapore
| | - Simi E George
- Cardiovascular and Metabolic Disorders Signature Research Program, Duke-NUS Medical School, Singapore
| | - Puay Hoon Tan
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Su Chi Lim
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore; Diabetes Centre, Khoo Teck Puat Hospital, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore; Lee Kong Chian School of Medicine, Singapore; Nanyang Technological University, Singapore
| | - Susan B Gurley
- Department of Medicine, Division of Nephrology and Hypertension, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Thomas M Coffman
- Cardiovascular and Metabolic Disorders Signature Research Program, Duke-NUS Medical School, Singapore; Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA.
| |
Collapse
|
30
|
Xing H, Jiang Z, Wu Y, Ou S, Qin J, Xue L, Wu W. The role of urinary Dickkopf-3 in the prediction of acute kidney injury: a systematic review meta-analysis. Int Urol Nephrol 2023; 55:3175-3188. [PMID: 37072601 DOI: 10.1007/s11255-023-03593-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/07/2023] [Indexed: 04/20/2023]
Abstract
BACKGROUND To systematically evaluate the diagnostic efficacy of urinary Dickkopf-Related Protein 3 (DKK-3) in acute kidney injury and to explore the clinical application value of urinary DKK-3. METHOD English databases (PubMed, Embase, Cochrane, and WOS) and Chinese databases (VIP, WanFang data, and China National Knowledge Internet) were screened for relevant papers published before March 12, 2023. After literature screening and data extraction, quality assessment was performed according to the QUADAS-2 scoring system. Then, the combined diagnostic and predictive parameters were calculated using a bivariate mixed effect meta-analysis model. Deek's funnel plot asymmetry test assessed publication bias, and Fagan's nomogram plot was used to verify its clinical utility. RESULT A total of 5 studies involving 2787 patients were included in this meta-analysis, of which 4 focused on contrast-induced acute kidney injury (CI-AKI) and 1 focused on AKI associated with cardiac surgery. The analysis showed that urine Dickkopf-3 has high diagnostic accuracy for AKI, with a sensitivity of 0.55 (95% CI [0.41, 0.68]), specificity of 0.80 (95% CI [0.70, 0.87]), positive likelihood ratio (PLR) of 2.7 [1.8, 4.1], negative likelihood ratio (NLR) of 0.56 [0.42, 0.75], diagnostic odds ratio (DOR) of 5 [3, 9], and AUC of 0.74 [0.70-0.77]. We did not perform subgroup analyses for predictive value due to the small number of included studies. CONCLUSION Urinary DKK3 may have limited predictive ability for acute kidney injury, especially for AKI associated with cardiac surgery. Therefore, urinary DKK3 may serve as a potential predictor for AKI. However, clinical studies with larger samples are still needed for validation.
Collapse
Affiliation(s)
- Huameng Xing
- Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory, Luzhou, China
| | - Zheng Jiang
- Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory, Luzhou, China
| | - Yuxuan Wu
- Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory, Luzhou, China
| | - Santao Ou
- Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory, Luzhou, China
| | - Jianhua Qin
- Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory, Luzhou, China
| | - Ling Xue
- Department of Urology, Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Sichuan, 646000, Luzhou, China.
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China.
| | - Weihua Wu
- Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, China.
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China.
- Metabolic Vascular Disease Key Laboratory, Luzhou, China.
| |
Collapse
|
31
|
Cattran DC, Floege J, Coppo R. Evaluating Progression Risk in Patients With Immunoglobulin A Nephropathy. Kidney Int Rep 2023; 8:2515-2528. [PMID: 38106572 PMCID: PMC10719597 DOI: 10.1016/j.ekir.2023.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 12/19/2023] Open
Abstract
The highly variable rate of decline in kidney function in patients with immunoglobulin A nephropathy (IgAN) provides a major clinical challenge. Predicting which patients will progress to kidney failure, and how quickly, is difficult. Multiple novel therapies are likely to be approved in the short-term, but clinicians lack the tools to identify patients most likely to benefit from specific treatments at the right time. Noninvasive and validated markers for selecting at-risk patients and longitudinal monitoring are urgently needed. This review summarizes what is known about demographic, clinical, and histopathologic prognostic markers in the clinician's toolkit, including the International IgAN Prediction Tool. We also briefly review what is known on these topics in children and adolescents with IgAN. Although helpful, currently used markers leave clinicians heavily reliant on histologic features from the diagnostic kidney biopsy and standard clinical data to guide treatment choice, and very few noninvasive markers reflect treatment efficacy over time. Novel prognostic and predictive markers are under clinical investigation, with considerable progress being made in markers of complement activation. Other areas of research are the interplay between gut microbiota and galactose-deficient IgA1 expression; microRNAs; imaging; artificial intelligence; and markers of fibrosis. Given the rate of therapeutic advancement, the remaining gaps in biomarker research need to be addressed. We finish by describing our route to clinical utility of predictive and prognostic markers in IgAN. This route will provide us with the chance to improve IgAN prognosis by using robust, clinically practical markers to inform patient care.
Collapse
Affiliation(s)
| | - Jürgen Floege
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Rosanna Coppo
- Fondazione Ricerca Molinette, Regina Margherita Hospital, Turin, Italy
| |
Collapse
|
32
|
Schmidt-Lauber C, Hänzelmann S, Schunk S, Petersen EL, Alabdo A, Lindenmeyer M, Hausmann F, Kuta P, Renné T, Twerenbold R, Zeller T, Blankenberg S, Fliser D, Huber TB. Kidney outcome after mild to moderate COVID-19. Nephrol Dial Transplant 2023; 38:2031-2040. [PMID: 36657383 PMCID: PMC10468748 DOI: 10.1093/ndt/gfad008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has a remarkable kidney tropism. While kidney effects are common in severe coronavirus disease 2019 (COVID-19), data on non-severe courses are limited. Here we provide a multilevel analysis of kidney outcomes after non-severe COVID-19 to test for eventual kidney sequela. METHODS This cross-sectional study investigates individuals after COVID-19 and matched controls recruited from the Hamburg City Health Study (HCHS) and its COVID-19 program. The HCHS is a prospective population-based cohort study within the city of Hamburg, Germany. During the COVID-19 pandemic the study additionally recruited subjects after polymerase chain reaction-confirmed SARS-CoV-2 infections. Matching was performed by age, sex and education. Main outcomes were estimated glomerular filtration rate (eGFR), albuminuria, Dickkopf3, haematuria and pyuria. RESULTS A total of 443 subjects in a median of 9 months after non-severe COVID-19 were compared with 1328 non-COVID-19 subjects. The mean eGFR was mildly lower in post-COVID-19 than non-COVID-19 subjects, even after adjusting for known risk factors {β = -1.84 [95% confidence interval (CI) -3.16 to -0.52]}. However, chronic kidney disease [odds ratio (OR) 0.90 (95% CI 0.48-1.66)] or severely increased albuminuria [OR 0.76 (95% CI 0.49-1.09)] equally occurred in post-COVID-19 and non-COVID-19 subjects. Haematuria, pyuria and proteinuria were also similar between the two cohorts, suggesting no ongoing kidney injury after non-severe COVID-19. Further, Dickkopf3 was not increased in the post-COVID-19 cohort, indicating no systematic risk for ongoing GFR decline [β = -72.19 (95% CI -130.0 to -14.4)]. CONCLUSION While mean eGFR was slightly lower in subjects after non-severe COVID-19, there was no evidence for ongoing or progressive kidney sequela.
Collapse
Affiliation(s)
| | - Sonja Hänzelmann
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Schunk
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University, Homburg/Saar, Germany
| | - Elina L Petersen
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany
| | - Ammar Alabdo
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maja Lindenmeyer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabian Hausmann
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Piotr Kuta
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Raphael Twerenbold
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany
- University Center of Cardiovascular Science, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK) Partner Site Hamburg–Kiel–Lübeck
| | - Tanja Zeller
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany
- University Center of Cardiovascular Science, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK) Partner Site Hamburg–Kiel–Lübeck
| | - Stefan Blankenberg
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK) Partner Site Hamburg–Kiel–Lübeck
| | - Danilo Fliser
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University, Homburg/Saar, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
33
|
Dziamałek-Macioszczyk P, Winiarska A, Pawłowska A, Wojtacha P, Stompór T. Patterns of Dickkopf-3 Serum and Urine Levels at Different Stages of Chronic Kidney Disease. J Clin Med 2023; 12:4705. [PMID: 37510820 PMCID: PMC10380869 DOI: 10.3390/jcm12144705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/08/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Dickkopf 3 (Dkk3) is a WNT/β-catenin signaling pathway regulator secreted by tubular epithelial cells upon the influence of different stressors. Recently Dkk3 was described as a biomarker of tubular cell injury and a tool that may estimate the risk of chronic kidney disease (CKD) progression. The data about Dkk3 concentrations at particular stages of CKD are lacking. The aim of this study was to measure serum and urine Dkk3 levels in patients with different 'renal status' and evaluate its role as a biomarker of renal damage. One hundred individuals, aged between 24 and 85 years (mean 53.1 ± 17.1), were enrolled in the study. Five groups of 20 subjects each were recruited based on their kidney function. Serum and urine Dkk3 levels were measured by ELISA. The highest median urinary Dkk3 normalized to urinary creatinine was found in patients with established CKD (7051 pg/mg). It was two times higher in renal transplant patients (5705 pg/mg) than in healthy individuals (2654 pg/mg) and the glomerulonephritis group (2470 pg/mg). Urinary Dkk3 was associated with serum creatinine in participants with established CKD and following transplantation. Our results confirm the potential role of Dkk3 as a biomarker of an ongoing renal injury.
Collapse
Affiliation(s)
- Paulina Dziamałek-Macioszczyk
- Department of Nephrology, Hypertension and Internal Medicine, University of Warmia and Mazury in Olsztyn, 10-561 Olsztyn, Poland
| | - Agata Winiarska
- Department of Nephrology, Hypertension and Internal Medicine, University of Warmia and Mazury in Olsztyn, 10-561 Olsztyn, Poland
| | - Anna Pawłowska
- Department of Nephrology, Hypertension and Internal Medicine, University of Warmia and Mazury in Olsztyn, 10-561 Olsztyn, Poland
| | - Paweł Wojtacha
- Department of Industrial and Food Microbiology, University of Warmia and Mazury in Olsztyn, 10-726 Olsztyn, Poland
| | - Tomasz Stompór
- Department of Nephrology, Hypertension and Internal Medicine, University of Warmia and Mazury in Olsztyn, 10-561 Olsztyn, Poland
| |
Collapse
|
34
|
Catanese L, Siwy J, Mischak H, Wendt R, Beige J, Rupprecht H. Recent Advances in Urinary Peptide and Proteomic Biomarkers in Chronic Kidney Disease: A Systematic Review. Int J Mol Sci 2023; 24:ijms24119156. [PMID: 37298105 DOI: 10.3390/ijms24119156] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Biomarker development, improvement, and clinical implementation in the context of kidney disease have been a central focus of biomedical research for decades. To this point, only serum creatinine and urinary albumin excretion are well-accepted biomarkers in kidney disease. With their known blind spot in the early stages of kidney impairment and their diagnostic limitations, there is a need for better and more specific biomarkers. With the rise in large-scale analyses of the thousands of peptides in serum or urine samples using mass spectrometry techniques, hopes for biomarker development are high. Advances in proteomic research have led to the discovery of an increasing amount of potential proteomic biomarkers and the identification of candidate biomarkers for clinical implementation in the context of kidney disease management. In this review that strictly follows the PRISMA guidelines, we focus on urinary peptide and especially peptidomic biomarkers emerging from recent research and underline the role of those with the highest potential for clinical implementation. The Web of Science database (all databases) was searched on 17 October 2022, using the search terms "marker *" OR biomarker * AND "renal disease" OR "kidney disease" AND "proteome *" OR "peptid *" AND "urin *". English, full-text, original articles on humans published within the last 5 years were included, which had been cited at least five times per year. Studies based on animal models, renal transplant studies, metabolite studies, studies on miRNA, and studies on exosomal vesicles were excluded, focusing on urinary peptide biomarkers. The described search led to the identification of 3668 articles and the application of inclusion and exclusion criteria, as well as abstract and consecutive full-text analyses of three independent authors to reach a final number of 62 studies for this manuscript. The 62 manuscripts encompassed eight established single peptide biomarkers and several proteomic classifiers, including CKD273 and IgAN237. This review provides a summary of the recent evidence on single peptide urinary biomarkers in CKD, while emphasizing the increasing role of proteomic biomarker research with new research on established and new proteomic biomarkers. Lessons learned from the last 5 years in this review might encourage future studies, hopefully resulting in the routine clinical applicability of new biomarkers.
Collapse
Affiliation(s)
- Lorenzo Catanese
- Department of Nephrology, Angiology and Rheumatology, Klinikum Bayreuth GmbH, 95447 Bayreuth, Germany
- Kuratorium for Dialysis and Transplantation (KfH), 95445 Bayreuth, Germany
- Medizincampus Oberfranken, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Justyna Siwy
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany
| | | | - Ralph Wendt
- Department of Nephrology, St. Georg Hospital Leipzig, 04129 Leipzig, Germany
| | - Joachim Beige
- Department of Nephrology, St. Georg Hospital Leipzig, 04129 Leipzig, Germany
- Department of Internal Medicine II, Martin-Luther-University Halle/Wittenberg, 06108 Halle/Saale, Germany
- Kuratorium for Dialysis and Transplantation (KfH), 04129 Leipzig, Germany
| | - Harald Rupprecht
- Department of Nephrology, Angiology and Rheumatology, Klinikum Bayreuth GmbH, 95447 Bayreuth, Germany
- Kuratorium for Dialysis and Transplantation (KfH), 95445 Bayreuth, Germany
- Medizincampus Oberfranken, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
35
|
Hu J, Zhou Y, Huang H, Kuai Y, Chen J, Bai Z, Li X, Li Y. Prediction of urinary dickkopf-3 for AKI, sepsis-associated AKI, and PICU mortality in children. Pediatr Res 2023; 93:1651-1658. [PMID: 36008594 DOI: 10.1038/s41390-022-02269-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Preoperative urinary dickkopf-3 (DKK3) is proposed as an early biomarker for the prediction of acute kidney injury (AKI) in patients undergoing cardiac surgery. We explored the clinical utility of urinary DKK3 for the early predictive value for AKI, sepsis-associated AKI (SA-AKI), and pediatric intensive care unit (PICU) mortality in critically ill children. METHODS Urine samples were collected during the first 24 h after admission for measurement of DKK3. AKI diagnosis was based on serum creatinine and urine output using the KDIGO criteria. SA-AKI was defined as AKI that occurred in children who met the sepsis criteria in accordance with the surviving sepsis campaign international guidelines for children. RESULTS Of the 420 children, 73 developed AKI, including 24 with SA-AKI, and 30 died during the PICU stay. The urinary DKK3 level was significantly associated with AKI, SA-AKI, and PICU mortality, even after adjustment for confounders. The area under the receiver operating characteristic curve of urinary DKK3 for the discrimination of AKI, SA-AKI, and PICU mortality was 0.70, 0.80, and 0.78, respectively. CONCLUSION Urinary DKK3 was independently associated with an increased risk for AKI, SA-AKI, and PICU mortality and may be predictive of the aforementioned issues in critically ill children. IMPACT Urinary dickkopf-3 (DKK3) has been identified as a preoperative biomarker for the prediction of acute kidney injury (AKI) following cardiac surgery or coronary angiography in adult patients. However, little is known about the clinical utility of urinary DKK3 in pediatric cohorts. This study demonstrated that urinary DKK3 is capable of early predicting AKI and pediatric intensive care unit (PICU) mortality and discriminating sepsis-associated AKI (SA-AKI) from other types of AKI. Urinary DKK3 may be an early biomarker for predicting AKI, SA-AKI, and PICU mortality in critically ill children.
Collapse
Affiliation(s)
- Junlong Hu
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Yueying Zhou
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Hui Huang
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Yuxian Kuai
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jiao Chen
- Pediatric Intensive Care Unit, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhenjiang Bai
- Pediatric Intensive Care Unit, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xiaozhong Li
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Yanhong Li
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China.
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China.
| |
Collapse
|
36
|
Speer T, Schunk SJ, Sarakpi T, Schmit D, Wagner M, Arnold L, Zewinger S, Azukaitis K, Bayazit A, Obrycki L, Kaplan Bulut I, Duzova A, Doyon A, Ranchin B, Caliskan S, Harambat J, Yilmaz A, Alpay H, Lugani F, Balat A, Arbeiter K, Longo G, Melk A, Querfeld U, Wühl E, Mehls O, Fliser D, Schaefer F. Urinary DKK3 as a biomarker for short-term kidney function decline in children with chronic kidney disease: an observational cohort study. THE LANCET. CHILD & ADOLESCENT HEALTH 2023; 7:405-414. [PMID: 37119829 DOI: 10.1016/s2352-4642(23)00049-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/04/2023] [Accepted: 02/09/2023] [Indexed: 05/01/2023]
Abstract
BACKGROUND Childhood-onset chronic kidney disease is a progressive condition that can have a major effect on life expectancy and quality. We evaluated the usefulness of the kidney tubular cell stress marker urinary Dickkopf-related protein 3 (DKK3) in determining the short-term risk of chronic kidney disease progression in children and identifying those who will benefit from specific nephroprotective interventions. METHODS In this observational cohort study, we assessed the association between urinary DKK3 and the combined kidney endpoint (ie, the composite of 50% reduction of the estimated glomerular filtration rate [eGFR] or progression to end-stage kidney disease) or the risk of kidney replacement therapy (ie, dialysis or transplantation), and the interaction of the combined kidney endpoint with intensified blood pressure reduction in the randomised controlled ESCAPE trial. Moreover, urinary DKK3 and eGFR were quantified in children aged 3-18 years with chronic kidney disease and urine samples available enrolled in the prospective multicentre ESCAPE (NCT00221845; derivation cohort) and 4C (NCT01046448; validation cohort) studies at baseline and at 6-monthly follow-up visits. Analyses were adjusted for age, sex, hypertension, systolic blood pressure SD score (SDS), BMI SDS, albuminuria, and eGFR. FINDINGS 659 children were included in the analysis (231 from ESCAPE and 428 from 4C), with 1173 half-year blocks in ESCAPE and 2762 in 4C. In both cohorts, urinary DKK3 above the median (ie, >1689 pg/mg creatinine) was associated with significantly greater 6-month eGFR decline than with urinary DKK3 at or below the median (-5·6% [95% CI -8·6 to -2·7] vs 1·0% [-1·9 to 3·9], p<0·0001, in ESCAPE; -6·2% [-7·3 to -5·0] vs -1·5% [-2·9 to -0·1], p<0·0001, in 4C), independently of diagnosis, eGFR, and albuminuria. In ESCAPE, the beneficial effect of intensified blood pressure control was limited to children with urinary DKK3 higher than 1689 pg/mg creatinine, in terms of the combined kidney endpoint (HR 0·27 [95% CI 0·14 to 0·55], p=0·0003, number needed to treat 4·0 [95% CI 3·7 to 4·4] vs 250·0 [66·9 to ∞]) and the need for kidney replacement therapy (HR 0·33 [0·13 to 0·85], p=0·021, number needed to treat 6·7 [6·1 to 7·2] vs 31·0 [27·4 to 35·9]). In 4C, inhibition of the renin-angiotensin-aldosterone system resulted in significantly lower urinary DKK3 concentrations (least-squares mean 12 235 pg/mg creatinine [95% CI 10 036 to 14 433] in patients not on angiotensin-converting enzyme inhibitors or angiotensin 2 receptor blockers vs 6861 pg/mg creatinine [5616 to 8106] in those taking angiotensin-converting enzyme inhibitors or angiotensin 2 receptor blockers, p<0·0001). INTERPRETATION Urinary DKK3 indicates short-term risk of declining kidney function in children with chronic kidney disease and might allow a personalised medicine approach by identifying those who benefit from pharmacological nephroprotection, such as intensified blood pressure lowering. FUNDING None.
Collapse
Affiliation(s)
- Thimoteus Speer
- Department of Internal Medicine 4, Nephrology, Goethe-University, Frankfurt, Germany; Else Kroener Fresenius Center for Nephrological Research, Goethe-University, Frankfurt, Germany
| | - Stefan J Schunk
- Department of Internal Medicine IV, Saarland University Medical Center, Homburg/Saar, Germany
| | - Tamim Sarakpi
- Department of Internal Medicine 4, Nephrology, Goethe-University, Frankfurt, Germany
| | - David Schmit
- Department of Internal Medicine IV, Saarland University Medical Center, Homburg/Saar, Germany
| | - Martina Wagner
- Department of Internal Medicine IV, Saarland University Medical Center, Homburg/Saar, Germany
| | - Ludger Arnold
- Department of Internal Medicine IV, Saarland University Medical Center, Homburg/Saar, Germany
| | - Stephen Zewinger
- Department of Internal Medicine IV, Saarland University Medical Center, Homburg/Saar, Germany
| | - Karolis Azukaitis
- Clinic of Pediatrics, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Aysun Bayazit
- Department of Pediatric Nephrology, Cukurova University, Adana, Turkey
| | - Lukasz Obrycki
- Department of Nephrology, Kidney Transplantation and Hypertension, The Children's Memorial Health Institute, Warsaw, Poland
| | - Ipek Kaplan Bulut
- Department of Pediatrics, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Ali Duzova
- Division of Pediatric Nephrology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Anke Doyon
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Bruno Ranchin
- Pediatric Nephrology Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Université de Lyon, Lyon, France
| | - Salim Caliskan
- Department of Pediatric Nephrology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Jerome Harambat
- Pediatrics Department, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Alev Yilmaz
- Pediatric Nephrology, Istanbul Medical Faculty, Istanbul, Turkey
| | - Harika Alpay
- Department of Pediatrics, School of Medicine, Marmara University, Istanbul, Turkey
| | - Francesca Lugani
- Pediatric Nephrology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Ayse Balat
- Department of Pediatric Nephrology, Gaziantep University, Gaziantep, Turkey
| | - Klaus Arbeiter
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Germana Longo
- Pediatric Nephrology, Dialysis and Transplant Unit, Department of Woman and Child Health, Azienda Ospedaliera-University of Padova, Padova, Italy
| | - Anette Melk
- Department of Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Uwe Querfeld
- Department of Pediatrics, Division of Gastroenterology, Nephrology and Metabolic Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Elke Wühl
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Otto Mehls
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Danilo Fliser
- Department of Internal Medicine IV, Saarland University Medical Center, Homburg/Saar, Germany; DiaRen, Homburg/Saar, Germany
| | - Franz Schaefer
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
37
|
Satrapova V, Sparding N, Genovese F, Karsdal MA, Bartonova L, Frausova D, Honsova E, Kollar M, Suchanek M, Koprivova H, Rysava R, Bednarova V, Tesar V, Hruskova Z. Biomarkers of fibrosis, kidney tissue injury and inflammation may predict severity and outcome of renal ANCA – associated vasculitis. Front Immunol 2023; 14:1122972. [PMID: 37020541 PMCID: PMC10067901 DOI: 10.3389/fimmu.2023.1122972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/24/2023] [Indexed: 03/22/2023] Open
Abstract
BackgroundActivity and chronicity of kidney involvement in ANCA-associated vasculitis (AAV) can be currently reliably evaluated only by kidney biopsy. In this study, we measured a panel of serum and urinary biomarkers collected at the time of kidney biopsy and hypothesized that they could reflect specific histopathological parameters in the biopsy and help to predict prognosis.MethodsWe examined a cohort of 45 patients with AAV and 10 healthy controls. Biomarker levels (DKK-3, CD163, EGF, PRO-C6 and C3M) were measured in this study by ELISA. Biopsies were scored with a scoring system for AAV (focal x crescentic x sclerotic x mixed class) and interstitial fibrosis was quantified.ResultsLevels of urinary DKK-3, CD163, EGF, PRO-C6 and C3M significantly differed among biopsy classes in AAV, with urinary DKK-3 and PRO-C6 levels being highest in the sclerotic class and lowest in the focal class, urinary CD163 levels highest in the crescentic class and urinary C3M levels highest in the focal class. Moreover, the urinary biomarkers were able to discriminate focal biopsy class from the other classes. Urinary DKK-3, EGF, PRO-C6 and C3M levels measured at the time of biopsy were also significantly related to the extent of fibrosis and to the final kidney function at the end of follow-up.ConclusionsThis small pilot study suggests that selected urinary biomarkers of fibrosis and inflammation may reflect changes in the kidney biopsy and be prognostic of kidney outcome in patients with AAV.
Collapse
Affiliation(s)
- Veronika Satrapova
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
- *Correspondence: Veronika Satrapova,
| | | | | | | | - Lenka Bartonova
- Department of Pathology, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Doubravka Frausova
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Eva Honsova
- Department of Pathology, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Marek Kollar
- Department of Pathology, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Miloslav Suchanek
- Faculty of Environment, Jan Evangelista Purkyně University in Ústí nad Labem, Ústí nad Labem, Czechia
| | - Helena Koprivova
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Romana Rysava
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Vladimira Bednarova
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Vladimir Tesar
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Zdenka Hruskova
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| |
Collapse
|
38
|
Huang R, Fu P, Ma L. Kidney fibrosis: from mechanisms to therapeutic medicines. Signal Transduct Target Ther 2023; 8:129. [PMID: 36932062 PMCID: PMC10023808 DOI: 10.1038/s41392-023-01379-7] [Citation(s) in RCA: 243] [Impact Index Per Article: 121.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 02/12/2023] [Accepted: 02/20/2023] [Indexed: 03/19/2023] Open
Abstract
Chronic kidney disease (CKD) is estimated to affect 10-14% of global population. Kidney fibrosis, characterized by excessive extracellular matrix deposition leading to scarring, is a hallmark manifestation in different progressive CKD; However, at present no antifibrotic therapies against CKD exist. Kidney fibrosis is identified by tubule atrophy, interstitial chronic inflammation and fibrogenesis, glomerulosclerosis, and vascular rarefaction. Fibrotic niche, where organ fibrosis initiates, is a complex interplay between injured parenchyma (like tubular cells) and multiple non-parenchymal cell lineages (immune and mesenchymal cells) located spatially within scarring areas. Although the mechanisms of kidney fibrosis are complicated due to the kinds of cells involved, with the help of single-cell technology, many key questions have been explored, such as what kind of renal tubules are profibrotic, where myofibroblasts originate, which immune cells are involved, and how cells communicate with each other. In addition, genetics and epigenetics are deeper mechanisms that regulate kidney fibrosis. And the reversible nature of epigenetic changes including DNA methylation, RNA interference, and chromatin remodeling, gives an opportunity to stop or reverse kidney fibrosis by therapeutic strategies. More marketed (e.g., RAS blockage, SGLT2 inhibitors) have been developed to delay CKD progression in recent years. Furthermore, a better understanding of renal fibrosis is also favored to discover biomarkers of fibrotic injury. In the review, we update recent advances in the mechanism of renal fibrosis and summarize novel biomarkers and antifibrotic treatment for CKD.
Collapse
Affiliation(s)
- Rongshuang Huang
- Kidney Research Institute, Division of Nephrology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ping Fu
- Kidney Research Institute, Division of Nephrology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Liang Ma
- Kidney Research Institute, Division of Nephrology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
39
|
Rodrigues CE, Endre ZH. Definitions, phenotypes, and subphenotypes in acute kidney injury-Moving towards precision medicine. Nephrology (Carlton) 2023; 28:83-96. [PMID: 36370326 PMCID: PMC10100386 DOI: 10.1111/nep.14132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 10/23/2022] [Accepted: 10/31/2022] [Indexed: 11/13/2022]
Abstract
The current definition of acute kidney injury (AKI) is generic and, based only on markers of function, is unsuitable for guiding individualized treatment. AKI is a complex syndrome with multiple presentations and causes. Targeted AKI management will only be possible if different phenotypes and subphenotypes of AKI are recognised, based on causation and related pathophysiology. Molecular signatures to identify subphenotypes are being recognised, as specific biomarkers reveal activated pathways. Assessment of individual clinical risk needs wider dissemination to allow identification of patients at high risk of AKI. New and more timely markers for glomerular filtration rate (GFR) are available. However, AKI diagnosis and classification should not be limited to GFR, but include tubular function and damage. Combining damage and stress biomarkers with functional markers enhances risk prediction, and identifies a population enriched for clinical trials targeting AKI. We review novel developments and aim to encourage implementation of these new techniques into clinical practice as a strategy for individualizing AKI treatment akin to a precision medicine-based approach.
Collapse
Affiliation(s)
- Camila Eleuterio Rodrigues
- Nephrology DepartmentPrince of Wales Clinical School – UNSW MedicineSydneyNew South WalesAustralia
- Nephrology DepartmentHospital das Clínicas – University of São Paulo School of MedicineSão PauloBrazil
| | - Zoltán H. Endre
- Nephrology DepartmentPrince of Wales Clinical School – UNSW MedicineSydneyNew South WalesAustralia
| |
Collapse
|
40
|
Urinary Dickkopf-3 (DKK3) Is Associated with Greater eGFR Loss in Patients with Resistant Hypertension. J Clin Med 2023; 12:jcm12031034. [PMID: 36769689 PMCID: PMC9918035 DOI: 10.3390/jcm12031034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/18/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Patients with resistant hypertension (HTN) demonstrate an increased risk of chronic kidney disease and progression to end-stage renal disease; however, the individual course of progression is hard to predict. Assessing the stress-induced, urinary glycoprotein Dickkopf-3 (uDKK3) may indicate ongoing renal damage and consecutive estimated glomerular filtration rate (eGFR) decline. The present study aimed to determine the association between uDKK3 levels and further eGFR changes in patients with resistant HTN. In total, 31 patients with resistant HTN were included. Blood pressure and renal function were measured at baseline and up to 24 months after (at months 12 and 24). uDKK3 levels were determined exclusively from the first available spot urine sample at baseline or up to a period of 6 months after, using a commercial ELISA kit. Distinctions between different patient groups were analyzed using the unpaired t-test or Mann-Whitney test. Correlation analysis was performed using Spearman's correlation. The median uDKK3 level was 303 (interquartile range (IQR) 150-865) pg/mg creatinine. Patients were divided into those with high and low eGFR loss (≥3 vs. <3 mL/min/1.73 m²/year). Patients with high eGFR loss showed a significantly higher median baseline uDKK3 level (646 (IQR 249-2555) (n = 13) vs. 180 (IQR 123-365) pg/mg creatinine (n = 18), p = 0.0412 (Mann-Whitney U)). Alternatively, patients could be classified into those with high and low uDKK3 levels (≥400 vs. <400 pg/mg creatinine). Patients with high uDKK3 levels showed significantly higher eGFR loss (-6.4 ± 4.7 (n = 11) vs. 0.0 ± 7.6 mL/min/1.73 m2/year (n = 20), p = 0.0172 (2-sided, independent t-test)). Within the entire cohort, there was a significant correlation between the uDKK3 levels and change in eGFR at the latest follow-up (Spearman's r = -0.3714, p = 0.0397). In patients with resistant HTN, high levels of uDKK3 are associated with higher eGFR loss up to 24 months later.
Collapse
|
41
|
Du Y, Cheng T, Liu C, Zhu T, Guo C, Li S, Rao X, Li J. IgA Nephropathy: Current Understanding and Perspectives on Pathogenesis and Targeted Treatment. Diagnostics (Basel) 2023; 13:diagnostics13020303. [PMID: 36673113 PMCID: PMC9857562 DOI: 10.3390/diagnostics13020303] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
Immunoglobulin A nephropathy (IgAN) is the most common primary glomerulonephritis worldwide, with varied clinical and histopathological features between individuals, particularly across races. As an autoimmune disease, IgAN arises from consequences of increased circulating levels of galactose-deficient IgA1 and mesangial deposition of IgA-containing immune complexes, which are recognized as key events in the widely accepted "multi-hit" pathogenesis of IgAN. The emerging evidence further provides insights into the role of genes, environment, mucosal immunity and complement system. These developments are paralleled by the increasing availability of diagnostic tools, potential biomarkers and therapeutic agents. In this review, we summarize current evidence and outline novel findings in the prognosis, clinical trials and translational research from the updated perspectives of IgAN pathogenesis.
Collapse
|
42
|
[Nephrological management and drug dosing in patients with rheumatic diseases and renal insufficiency]. Z Rheumatol 2022; 81:811-828. [PMID: 36350405 DOI: 10.1007/s00393-022-01283-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND In patients with inflammatory rheumatic diseases and renal insufficiency there are two challenges for physicians: to adapt the antirheumatic medication to the renal function and to carry out a nephroprotective treatment that prevents long-term deterioration of renal function and reduces the elevated cardiovascular risk. METHODS A literature search (in PubMed) was carried out and the current state of knowledge on nephroprotective treatment strategies and the treatment of rheumatic diseases in the presence of renal insufficiency was collated, evaluated and summarized. RESULTS Lifestyle interventions, especially the cessation of smoking and drug treatment strategies form the basis of nephroprotection including the control of diabetes mellitus with metformin, sodium glucose transporter 2 (SGLT2) inhibitors, glucagon-like peptide 1 (GLP1) analogues and control of hypertension with blockade of the renin-angiotensin-aldosterone system (RAAS), hyperlipidemia, hyperphosphatemia and metabolic acidosis. The SGLT2 inhibitors are also effective for nondiabetic nephropathy. The elevated cardiovascular risk is further reduced by effective control of inflammatory rheumatic activity. Numerous conventional disease modifying antirheumatic drugs, especially methotrexate and the Janus kinase (JAK) inhibitors baricitinib and filgotinib, must mostly be adapted to the renal function. In contrast, biologics can be given in standard doses with the exception of anakinra. The increased cardiovascular risk currently limits the use of tofacitinib in patients with renal insufficiency. CONCLUSION The antirheumatic medication should be modified and a complex nephroprotective treatment concept is mandatory in the management of patients with rheumatic disease and renal insufficiency, that in the best-case scenario can be guaranteed by a close interdisciplinary cooperation of rheumatologists and nephrologists.
Collapse
|
43
|
Kramer T, Brinkkoetter P, Rosenkranz S. Right Heart Function in Cardiorenal Syndrome. Curr Heart Fail Rep 2022; 19:386-399. [PMID: 36166185 PMCID: PMC9653308 DOI: 10.1007/s11897-022-00574-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/12/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE OF REVIEW Since CRS is critically dependent on right heart function and involved in interorgan crosstalk, assessment and monitoring of both right heart and kidney function are of utmost importance for clinical outcomes. This systematic review aims to comprehensively report on novel diagnostic and therapeutic paradigms that are gaining importance for the clinical management of the growing heart failure population suffering from CRS. RECENT FINDINGS Cardiorenal syndrome (CRS) in patients with heart failure is associated with poor outcome. Although systemic venous congestion and elevated central venous pressure have been recognized as main contributors to CRS, they are often neglected in clinical practice. The delicate hemodynamic balance in CRS is particularly determined by the respective status of the right heart. The consideration of hemodynamic and CRS profiles is advantageous in tailoring treatment for better preservation of renal function. Assessment and monitoring of right heart and renal function by known and emerging tools like renal Doppler ultrasonography or new biomarkers may have direct clinical implications.
Collapse
Affiliation(s)
- Tilmann Kramer
- Klinik III Für Innere Medizin, Herzzentrum Der Universität Zu Köln, Köln, Germany.
- Cologne Cardiovascular Research Center (CCRC), Klinikum Der Universität Zu Köln, Köln, Germany.
| | - Paul Brinkkoetter
- Cologne Cardiovascular Research Center (CCRC), Klinikum Der Universität Zu Köln, Köln, Germany
- Klinik II Für Innere Medizin, Nephrologie, Universität Zu Köln, Köln, Germany
- Center for Molecular Medicine Cologne (CMMC), Universität Zu Köln, Köln, Germany
| | - Stephan Rosenkranz
- Klinik III Für Innere Medizin, Herzzentrum Der Universität Zu Köln, Köln, Germany
- Cologne Cardiovascular Research Center (CCRC), Klinikum Der Universität Zu Köln, Köln, Germany
- Center for Molecular Medicine Cologne (CMMC), Universität Zu Köln, Köln, Germany
| |
Collapse
|
44
|
Cottam D, Azzopardi G, Forni LG. Biomarkers for early detection and predicting outcomes in acute kidney injury. Br J Hosp Med (Lond) 2022; 83:1-11. [DOI: 10.12968/hmed.2022.0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The current diagnosis of acute kidney injury relies on the measurement of serum creatinine levels and urine output. However, both measures are subject to considerable limitations; for example, change in serum creatinine levels ideally requires a knowledge of baseline function that is often not available. Furthermore, creatinine levels are influenced by many factors including diet, drug therapy, muscle mass, gender and ethnicity, which may lead to underestimation of the extent of renal dysfunction. Similarly, urine output lacks both specificity and sensitivity as a marker of acute kidney injury given that oliguria may be an appropriate physiological response to a multitude of stressors and that output may be maintained until significant renal damage has already occurred. Given the well-documented consequences of acute kidney injury and the considerable burden associated with its development, much attention has focused on early identification of patients at high risk to try and improve outcomes. Many studies have focused on the identification of candidate molecules that may enable the early detection of individuals at risk of developing acute kidney injury, including constitutive proteins associated with kidney damage, as well as molecules upregulated in response to injury, non-renal products that may be filtered, reabsorbed or secreted by the kidney, and markers of renal stress. Such biomarkers may also aid stratification for adverse events, such as the need for kidney replacement therapy or progression to chronic kidney disease and end-stage kidney disease. This article discusses some of these novel biomarkers and assesses the role they may have in the understanding, management, diagnosis and prognostication of acute kidney injury.
Collapse
Affiliation(s)
- Daniel Cottam
- Intensive Care Unit, Royal Surrey Hospital Foundation Trust, Guildford, UK
| | - Giada Azzopardi
- South West Thames Renal and Transplantation Unit, Epsom and St Helier University Hospitals, Carshalton, UK
| | - Lui G Forni
- Intensive Care Unit, Royal Surrey Hospital Foundation Trust, Guildford, UK
- Department of Clinical and Experimental Medicine, Faculty of Health Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
45
|
Husain-Syed F, Reis T, Kashani K, Ronco C. Advances in laboratory detection of acute kidney injury. Pract Lab Med 2022; 31:e00283. [PMID: 35677313 PMCID: PMC9168173 DOI: 10.1016/j.plabm.2022.e00283] [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: 01/03/2022] [Revised: 04/27/2022] [Accepted: 05/24/2022] [Indexed: 12/14/2022] Open
Abstract
Recent advances have improved our understanding of the epidemiology and pathophysiology of acute kidney injury (AKI). So far, the Kidney Disease: Improving Global Outcome guidelines define and stratify kidney injury based on increases in serum creatinine level and/or decreases in urine output. Although the term AKI acknowledges the existence of cellular injury, its diagnosis is still only defined by the reduced excretory function of the kidney. New biomarkers that aid a better understanding of the relationship between acute tubular injury and kidney dysfunction have been identified, reflecting the advances in molecular biology. The expression of some of these novel biomarkers precedes changes in conventional biomarkers or can increase their predictive power. Therefore, they might enhance the clinical accuracy of the definition of AKI. This review summarizes the limitations of the current AKI classification and a panel of candidate biomarkers for augmenting AKI classification and recognition of AKI subphenotypes. We expect that the integration of appropriately selected biomarkers in routine clinical practice can improve AKI care.
Collapse
Affiliation(s)
- Faeq Husain-Syed
- Department of Internal Medicine II, University Hospital Giessen and Marburg, Justus-Liebig-University Giessen, Klinikstraße 33, 35392, Giessen, Germany
| | - Thiago Reis
- Laboratory of Molecular Pharmacology, Faculty of Health Sciences, University of Brasília, Brasília, Distrito Federal, Brazil
- Department of Nephrology and Kidney Transplantation, Clínica de Doenças Renais de Brasília, DF Star Hospital, Rede D'Or São Luiz, Brasília, Distrito Federal, Brazil
| | - Kianoush Kashani
- Division of Nephrology and Hypertension, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Claudio Ronco
- Department of Medicine (DIMED), Università di Padova, Via Giustiniani, 2–35128, Padua, Italy
- International Renal Research Institute of Vicenza, Via Rodolfi, 37–36100, Vicenza, Italy
- Department of Nephrology, Dialysis and Transplantation, San Bortolo Hospital, Via Rodolfi, 37–36100, Vicenza, Italy
| |
Collapse
|
46
|
Biochemical interaction of pyrvinium in gentamicin-induced acute kidney injury by modulating calcium dyshomeostasis and mitochondrial dysfunction. Chem Biol Interact 2022; 363:110020. [PMID: 35750223 DOI: 10.1016/j.cbi.2022.110020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 06/03/2022] [Accepted: 06/15/2022] [Indexed: 11/21/2022]
Abstract
Acute kidney injury (AKI) has a poor clinical prognosis and increases the risk of chronic kidney failure (CKD). It is a common complication of organ failure in hospitalised patients (10-15% of all hospitalizations) and in intensive care unit (ICU) patients, with an incidence of up to 50%. Concerning ICU, AKI has a mortality rate ranging from 27% to 35%, rising to 60%-65% when dialysis is needed, with roughly 5%-20% of survivors requiring dialysis on discharge. AKI is believed to cause over 7 million deaths per year worldwide. Currently, there is no treatment for AKI or its progression to CKD. When activated by AKI, numerous pathways have been suggested as possible contributors to CKD progression. Wnt/β-catenin is a crucial regulator of kidney development that increases following the injury. Despite the overwhelming evidence that Wnt/β-catenin promotes AKI, tubulointerstitial fibrosis, a hallmark of CKD progression, is also promoted by this pathway. The therapeutic potential of Wnt/β-catenin in the treatment of AKI and the progression from AKI to CKD is being studied. This hypothesis aims to determine whether the Wnt/β-catenin inhibitor pyrvinium has a beneficial effect on the renal dysfunction and damage caused by Gentamicin.
Collapse
|
47
|
Schuster A, Steines L, Müller K, Zeman F, Findeisen P, Banas B, Bergler T. Dickkopf 3-A New Indicator for the Deterioration of Allograft Function After Kidney Transplantation. Front Med (Lausanne) 2022; 9:885018. [PMID: 35646976 PMCID: PMC9130628 DOI: 10.3389/fmed.2022.885018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/19/2022] [Indexed: 11/20/2022] Open
Abstract
Evidence of tubular atrophy and interstitial fibrosis is prognostically unfavorable and associated with a premature graft loss after kidney transplantation. Recently, Dickkopf 3 (DKK3), a profibrotic glycoprotein released by stressed tubular epithelial cells, has been identified to cause IF/TA by regulating the Wnt/β-catenin signaling and seems to engage a T-cell response. The aim of our study was to determine if a correlation between DKK3 and graft function exists and if DKK3 could be a new indicator to identify patients at risk for a deterioration in graft function. Patients, transplanted between 2016 and 2018, were analyzed with regard to DKK3 in the urine and graft function (creatinine, eGFR, albuminuria). Multivariable analyzes were used including known factors influencing graft function (PRA, donor age) to stress robustness of DKK3. The 3 and 12 month DKK3 values were significant predictors for subsequent graft function up to 36 months. An increase of DKK3 from month 3 to 12 of ≥ 25% showed a higher risk of an impaired graft function, with, e.g., a reduction in eGFR of about 9–10 ml/min in contrast to patients without intensified DKK3 increase. Induction therapy has an influence on DKK3 as patients induced with a T-cell depleting therapy showed a trend toward lower DKK3 values. In summary, our study is the first investigation of DKK3 in kidney transplant recipients and was able to show that DKK3 could forecast graft function. It is recommended to investigate the potential of DKK3 as a predictor of kidney function after transplantation in further studies.
Collapse
Affiliation(s)
- Antonia Schuster
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Louisa Steines
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Karolina Müller
- Center for Clinical Studies, University Hospital Regensburg, Regensburg, Germany
| | - Florian Zeman
- Center for Clinical Studies, University Hospital Regensburg, Regensburg, Germany
| | | | - Bernhard Banas
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Tobias Bergler
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
48
|
Sparding N, Genovese F, Rasmussen DGK, Karsdal MA, Neprasova M, Maixnerova D, Satrapova V, Frausova D, Hornum M, Bartonova L, Honsova E, Kollar M, Koprivova H, Hruskova Z, Tesar V. Endotrophin, a collagen type VI-derived matrikine, reflects the degree of renal fibrosis in patients with IgA nephropathy and in patients with ANCA-associated vasculitis. Nephrol Dial Transplant 2022; 37:1099-1108. [PMID: 33914059 PMCID: PMC9130028 DOI: 10.1093/ndt/gfab163] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Renal fibrosis is the hallmark of chronic kidney disease (CKD) and is characterized by an imbalanced extracellular matrix remodelling. Endotrophin (ETP) is a signalling molecule released from collagen type VI (COL VI). ETP can be measured by the PRO-C6 assay, which quantifies the levels of COL VI formation. ETP levels were previously associated with mortality and disease progression in patients with CKD. We hypothesized that serum and urinary ETP levels correlate with the degree of interstitial fibrosis in kidney biopsies from patients with immunoglobulin A nephropathy (IgAN) and patients with anti-neutrophil cytoplasmic antibody-associated vasculitis (AAV). METHODS We examined a cohort of 49 IgAN and 47 AAV patients. A validation cohort of 85 IgAN patients was included. ETP was measured in serum (S-ETP) and urine (U-ETP/Cr) samples, taken on the same day before renal biopsy was performed, using the enzyme-linked immunosorbent assay PRO-C6. The biopsies were evaluated for interstitial fibrosis and tubular atrophy according to the Banff and MEST-C scores. RESULTS S-ETP and U-ETP/Cr levels correlated with kidney function, increased CKD severity, correlated with the extent of interstitial fibrosis and gradually increased with increasing degree of interstitial fibrosis and tubular atrophy. ETP outperformed the known fibrosis biomarker Dickkopf-3 for discrimination of patients with high fibrotic burden. The association of S-ETP and U-ETP/Cr with the level of kidney fibrosis was confirmed in the validation cohort. CONCLUSIONS We demonstrated that high levels of circulating and excreted ETP are not only indicative of lower kidney function, but also reflect the burden of fibrosis in the kidneys.
Collapse
Affiliation(s)
- Nadja Sparding
- Nordic Bioscience, Herlev, Denmark
- Faculty of Health and Medical Science, Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Michaela Neprasova
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Dita Maixnerova
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Veronika Satrapova
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Doubravka Frausova
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Mads Hornum
- Department of Nephrology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Lenka Bartonova
- Department of Pathology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Eva Honsova
- Department of Pathology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Marek Kollar
- Department of Pathology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Helena Koprivova
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Zdenka Hruskova
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Vladimir Tesar
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| |
Collapse
|
49
|
Gonzalez J, Jatem E, Roig J, Valtierra N, Ostos E, Abo A, Santacana M, Garcia A, Segarra A. Usefulness of urinary biomarkers to estimate the interstitial fibrosis surface in diabetic nephropathy with normal kidney function. Nephrol Dial Transplant 2022; 37:2102-2110. [PMID: 35583251 DOI: 10.1093/ndt/gfac185] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Kidney biopsies of patients with diabetic nephropathy (DN) and normal kidney function may exhibit interstitial fibrosis (IF) without reduction of glomerular filtration rate because of hyperfiltration. The aim of our study was to analyze the performance of a set of biomarkers of tubular injury to estimate the extent of IF in patients with DN and normal kidney function. PATIENTS AND METHODS This cross-sectional study included 118 adults with DN diagnosed by kidney biopsy and GFR ≥ 90 mL/min/1.73 m2 and a control group of healthy subjects. We measured the urinary excretion of MCP-1, NGAL, KIM-1, L-FABP, β2-microglobulin and DKK-3 at the time of kidney biopsy. GFR was measured by Cr-EDTA (mGFR). IF was quantified using a quantitative morphometric procedure. Predictive multivariate models were developed to estimate the IF surface. RESULTS patients with DN showed significantly higher levels of DKK-3, MCP-1 and L-FABP and significantly lower levels of EGF than healthy controls. There were no significant between-group differences in the levels of β2-microglobulin, KIM-1 or NGAL. IF was negatively associated with EGF and a positively with age, proteinuria, MCP-1, DKK-3 and L-FABP but no with β2-microglobulin, KIM-1, NGAL or GFR. The best model to predict IF surface accounted for 59% of its variability and included age, proteinuria, EGF, DKK-3 and MCP-1. CONCLUSIONS our study provides a model to estimate the IF in DN that can be useful to assess the progression of IF in patients with normal kidney function.
Collapse
Affiliation(s)
- Jorge Gonzalez
- Servicio de Nefrologia Hospital Arnau de Vilanova, Lleida
| | - Elias Jatem
- Servicio de Nefrologia Hospital Arnau de Vilanova, Lleida
| | - Jordi Roig
- Servicio de Nefrologia Hospital Arnau de Vilanova, Lleida
| | | | - Elena Ostos
- Vall d'Hebrón Institut de Recerca, Barcelona
| | - Anabel Abo
- Servicio de Anatomia Patológica, Hospital Arnau de Vilanova, Lleida
| | - Maria Santacana
- Servicio de Anatomia Patológica, Hospital Arnau de Vilanova, Lleida.,Institut de Recerca Biomèdica August Pi i Sunyer, Lleida
| | - Alicia Garcia
- Institut de Recerca Biomèdica August Pi i Sunyer, Lleida
| | - Alfons Segarra
- Servicio de Nefrologia Hospital Arnau de Vilanova, Lleida.,Vall d'Hebrón Institut de Recerca, Barcelona.,Institut de Recerca Biomèdica August Pi i Sunyer, Lleida.,Servicio de Nefrologia Hospital Vall d'Hebrón, Barcelona, Spain
| |
Collapse
|
50
|
Labes R, Brinkmann L, Kulow VA, Roegner K, Mathia S, Balcerek B, Persson PB, Rosenberger C, Fähling M. Daprodustat prevents Cyclosporine-A mediated anemia and peritubular capillary loss. Kidney Int 2022; 102:750-765. [DOI: 10.1016/j.kint.2022.04.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 04/06/2022] [Accepted: 04/18/2022] [Indexed: 12/24/2022]
|