1
|
Rao J, Li Y, Zhang X, Zhao W, Chen Y, Zhang J, Peng H, Ye Z. The prognostic value of the neutrophil-percentage-to-albumin ratio for all-cause and cardiovascular mortality in chronic kidney disease stages G3a to G5: insights from NHANES 2003-2018. Ren Fail 2025; 47:2495861. [PMID: 40336184 PMCID: PMC12064118 DOI: 10.1080/0886022x.2025.2495861] [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: 09/22/2024] [Revised: 01/30/2025] [Accepted: 04/05/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Patients with chronic kidney disease (CKD) stages G3a to G5 frequently experience heightened systemic inflammation and nutritional loss. Identifying laboratory-accessible, cost-effective markers that can effectively predict the prognosis of CKD stages G3a to G5 is crucial. METHODS This prospective cohort study included 3,331 patients with CKD stages G3a to G5 who participated in the National Health and Nutrition Examination Survey (NHANES) from 2003 to 2018. Multivariable adjusted Cox proportional hazards regression models and restricted cubic spline analyses were used to assess the associations of neutrophil percentage-to-albumin ratio (NPAR) levels with all-cause mortality, CVD, and non-CVD mortality. RESULTS The cohort study encompassed data from 3,331 participants for analysis. Nonlinear J-shaped associations were observed between NPAR levels and the risk of all-cause, CVD, and non-CVD mortality in patients with CKD stages G3a to G5. High levels NPAR exhibited a significantly elevated risk of both all-cause and CVD mortality in the fully adjusted model. The respective hazard ratios (HRs) for all-cause mortality were 1.23 [95% confidence interval (CI), 1.05-1.44], and for CVD mortality, 1.513 (95% CI, 1.131-2.024). CONCLUSIONS Elevated NPAR can predict both all-cause and CVD deaths in advanced CKD patients. Individuals with high NPAR levels face an elevated risk of mortality and exhibit a decreased survival rate in the context of CKD. This finding offers evidence supporting the timely evaluation and intervention for inflammation and nutritional status in individuals with CKD stages G3a to G5.
Collapse
Affiliation(s)
- Jialing Rao
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yuanqing Li
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaohao Zhang
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wenbo Zhao
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yanru Chen
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jun Zhang
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hui Peng
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zengchun Ye
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
2
|
Wang JN, Suo XG, Yu JT, Luo QC, Ji ML, Zhang MM, Zhu Q, Cheng XR, Hou C, Chen X, Wang F, Xu CH, Li C, Xie SS, Wei J, Zhang DF, Zhang XR, Wang ZJ, Dong YH, Zhu S, Peng LJ, Li XY, Chen HY, Xu T, Jin J, Chen FX, Meng XM. NAT10 exacerbates acute renal inflammation by enhancing N4-acetylcytidine modification of the CCL2/CXCL1 axis. Proc Natl Acad Sci U S A 2025; 122:e2418409122. [PMID: 40261924 PMCID: PMC12054813 DOI: 10.1073/pnas.2418409122] [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: 09/12/2024] [Accepted: 03/08/2025] [Indexed: 04/24/2025] Open
Abstract
Inflammation plays an essential role in eliminating microbial pathogens and repairing tissues, while sustained inflammation accelerates kidney damage and disease progression. Therefore, understanding the mechanisms of the inflammatory response is vital for developing therapies for inflammatory kidney diseases like acute kidney injury (AKI), which currently lacks effective treatment. Here, we identified N-acetyltransferase 10 (NAT10) as an important regulator for acute inflammation. NAT10, the only known "writer" protein for N4-acetylcytidine (ac4C) acetylation, is elevated in renal tubules across various AKI models, human biopsies, and cultured tubular epithelial cells (TECs). Conditional knockout (cKO) of NAT10 in mouse kidneys attenuates renal dysfunction, inflammation, and infiltration of macrophages and neutrophils, whereas its conditional knock-in (cKI) exacerbates these effects. Mechanistically, our findings from ac4C-RIP-seq and RNA-seq analyses revealed that NAT10-mediated ac4C acetylation enhances the mRNA stability of a range of key chemokines, including C-C motif chemokine ligand 2 (CCL2) and C-X-C motif chemokine ligand 1(CXCL1), promoting macrophage and neutrophil recruitment and accelerating renal inflammation. Additionally, CCL2 and CXCL1 neutralizing antibodies or their receptor inhibitors, abrogated renal inflammation in NAT10-overexpression TECs or NAT10-cKI mice. Importantly, inhibiting NAT10, either through Adeno-associated virus 9 (AAV9)-mediated silencing or pharmacologically with our found inhibitor Cpd-155, significantly reduces renal inflammation and injury. Thus, targeting the NAT10/CCL2/CXCL1 axis presents a promising therapeutic strategy for treating inflammatory kidney diseases.
Collapse
Affiliation(s)
- Jia-nan Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Xiao-guo Suo
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Ju-tao Yu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Qi-chao Luo
- School of Basic Medicine, Anhui Medical University, Hefei230032, China
| | - Ming-lu Ji
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Meng-meng Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Qi Zhu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Xin-ran Cheng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Chao Hou
- School of Basic Medicine, Anhui Medical University, Hefei230032, China
| | - Xin Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Fang Wang
- Department of Pharmacy, Lu’an Hospital of Anhui Medical University, Lu’an People’s Hospital of Anhui Province, Lu’an237006, China
| | - Chuan-hui Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Chao Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Shuai-shuai Xie
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Jie Wei
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei230601, Anhui, China
| | - Dan-feng Zhang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei230601, Anhui, China
| | - Xin-ru Zhang
- School of Basic Medicine, Anhui Medical University, Hefei230032, China
| | - Zhi-juan Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei230601, Anhui, China
| | - Yu-hang Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Sai Zhu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Li-jin Peng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Xiang-yu Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Hai-yong Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong999077, Hong Kong
- Department of Chinese Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen518000, China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Juan Jin
- School of Basic Medicine, Anhui Medical University, Hefei230032, China
| | - Fei Xavier Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Xiao-ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| |
Collapse
|
3
|
Anderson WA, Domingo-Relloso A, Galvez-Fernandez M, Schilling K, Glabonjat RA, Basu A, Nigra AE, Gutierrez OM, Scherzer R, Goldsmith J, Sarnak MJ, Bonventre JV, Kimmel PL, Vasan RS, Ix JH, Shlipak MG, Navas-Acien A. Uranium exposure and kidney tubule biomarkers in the Multi-Ethnic Study of Atherosclerosis (MESA). ENVIRONMENTAL RESEARCH 2025; 271:121060. [PMID: 39922262 PMCID: PMC11959630 DOI: 10.1016/j.envres.2025.121060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND Experimental studies indicate that uranium exposure is toxic to the kidney tubules. We evaluated the association of urinary uranium concentrations with biomarkers of tubule cell dysfunction (alpha-1-microglobulin [A1M], uromodulin [UMOD], epidermal growth factor [EGF]), and tubule cell injury (kidney injury molecule-1 [KIM-1], monocyte chemoattractant protein-1 [MCP-1], and chitinase-3-like protein 1 [YKL-40]), as well as with albuminuria and estimated glomerular filtration rate (eGFR) among participants in the Multi-Ethnic Study of Atherosclerosis (MESA). METHODS We conducted a cross-sectional study that included 461 participants selected for the absence of diabetes, chronic kidney disease (CKD), and cardiovascular disease, evaluated with six kidney tubule biomarker measurements. Urinary uranium concentrations were measured using inductively coupled plasma mass spectrometry in spot urine samples. Linear models were used to determine associations of urinary uranium concentrations with each kidney tubule biomarker, calculated by the geometric mean ratio (GMR), after adjustment for participant's urinary creatinine concentrations, age, sex, race/ethnicity, MESA field center, highest level of education completed, cigarette smoking status, alcohol consumption, body mass index (BMI), albuminuria levels, and eGFR. RESULTS Median (interquartile range) urinary uranium concentration was 5.2 (2.9, 10.4) ng/L, and mean (standard deviation) eGFR was 99 (16) mL/min/1.73 m2. The adjusted GMRs (95%CI) of KIM-1 and MCP-1 were 1.11 (1.01, 1.22) and 1.10 (1.01, 1.20), respectively per 7.5 ng/L (interquartile range) higher urinary uranium concentration, while no statistically significant associations were observed for YKL-40, A1M, UMOD, EGF, albuminuria, or eGFR. In flexible dose-response models, the associations were positive and largely linear between urinary uranium concentrations and higher KIM-1 and MCP-1. CONCLUSIONS Among healthy community-living individuals, chronic low-level uranium exposure, as measured in urine, was associated with markers of kidney tubule cell injury. Chronic low-level uranium exposure observed in contemporary US urban centers may adversely affect kidney tubule health and related outcomes.
Collapse
Affiliation(s)
- William A Anderson
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA.
| | - Arce Domingo-Relloso
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Marta Galvez-Fernandez
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Kathrin Schilling
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Ronald A Glabonjat
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Anirban Basu
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Anne E Nigra
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Orlando M Gutierrez
- Department of Medicine and Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rebecca Scherzer
- Kidney Health Research Collaborative, San Francisco Veterans Affairs Healthcare System, and Department of Medicine, University of California, San Francisco, CA, USA
| | - Jeff Goldsmith
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Mark J Sarnak
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Joseph V Bonventre
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Paul L Kimmel
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ramachandran S Vasan
- University of Texas School of Public Health and University of Texas Health Sciences Center, San Antonio, TX, USA; Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Joachim H Ix
- Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, and Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Michael G Shlipak
- Kidney Health Research Collaborative, San Francisco Veterans Affairs Healthcare System, and Department of Medicine, University of California, San Francisco, CA, USA
| | - Ana Navas-Acien
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| |
Collapse
|
4
|
Geraud N, Casemayou A, Alves M, Breuil B, Tkaczyk M, Stańczyk M, Szaflik K, Talar T, Decramer S, Klein J, Schanstra JP, Meyer BB. Evaluation of predictive performance of fetal urinary inflammatory markers of postnatal kidney function in fetuses with posterior urethral valves. Pediatr Nephrol 2025; 40:1023-1032. [PMID: 39614902 PMCID: PMC11885327 DOI: 10.1007/s00467-024-06608-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/03/2024] [Accepted: 11/07/2024] [Indexed: 03/08/2025]
Abstract
BACKGROUND There are proposed roles for inflammation in the development of congenital obstructive uropathy in the setting of posterior urethral valves (PUV). However, the value of inflammatory proteins as predictive markers of postnatal kidney function, key in the management of fetuses with PUV, has not been explored. We screened fetal urine of fetuses with PUV with a panel of inflammatory proteins to determine their predictive value of postnatal kidney function. METHODS Twenty-five different chemokines and cytokines were measured using a multiplex immunoassay in fetal urine of 79 PUV patients from retrospective cohorts, separated in discovery (n = 52) and validation (n = 27). The candidate markers were also quantified in amniotic fluid samples obtained from 16 PUV and 25 other congenital anomalies of the kidney and the urinary tract pregnancies. The performance of validated candidate inflammatory proteins was compared to the previously published 12PUV fetal urine peptide signature. RESULTS Fetal urine chemokines CCL2 (MCP-1), CXCL9 (MIG), and CCL4 (MIP-1β) were identified as predictive of postnatal kidney failure in fetuses with PUV from the discovery cohort. Their predictive potential was confirmed in the validation cohort (AUCs of 0.87, 0.81, and 0.86, respectively). The performance of these individual chemokines was lower than the previously published 12PUV fetal urine peptide signature. However, the combination of the three chemokines performed similarly to 12PUV. In contrast, these three chemokines were not predictive of outcome in amniotic fluid. CONCLUSIONS We identified chemokines in fetal urine of PUV pregnancies that, after external validation, could serve as predictive biomarkers of postnatal outcome and contribute to improve prenatal PUV management.
Collapse
Affiliation(s)
- Nicolas Geraud
- National Institute of Health and Medical Research (INSERM), UMR 1297, Institute of Metabolic and Cardiovascular Disease, Toulouse, France
- University Paul Sabatier, Toulouse III, Toulouse, France
| | - Audrey Casemayou
- National Institute of Health and Medical Research (INSERM), UMR 1297, Institute of Metabolic and Cardiovascular Disease, Toulouse, France
- University Paul Sabatier, Toulouse III, Toulouse, France
- Centre De Référence Des Maladies Rénales Rares du Sud-Ouest (SORARE), Toulouse University Hospital, Toulouse, France
| | - Melinda Alves
- National Institute of Health and Medical Research (INSERM), UMR 1297, Institute of Metabolic and Cardiovascular Disease, Toulouse, France
- University Paul Sabatier, Toulouse III, Toulouse, France
| | - Benjamin Breuil
- National Institute of Health and Medical Research (INSERM), UMR 1297, Institute of Metabolic and Cardiovascular Disease, Toulouse, France
- University Paul Sabatier, Toulouse III, Toulouse, France
| | - Marcin Tkaczyk
- Department of Pediatric, Immunology and Nephrology, Polish Mother's Memorial Hospital Research Institute, Łódź, Poland
| | - Małgorzata Stańczyk
- Department of Pediatric, Immunology and Nephrology, Polish Mother's Memorial Hospital Research Institute, Łódź, Poland
| | - Krzysztof Szaflik
- Department of Gynecology, Fertility and Fetal Therapy, Polish Mother's Memorial Hospital Research Institute, Łódź, Poland
| | - Tomasz Talar
- Department of Neonatology and Intensive Therapy, Polish Mother's Memorial Hospital Research Institute, Łódź, Poland
| | - Stéphane Decramer
- National Institute of Health and Medical Research (INSERM), UMR 1297, Institute of Metabolic and Cardiovascular Disease, Toulouse, France
- University Paul Sabatier, Toulouse III, Toulouse, France
- Centre De Référence Des Maladies Rénales Rares du Sud-Ouest (SORARE), Toulouse University Hospital, Toulouse, France
- Department of Pediatric Internal Medicine, Rheumatology and Nephrology, Toulouse University Hospital, Toulouse, France
| | - Julie Klein
- National Institute of Health and Medical Research (INSERM), UMR 1297, Institute of Metabolic and Cardiovascular Disease, Toulouse, France
- University Paul Sabatier, Toulouse III, Toulouse, France
| | - Joost P Schanstra
- National Institute of Health and Medical Research (INSERM), UMR 1297, Institute of Metabolic and Cardiovascular Disease, Toulouse, France.
- University Paul Sabatier, Toulouse III, Toulouse, France.
| | - Bénédicte Buffin Meyer
- National Institute of Health and Medical Research (INSERM), UMR 1297, Institute of Metabolic and Cardiovascular Disease, Toulouse, France
- University Paul Sabatier, Toulouse III, Toulouse, France
| |
Collapse
|
5
|
Musiał K, Stojanowski J, Bargenda-Lange A, Gołębiowski T. GDF15, EGF, and Neopterin in Assessing Progression of Pediatric Chronic Kidney Disease Using Artificial Intelligence Tools-A Pilot Study. Int J Mol Sci 2025; 26:2344. [PMID: 40076962 PMCID: PMC11899747 DOI: 10.3390/ijms26052344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Cell-mediated immunity and chronic inflammation are hallmarks of chronic kidney disease (CKD). Growth differentiation factor 15 (GDF15) is a marker of inflammation and an integrative signal in stress conditions. Epidermal growth factor (EGF) is a tubule-specific protein that modulates the regeneration of injured renal tubules. Neopterin is a product of activated monocytes and macrophages and serves as a marker of cell-mediated immunity. Our aim was to assess the role of the above-mentioned parameters in the progression of CKD in children using artificial intelligence tools. The study group consisted of 151 children with CKD stages 1-5. EGF, GDF15, and neopterin serum concentrations were assessed by ELISA. The patients' anthropometric data, biochemical parameters, EGF, GDF15, and neopterin serum values were implemented into the artificial neural network (ANN). The most precise model contained EGF, GDF15, and neopterin as input parameters and classified patients into either CKD 1-3 or CKD 4-5 groups with an excellent accuracy of 96.77%. The presented AI model, with serum concentrations of EGF, GDF15, and neopterin as input parameters, may serve as a useful predictor of CKD progression. It suggests the essential role of inflammatory processes in the renal function decline in the course of CKD in children.
Collapse
Affiliation(s)
- Kinga Musiał
- Department of Pediatric Nephrology, Wrocław Medical University, Borowska 213, 50-556 Wrocław, Poland
| | - Jakub Stojanowski
- Department of Nephrology and Transplantation Medicine, Wrocław Medical University, Borowska 213, 50-556 Wrocław, Poland
| | - Agnieszka Bargenda-Lange
- Department of Pediatric Nephrology, Wrocław Medical University, Borowska 213, 50-556 Wrocław, Poland
| | - Tomasz Gołębiowski
- Department of Nephrology and Transplantation Medicine, Wrocław Medical University, Borowska 213, 50-556 Wrocław, Poland
| |
Collapse
|
6
|
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
|
7
|
Geurts F, van Heugten MH, Blijdorp CJ, Fenton RA, Chaker L, Hoorn EJ. Urinary EGF Reflects Distal Tubular Mass and Is Associated with Hypertension, Serum Magnesium, and Kidney Outcomes. KIDNEY360 2025; 6:451-460. [PMID: 39714970 PMCID: PMC11970850 DOI: 10.34067/kid.0000000687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
Key Points Donor nephrectomy reduced urinary EGF (uEGF) by half and correlated with the reduction in kidney volume, suggesting that uEGF reflects tubular mass. In the general population, lower uEGF/creatinine was associated with lower eGFR, lower serum magnesium, and higher BP. Lower uEGF/creatinine, was associated with incident CKD, and this association was stronger in people without hypertension. Background EGF is expressed in the distal tubule and secreted in urine (urinary EGF [uEGF]) after cleavage of membrane-bound pro-EGF. Lower uEGF is associated with kidney disease progression. EGF also plays a role in the regulation of serum magnesium and BP, but whether uEGF is associated with these parameters is unknown. We hypothesized that uEGF is a distal tubule marker associated with serum magnesium, BP, and kidney outcomes. Methods We first used a cohort of kidney donors (N =20) and measured uEGF to analyze the association with tubular mass and pro-EGF in urinary extracellular vesicles as proxy for tubular expression. Next, we measured uEGF in a population-based cohort (N =2382) to investigate the associations with serum magnesium, hypertension, and kidney outcomes (incident eGFR <60 or <45 ml/min per 1.73 m2, 40% loss of eGFR, or kidney failure). Results Kidney donation decreased eGFR from 86 to 54 ml/min per 1.73 m2 (36% reduction; 95% confidence interval [CI], 31% to 42%), uEGF from 28 to 14 µ g/24 hours (49% reduction; 95% CI, 42% to 55%), and pro-EGF by 29% (95% CI, 12% to 45%). The decrease in uEGF correlated with the decrease in kidney volume. In the population cohort, lower uEGF was significantly associated with hypertension and lower serum magnesium. The association between uEGF and serum magnesium was stronger in participants with lower eGFR, hypertension, and diuretic use. Lower uEGF at baseline was also associated with worse kidney outcomes, and this association was stronger for normotensive participants. Conclusions uEGF is a marker of distal tubular mass that is not only associated with kidney disease progression, but also with serum magnesium and BP. Future studies should address whether normotensive people with low uEGF excretion represent a group that may benefit from kidney-protective treatment.
Collapse
Affiliation(s)
- Frank Geurts
- Division of Nephrology and Transplantation, Erasmus Medical Center, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Martijn H. van Heugten
- Division of Nephrology and Transplantation, Erasmus Medical Center, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Charles J. Blijdorp
- Division of Nephrology and Transplantation, Erasmus Medical Center, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Layal Chaker
- Division of Nephrology and Transplantation, Erasmus Medical Center, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ewout J. Hoorn
- Division of Nephrology and Transplantation, Erasmus Medical Center, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
8
|
Greenberg JH, Abraham AG, Xu Y, Schelling JR, Coca SG, Schrauben SJ, Wilson FP, Waikar SS, Vasan RS, Gutierrez OM, Shlipak MG, Ix JH, Warady BA, Kimmel PL, Bonventre JV, Parikh CR, Denburg M, Furth S. Biomarker Panels for Discriminating Risk of CKD Progression in Children. J Am Soc Nephrol 2025:00001751-990000000-00529. [PMID: 39820177 DOI: 10.1681/asn.0000000602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 01/10/2025] [Indexed: 01/19/2025] Open
Abstract
Key Points
Plasma biomarkers (kidney injury molecule-1, KIM-1), urine biomarkers (EGF/creatinine and urine albumin-creatinine ratio), and eGFR identified four prognostic groups in children with CKD progression.A panel of biomarkers may better capture the complexity of kidney disease in children and may allow for a broader assessment of kidney health.
Background
We have previously studied biomarkers of tubular health (EGF), injury (kidney injury molecule-1 [KIM-1]), dysfunction (α-1 microglobulin), and inflammation (TNF receptor-1, TNF receptor-2, monocyte chemoattractant protein-1, YKL-40, and soluble urokinase plasminogen activator receptor) and demonstrated that plasma KIM-1, TNF receptor-1, TNF receptor-2, urine KIM-1, EGF, monocyte chemoattractant protein-1, and urine α-1 microglobulin are each independently associated with CKD progression in children. In this study, we used bootstrapped survival trees to identify a combination of biomarkers to predict CKD progression in children.
Methods
The Chronic Kidney Disease in Children (CKiD) Cohort Study prospectively enrolled children aged 6 months to 16 years with an eGFR of 30–90 ml/min per 1.73 m2. We measured biomarkers in stored plasma and urine collected 5 months after study enrollment. The primary outcome of CKD progression was a composite of 50% eGFR decline or kidney failure. We constructed a regression tree–based model for predicting the time to the composite event, using a panel of clinically relevant biomarkers with empirically derived thresholds, in addition to conventional risk factors.
Results
Of the 599 children included, the median age was 12 years (interquartile range [IQR], 8–15), 371 (62%) were male, baseline urine protein-creatinine ratio was 0.33 (IQR, 0.12–0.95) mg/mg, and baseline eGFR was 53 (IQR, 40–66) ml/min per 1.73 m2. Overall, 205 children (34%) reached the primary outcome of CKD progression. A single regression tree–based model using the most informative predictors with data-driven biomarker thresholds suggested a final set of four prognosis groups. In the final model, urine albumin/creatinine was the variable with the highest importance and along with urine EGF/creatinine identified the highest risk group of 24 children, 100% of whom developed CKD progression at a median time of 1.3 years (95% confidence interval [CI], 1.0 to 1.7). When the regression tree–derived risk group classifications were added to prediction models including the clinical risk factors, the C-statistic increased from 0.76 (95% CI, 0.71 to 0.80) to 0.85 (95% CI, 0.81 to 0.88).
Conclusions
Using regression tree–based methods, we identified a biomarker panel of urine albumin/creatinine, urine EGF/creatinine, plasma KIM-1, and eGFR, which significantly improved discrimination for CKD progression.
Collapse
Grants
- K08DK110536 NIDDK NIH HHS
- R01DK135650 NIDDK NIH HHS
- U01DK106982 Division of Diabetes, Endocrinology, and Metabolic Diseases
- U01 DK085689, U01 DK102730, U01 DK103225, U01 DK085660 NIDDK NIH HHS
- U01 DK085689, U01 DK102730, U01 DK103225, U01 DK085660 NIDDK NIH HHS
- U01 DK085689, U01 DK102730, U01 DK103225, U01 DK085660 NIDDK NIH HHS
- U01 DK085689, U01 DK102730, U01 DK103225, U01 DK085660 NIDDK NIH HHS
- U01 DK085689, U01 DK102730, U01 DK103225, U01 DK085660 NIDDK NIH HHS
- U01 DK085689, U01 DK102730, U01 DK103225, U01 DK085660 NIDDK NIH HHS
- U01 DK085689, U01 DK102730, U01 DK103225, U01 DK085660 NIDDK NIH HHS
- U01 DK085689, U01 DK102730, U01 DK103225, U01 DK085660 NIDDK NIH HHS
- U01 DK085689, U01 DK102730, U01 DK103225, U01 DK085660 NIDDK NIH HHS
- U01 DK085689, U01 DK102730, U01 DK103225, U01 DK085660 NIDDK NIH HHS
- U01 DK085689, U01 DK102730, U01 DK103225, U01 DK085660 NIDDK NIH HHS
- U01 DK085689, U01 DK102730, U01 DK103225, U01 DK085660 NIDDK NIH HHS
Collapse
Affiliation(s)
- Jason H Greenberg
- Section of Nephrology, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Yale University School of Medicine, New Haven, Connecticut
| | - Alison G Abraham
- Department of Epidemiology, University of Colorado, Denver, Colorado
| | - Yunwen Xu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, New York
| | - Jeffrey R Schelling
- Division of Nephrology, Department of Physiology and Biophysics and Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Steven G Coca
- Section of Nephrology, Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sarah J Schrauben
- Departments of Medicine and Epidemiology, Clinical Center for Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - F Perry Wilson
- Clinical and Translational Research Accelerator, Yale University School of Medicine, New Haven, Connecticut
| | - Sushrut S Waikar
- Section of Nephrology, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts
| | - Ramachandran S Vasan
- Departments of Medicine and Epidemiology, Boston University School of Medicine, Boston, Massachusetts
| | - Orlando M Gutierrez
- Section of Nephrology, Department of Medicine, University of Alabama, Birmingham, Alabama
| | - Michael G Shlipak
- Kidney Health Research Collaborative, Department of Medicine, San Francisco Veterans Affairs Healthcare System, University of California, San Francisco, California
| | - Joachim H Ix
- Nephrology Section, Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Bradley A Warady
- Division of Nephrology, Department of Pediatrics, Children's Mercy Kansas City, Kansas City, Missouri
| | - Paul L Kimmel
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Joseph V Bonventre
- Section of Nephrology, Department of Internal Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Chirag R Parikh
- Section of Nephrology, Department of Internal Medicine, Johns Hopkins School of Medicine, Baltimore, New York
| | - Michelle Denburg
- Division of Nephrology, Department of Pediatrics, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Susan Furth
- Section of Nephrology, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
9
|
Chen TK, Surapaneni AL, Schmidt IM, Waikar SS, Coresh J, Liu H, Susztak K, Rhee EP, Liu C, Schlosser P, Grams ME. Proteomics and Incident Kidney Failure in Individuals With CKD: The African American Study of Kidney Disease and Hypertension and the Boston Kidney Biopsy Cohort. Kidney Med 2024; 6:100921. [PMID: 39634331 PMCID: PMC11615895 DOI: 10.1016/j.xkme.2024.100921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
Rationale & Objective Individuals with chronic kidney disease (CKD) are at increased risk of morbidity and mortality, particularly as they progress to kidney failure. Identifying circulating proteins that underlie kidney failure development may guide the discovery of new targets for intervention. Study Design Prospective cohort. Setting & Participants 703 African American Study of Kidney Disease and Hypertension (AASK) and 434 Boston Kidney Biopsy Cohort (BKBC) participants with baseline proteomics data. Exposures Circulating proteins measured using SomaScan. Outcomes Kidney failure, defined as dialysis initiation or kidney transplantation. Analytical Approach Using adjusted Cox models, we studied associations of 6,284 circulating proteins with kidney failure risk separately in AASK and BKBC and meta-analyzed results. We then performed gene set enrichment analyses to identify underlying perturbations in biological pathways. In separate data sets with kidney-tissue level gene expression, we ascertained dominant regions of expression and correlated kidney tubular gene expression with fibrosis and estimated glomerular filtration rate (eGFR). Results Over median follow-up periods of 8.8 and 3.1 years, 210 AASK (mean age: 55 years, 39% female, mean GFR: 46 mL/min/1.73 m2) and 115 BKBC (mean age: 54 years, 47% female, mean eGFR: 51 mL/min/1.73 m2) participants developed kidney failure, respectively. We identified 143 proteins that were associated with incident kidney failure, of which only 1 (Testican-2) had a lower risk. Notable proteins included those related to vascular permeability (endothelial cell-selective adhesion molecule), glomerulosclerosis (ephrin-A1), glomerular development (ephrin-B2), intracellular sorting/transport (vesicular integral-membrane protein VIP36), podocyte effacement (pigment epithelium-derived factor), complement activation (complement decay-accelerating factor), and fibrosis (ephrin-A1, ephrin-B2, and pigment epithelium-derived factor). Gene set enrichment analyses detected overrepresented pathways that could be related to CKD progression, such as ephrin signaling, cell-cell junctions, intracellular transport, immune response, cell proliferation, and apoptosis. At the kidney level, glomerular expression predominated for genes corresponding to circulating proteins of interest, and several gene expression levels were correlated with eGFR and/or fibrosis. Limitations Possible residual confounding. Conclusions Multimodal data identified proteins and pathways associated with the development of kidney failure.
Collapse
Affiliation(s)
- Teresa K. Chen
- Kidney Health Research Collaborative and Division of Nephrology, Department of Medicine, University of California, San Francisco, CA
- San Francisco VA Health Care System, San Francisco, CA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Aditya L. Surapaneni
- Department of Medicine, New York University Langone School of Medicine, New York, NY
| | | | | | - Josef Coresh
- Department of Medicine, New York University Langone School of Medicine, New York, NY
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Hongbo Liu
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Katalin Susztak
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Eugene P. Rhee
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Celina Liu
- Department of Medicine, New York University Langone School of Medicine, New York, NY
| | - Pascal Schlosser
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Faculty of Medicine and Medical Center, Institute of Genetic Epidemiology, University of Freiburg, Freiburg, Germany
| | - Morgan E. Grams
- Department of Medicine, New York University Langone School of Medicine, New York, NY
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
10
|
Degenaar A, Kruger R, Jacobs A, Mels CMC. Phenotyping Kidney Function in Young Adults With High Blood Pressure: The African-PREDICT Study. J Clin Hypertens (Greenwich) 2024; 26:1291-1300. [PMID: 39368068 PMCID: PMC11555542 DOI: 10.1111/jch.14911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 10/07/2024]
Abstract
Biomarkers of kidney function, including glomerular, tubular, and fibrotic markers, have been associated with blood pressure in elderly populations and individuals with kidney and cardiovascular diseases. However, limited information is available in young adults. In this study, we compared levels of several kidney function biomarkers between normotensive and hypertensive young adults and explored the associations of these biomarkers with blood pressure within these groups. In this cross-sectional assessment, twenty-four-hour (24-h) blood pressure measurements of 1055 participants (mean age = 24.6 years) were used to classify hypertension as per the 2018 ESC/ESH guidelines. Biomarkers of kidney function included estimated glomerular filtration rate, urinary albumin, alpha-1 microglobulin (uA1M), neutrophil gelatinase-associated lipocalin (uNGAL), uromodulin (uUMOD), and the CKD273 classifier. All urinary biomarkers, except for the CKD273 classifier, were standardized for urinary creatinine (Cr). In the hypertensive group (61.0% White; 73.2% men), urinary albumin-to-creatinine ratio (uACR), uNGAL/Cr and uUMOD/Cr were lower than the normotensive group. In multiple regression analyses, 24-h systolic blood pressure (SBP) (β = 0.14; p = 0.042), 24-h diastolic blood pressure (DBP) (β = 0.14; p = 0.040), and 24-h mean arterial pressure (MAP) (β = 0.16; p = 0.020) associated positively with uA1M/Cr in the hypertensive group, while 24-h MAP positively associated with uACR (β = 0.17; p = 0.017). In exploratory factor analysis, positive associations of 24-h DBP and 24-h MAP with a factor pattern including tubular biomarkers were observed in the hypertensive group (24-h DBP: β = 0.18; p = 0.026, 24-h MAP: β = 0.17; p = 0.032). In the setting of hypertension, high perfusion pressure in the kidneys may play a role in the development of proximal tubule damage and promote early deterioration in kidney function in young adults. Trial Registration: ClinicalTrials.gov identifier: NCT03292094.
Collapse
Affiliation(s)
- Anja Degenaar
- Hypertension in Africa Research Team (HART)North‐West UniversityPotchefstroomSouth Africa
| | - Ruan Kruger
- Hypertension in Africa Research Team (HART)North‐West UniversityPotchefstroomSouth Africa
- Medical Research Council: Research Unit for Hypertension and Cardiovascular Disease, North‐West UniversityPotchefstroomSouth Africa
| | - Adriaan Jacobs
- Hypertension in Africa Research Team (HART)North‐West UniversityPotchefstroomSouth Africa
- Medical Research Council: Research Unit for Hypertension and Cardiovascular Disease, North‐West UniversityPotchefstroomSouth Africa
| | - Catharina M. C. Mels
- Hypertension in Africa Research Team (HART)North‐West UniversityPotchefstroomSouth Africa
- Medical Research Council: Research Unit for Hypertension and Cardiovascular Disease, North‐West UniversityPotchefstroomSouth Africa
| |
Collapse
|
11
|
Litt JS, Belfort MB, Everson TM, Haneuse S, Tiemeier H. Neonatal multimorbidity and the phenotype of premature aging in preterm infants. Pediatr Res 2024:10.1038/s41390-024-03617-2. [PMID: 39455859 DOI: 10.1038/s41390-024-03617-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024]
Abstract
Multimorbidity is the co-occurrence of multiple chronic health problems, associated with aging, frailty, and poor functioning. Children born preterm experience more multimorbid conditions in early life compared to term-born peers. Though neonatal multimorbidity is linked to poor health-related quality of life, functional outcomes, and peer group participation, gaps in our theoretical understanding and conceptualization remain. Drawing from life course epidemiology and the Developmental Origins of Heath and Disease models, we offer a framework that neonatal multimorbidity reflects maturational vulnerability posed by preterm birth. The impact of such vulnerability on health and development may be further amplified by adverse exposures and interventions within the environment of the neonatal intensive care unit. This can be exacerbated by disadvantaged home or community contexts after discharge. Uncovering the physiologic and social antecedents of multiple morbid conditions in the neonatal period and their biological underpinnings will allow for more accurate risk-prediction, counseling, and care planning for preterm infants and their families. According to this framework, the maturational vulnerability to multimorbidity imparted by preterm birth and its negative effects on health and development are not predetermined or static. Elucidating pathways of early biologic and physical aging will lead to improvements in care and outcomes. IMPACT: Multimorbidity is associated with significant frailty and dysfunction among older adults and is indicative of early physiologic aging. Preterm infants commonly experience multimorbidities in the newborn period, an underrecognized threat to long-term health and development. We offer a novel framework incorporating multimorbidity, early cellular aging, and life course health development to innovate risk-prediction, care-planning, and therapeutics.
Collapse
Affiliation(s)
- Jonathan S Litt
- Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, USA.
- Department of Pediatrics, Harvard Medical School, Boston, USA.
- Department of Social and Behavioral Sciences, Harvard TH Chan School of Public Health, Boston, USA.
| | - Mandy Brown Belfort
- Department of Pediatrics, Harvard Medical School, Boston, USA
- Department of Pediatrics, Brigham and Women's Hospital, Boston, USA
| | - Todd M Everson
- Department of Environmental Health, Emory University, Atlanta, USA
| | - Sebastien Haneuse
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, USA
| | - Henning Tiemeier
- Department of Social and Behavioral Sciences, Harvard TH Chan School of Public Health, Boston, USA
| |
Collapse
|
12
|
Harita Y. Urinary extracellular vesicles in childhood kidney diseases. Pediatr Nephrol 2024; 39:2293-2300. [PMID: 38093081 PMCID: PMC11199279 DOI: 10.1007/s00467-023-06243-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 06/26/2024]
Abstract
Most biological fluids contain extracellular vesicles (EVs). EVs are surrounded by a lipid bilayer and contain biological macromolecules such as proteins, lipids, RNA, and DNA. They lack a functioning nucleus and are incapable of replicating. The physiological characteristics and molecular composition of EVs in body fluids provide valuable information about the status of originating cells. Consequently, they could be effectively utilized for diagnostic and prognostic applications. Urine contains a heterogeneous population of EVs. To date, these urinary extracellular vesicles (uEVs) have been ignored in the standard urinalysis. In recent years, knowledge has accumulated on how uEVs should be separated and analyzed. It has become clear how uEVs reflect the expression of each molecule in cells in nephron segments and how they are altered in disease states such as glomerular/tubular disorders, rare congenital diseases, acute kidney injury (AKI), and chronic kidney disease (CKD). Significant promise exists for the molecular expression signature of uEVs detected by simple techniques such as enzyme-linked immunosorbent assay (ELISA), making them more applicable in clinical settings. This review presents the current understanding regarding uEVs, emphasizing the potential for non-invasive diagnostics, especially for childhood kidney diseases.
Collapse
Affiliation(s)
- Yutaka Harita
- Department of Pediatrics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
13
|
Troost JP, D’Souza J, Buxton M, Kshirsagar AV, Engel LS, O’Lenick CR, Smoyer WE, Klein J, Ju W, Eddy S, Helmuth M, Mariani LH, Kretzler M, Trachtman H. Elevated Exposure to Air Pollutants Accelerates Primary Glomerular Disease Progression. Kidney Int Rep 2024; 9:2527-2536. [PMID: 39156153 PMCID: PMC11328569 DOI: 10.1016/j.ekir.2024.05.013] [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: 02/09/2024] [Revised: 05/02/2024] [Accepted: 05/13/2024] [Indexed: 08/20/2024] Open
Abstract
Introduction Environmental contributors to kidney disease progression remain elusive. We explored how residential air pollution affects disease progression in patients with primary glomerulopathies. Methods Nephrotic Syndrome Study Network (NEPTUNE) and CureGlomerulonephropathy (CureGN) participants with residential census tract data and ≥2 years of follow-up were included. Using Cox proportional hazards models, the associations per doubling in annual average baseline concentrations of total particulate matter with diameter ≤2.5 μm (PM2.5) and its components, black carbon (BC), and sulfate, with time to ≥40% decline in estimated glomerular filtration rate (eGFR) or kidney failure were estimated. Serum tumour necrosis factor levels and kidney tissue transcriptomic inflammatory pathway activation scores were used as molecular markers of disease progression. Results PM2.5, BC, and sulfate exposures were comparable in NEPTUNE (n = 228) and CureGN (n = 697). In both cohorts, participants from areas with higher levels of pollutants had lower eGFR, were older and more likely self-reported racial and ethnic minorities. In a fully adjusted model combining both cohorts, kidney disease progression was associated with PM2.5 (adjusted hazard ratio 1.55 [95% confidence interval: 1.00-2.38], P = 0.0489) and BC (adjusted hazard ratio 1.43 [95% confidence interval: 0.98-2.07], P = 0.0608) exposure. Sulfate and PM2.5 exposure were positively correlated with serum tumour necrosis factor (TNF) (P = 0.003) and interleukin-1β levels (P = 0.03), respectively. Sulfate exposure was also directly associated with transcriptional activation of the TNF and JAK-STAT signaling pathways in kidneys (r = 0.55-0.67, P-value <0.01). Conclusion Elevated exposure to select air pollutants is associated with increased risk of disease progression and systemic inflammation in patients with primary.
Collapse
Affiliation(s)
- Jonathan P. Troost
- Michigan Institute for Clinical & Health Research, University of Michigan, Ann Arbor Michigan, USA
| | - Jennifer D’Souza
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Miatta Buxton
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Abhijit V. Kshirsagar
- Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Lawrence S. Engel
- Departments of Epidemiology and Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Cassandra R. O’Lenick
- Departments of Epidemiology and Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, North Carolina, USA
| | - William E. Smoyer
- Department of Pediatrics, Ohio State University, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Jon Klein
- Division of Nephrology and Hypertension, Department of Medicine, Christina Lee Brown Environment Institute, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Robley Rex Veterans Administration Medical Center, Louisville, Kentucky, USA
| | - Wenjun Ju
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor Michigan, USA
| | - Sean Eddy
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Margaret Helmuth
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Laura H. Mariani
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthias Kretzler
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor Michigan, USA
| | - Howard Trachtman
- Division of Nephrology, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
14
|
Ono K, Maeshima A, Nagayama I, Kubo T, Yagisawa T, Nagata D. Urinary Epidermal Growth Factor Level as a Noninvasive Indicator of Tubular Repair in Patients with Acute Kidney Injury. Diagnostics (Basel) 2024; 14:947. [PMID: 38732362 PMCID: PMC11083164 DOI: 10.3390/diagnostics14090947] [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: 03/28/2024] [Revised: 04/28/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
Epidermal growth factor (EGF), an essential factor for the proliferation and survival of renal tubular cells, is expressed by distal tubules and normally excreted via urine. Previous studies in rats demonstrated that acute tubular injury reduces urinary EGF levels. However, it is unclear whether urinary EGF is a suitable monitoring marker of tubular repair status after acute kidney injury (AKI) in humans. To address this question, we measured serum and urinary EGF in patients with AKI (n = 99) using ELISA and investigated whether urinary EGF levels were associated with the severity of tubular injury and renal prognosis. Urinary EGF was abundant in healthy controls but showed a significant decrease in AKI patients (14,522 ± 2190 pg/mL vs. 3201 ± 459.7 pg/mL, p < 0.05). The urinary EGF level in patients with renal AKI was notably lower than that in patients with pre-renal AKI. Furthermore, the urinary EGF level in patients with AKI stage 3 was significantly lower than that in patients with AKI stage 1. Urinary EGF levels were negatively correlated with urinary β-2MG and serum creatinine levels but positively correlated with hemoglobin levels and eGFR. Urinary EGF was not significantly correlated with urinary NAG, α-1MG, L-FABP, NGAL, KIM-1, or urinary protein concentrations. No significant correlation was observed between serum and urinary EGF levels, suggesting that urinary EGF is derived from the renal tubules rather than the blood. In living renal transplantation donors, the urinary EGF/Cr ratio was approximately half the preoperative urinary EGF/Cr ratio after unilateral nephrectomy. Collectively, these data suggest that urinary EGF is a suitable noninvasive indicator of not only the volume of functional normal renal tubules but also the status of tubular repair after AKI.
Collapse
Affiliation(s)
- Kazutoshi Ono
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Akito Maeshima
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, Kawagoe 350-8550, Japan
| | - Izumi Nagayama
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, Kawagoe 350-8550, Japan
| | - Taro Kubo
- Department of Renal Surgery and Transplantation, Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Takashi Yagisawa
- Department of Renal Surgery and Transplantation, Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Daisuke Nagata
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke 329-0498, Japan
| |
Collapse
|
15
|
Hingorani SR, Schmicker RH, Halloran B, Brophy P, Heagerty PJ, Juul S, Goldstein SL, Askenazi D. Association Between Urinary Biomarkers and CKD in Extremely Low Gestational Age Neonates. Am J Kidney Dis 2024; 83:497-507. [PMID: 37926336 PMCID: PMC10960703 DOI: 10.1053/j.ajkd.2023.09.008] [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: 05/16/2023] [Revised: 08/22/2023] [Accepted: 09/01/2023] [Indexed: 11/07/2023]
Abstract
RATIONALE & OBJECTIVE Children born before 28 weeks' gestation are at increased risk of chronic kidney disease (CKD). Urine biomarkers may shed light on mechanistic pathways and improve the ability to forecast CKD. We evaluated whether urinary biomarkers in neonates of low gestational age (GA) are associated with a reduced estimated glomerular filtration rate (eGFR) over time. STUDY DESIGN A cohort study of neonates with an exploratory case-control study of a subset of the cohort. SETTING & PARTICIPANTS 327 neonates born at 24-27 weeks' gestation with 2-year eGFR data from the PENUT (Preterm Erythropoietin Neuroprotection Trial) and the REPaIReD (Recombinant Erythropoietin for Prevention of Infant Renal Disease) study. EXPOSURES 11 urinary biomarkers measured at 27, 30, and 34 weeks' postmenstrual age for the primary cohort study and 10 additional biomarkers for the exploratory case-control study. OUTCOMES eGFR<90mL/min/1.73m2 at 2 years corrected for GA. ANALYTICAL APPROACH Linear mixed models to assess differences in biomarker values between neonates in whom CKD did and did not develop, accounting for multiple comparisons using Bonferroni-Holm correction in the cohort study only. Cohort analyses were adjusted for sex, GA, and body mass index. Cases were matched to controls on these variables in the case-control study. RESULTS After adjusting for weeks of GA, urinary levels of α-glutathione-S-transferase (log difference, 0.27; 95% CI, 0.12-0.43), albumin (log difference, 0.13; 95% CI, 0.02-0.25), and cystatin C (log difference, 0.19; 95% CI, 0.04-0.34) were higher in those in whom CKD developed than in those in whom it did not. Urinary albumin and cystatin C levels did not remain significantly different after Bonferroni-Holm correction. In the exploratory case-control analysis, there were no differences in any biomarkers between cases and controls. LIMITATIONS Early deaths and a high number of subjects without eGFR at 2 years corrected for GA. CONCLUSIONS Measurement of urinary biomarkers may assist in monitoring neonates who are at risk for CKD. Additional studies are needed to confirm these findings. FUNDING Grants from government (National Institutes of Health). TRIAL REGISTRATION Registered at ClinicalTrials.gov with study number NCT01378273. PLAIN-LANGUAGE SUMMARY Approximately 15 million neonates worldwide are born prematurely, and 2 million are born before 28 weeks' gestation. Many of these children go on to experience chronic kidney disease. Urine biomarkers may allow for early recognition of those at risk for the development of kidney disease. In this study of more than 300 children born before 28 weeks' gestational age, we found higher mean urinary levels of α-glutathione-S-transferase at 27, 30, and 34 weeks in children whose estimated glomerular filtration rate was<90mL/min/1.73m2 at 2 years compared with children whose estimated glomerular filtration rate was>90mL/min/1.73m2 at 2 years. Measurement of urinary biomarkers may assist in monitoring neonates who are at risk for chronic kidney disease. Additional studies are needed to confirm our findings.
Collapse
Affiliation(s)
- Sangeeta R Hingorani
- Divisions of Nephrology, Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, Washington.
| | - Robert H Schmicker
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Brian Halloran
- Division of Nephrology, Department of Pediatrics, University of Alabama at Birmingham, Children's of Alabama, Birmingham, Alabama
| | - Patrick Brophy
- Department of Pediatrics, University of Rochester School of Medicine, Rochester, New York
| | - Patrick J Heagerty
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Sandra Juul
- Neonatology, Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, Washington
| | - Stuart L Goldstein
- Division of Nephrology and Hypertension, Department of Pediatrics, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David Askenazi
- Division of Nephrology, Department of Pediatrics, University of Alabama at Birmingham, Children's of Alabama, Birmingham, Alabama
| |
Collapse
|
16
|
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
|
17
|
Pan Y, Wong CYP, Ma H, Tse RTH, Cheng CKL, Tan M, Chiu PKF, Teoh JYC, Wang X, Ng CF, Zhang L. Quantitative comparison of the renal pelvic urine and bladder urine to examine modifications of the urine proteome by the lower urinary tract. Proteomics Clin Appl 2024; 18:e2300004. [PMID: 37574260 DOI: 10.1002/prca.202300004] [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: 01/13/2023] [Revised: 06/21/2023] [Accepted: 08/04/2023] [Indexed: 08/15/2023]
Abstract
PURPOSE Urine proteome is a valuable reservoir of biomarkers for disease diagnosis and monitoring. Following formation as the plasma filtrate in the kidney, urine is progressively modified by the active reabsorption and secretion of the urinary tract. However, little is known about how the urine proteome changes as it passes along the urinary tract. EXPERIMENTAL DESIGN To investigate this, we compared the proteome composition of the renal pelvis urine (RPU) and individually self-voided bladder urine (BU) collected from seven unilateral urinary tract obstruction male patients by LC-MS/MS screening. To our knowledge, this is the first proteomic comparison of RPU and BU samples from the same individual. RESULTS Overall, RPU and BU proteomes did not exhibit proteins that were exclusively present in all samples of one urine type while in none of the other type. Nonetheless, BU had more overrepresented proteins that were observed at a higher frequency than RPU. Label-free quantitative analyses revealed BU-RPU differential proteins that are enriched in exosomes and extracellular proteins. However, the differences were not significant after corrections for multiple testing. Interestingly, we observed a significant increase of collagen peptides with hydroxyproline modifications in the BU samples, suggesting differences in protein modifications. CONCLUSIONS AND CLINICAL RELEVANCE Our study revealed no substantial differences at the protein level between the BU and RPU samples. Future investigations with expanded cohorts would provide more insights about the urothelial-urinary interactions.
Collapse
Affiliation(s)
- Yilin Pan
- Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, Kowloon, Hong Kong SAR, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong, China
| | - Christine Yim-Ping Wong
- S.H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Haiying Ma
- Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Ryan Tsz-Hei Tse
- S.H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Carol Ka-Lo Cheng
- S.H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Miaomiao Tan
- Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Peter Ka-Fung Chiu
- S.H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Jeremy Yuen-Chun Teoh
- S.H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Xin Wang
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi-Fai Ng
- S.H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Liang Zhang
- Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, Kowloon, Hong Kong SAR, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong, China
| |
Collapse
|
18
|
Liu Y, Xu K, Xiang Y, Ma B, Li H, Li Y, Shi Y, Li S, Bai Y. Role of MCP-1 as an inflammatory biomarker in nephropathy. Front Immunol 2024; 14:1303076. [PMID: 38239353 PMCID: PMC10794684 DOI: 10.3389/fimmu.2023.1303076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 12/12/2023] [Indexed: 01/22/2024] Open
Abstract
The Monocyte chemoattractant protein-1 (MCP-1), also referred to as chemokine ligand 2 (CCL2), belongs to the extensive chemokine family and serves as a crucial mediator of innate immunity and tissue inflammation. It has a notable impact on inflammatory conditions affecting the kidneys. Upon binding to its receptor, MCP-1 can induce lymphocytes and NK cells' homing, migration, activation, differentiation, and development while promoting monocytes' and macrophages' infiltration, thereby facilitating kidney disease-related inflammation. As a biomarker for kidney disease, MCP-1 has made notable advancements in primary kidney diseases such as crescentic glomerulonephritis, chronic glomerulonephritis, primary glomerulopathy, idiopathic proteinuria glomerulopathy, acute kidney injury; secondary kidney diseases like diabetic nephropathy and lupus nephritis; hereditary kidney diseases including autosomal dominant polycystic kidney disease and sickle cell kidney disease. MCP-1 not only predicts the occurrence, progression, prognosis of the disease but is also closely associated with the severity and stage of nephropathy. When renal tissue is stimulated or experiences significant damage, the expression of MCP-1 increases, demonstrating a direct correlation with the severity of renal injury.
Collapse
Affiliation(s)
- Yanlong Liu
- Heilongjiang Provincial Health Commission, Harbin, China
| | - Ke Xu
- Heilongjiang University of Chinese Medicine, The Second Clinical Medical College, Harbin, China
| | - Yuhua Xiang
- Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Boyan Ma
- Heilongjiang University of Chinese Medicine, The Second Clinical Medical College, Harbin, China
| | - Hailong Li
- Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Yuan Li
- The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yue Shi
- Heilongjiang University of Chinese Medicine, The Second Clinical Medical College, Harbin, China
| | - Shuju Li
- Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Yan Bai
- Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| |
Collapse
|
19
|
de Fontnouvelle C, Zappitelli M, Thiessen-Philbrook HR, Jia Y, Kimmel PL, Kaufman JS, Devarajan P, Parikh CR, Greenberg JH. Biomarkers of eGFR decline after cardiac surgery in children: findings from the ASSESS-AKI study. Pediatr Nephrol 2023; 38:2851-2860. [PMID: 36790467 DOI: 10.1007/s00467-023-05886-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 01/09/2023] [Accepted: 01/09/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND Children who require surgery for congenital heart disease have increased risk for long-term chronic kidney disease (CKD). Clinical factors as well as urine biomarkers of tubular health and injury may help improve the prognostication of estimated glomerular filtration rate (eGFR) decline. METHODS We enrolled children from 1 month to 18 years old undergoing cardiac surgery in the ASSESS-AKI cohort. We used mixed-effect models to assess the association between urinary biomarkers (log2-transformed uromodulin, NGAL, KIM-1, IL-18, L-FABP) measured 3 months after cardiac surgery and cyanotic heart disease with the rate of eGFR decline at annual in-person visits over 4 years. RESULTS Of the 117 children enrolled, 30 (24%) had cyanotic heart disease. During 48 months of follow-up, the median eGFR in the subgroup of children with cyanotic heart disease was lower at all study visits as compared with children with acyanotic heart disease (p = 0.01). In the overall cohort, lower levels of both urine uromodulin and IL-18 after discharge were associated with eGFR decline. After adjustment for age, RACHS-1 surgical complexity score, proteinuria, and eGFR at the 3-month study visit, lower concentrations of urine uromodulin and IL-18 were associated with a monthly decline in eGFR (uromodulin β = 0.04 (95% CI: 0.00-0.09; p = 0.07) IL-18 β = 0.07 (95% CI: 0.01-0.13; p = 0.04), ml/min/1.73 m2 per month). CONCLUSIONS At 3 months after cardiac surgery, children with lower urine uromodulin and IL-18 concentrations experienced a significantly faster decline in eGFR. Children with cyanotic heart disease had a lower median eGFR at all time points but did not experience faster eGFR decline. A higher-resolution version of the Graphical abstract is available as Supplementary information.
Collapse
Affiliation(s)
| | - Michael Zappitelli
- Department of Pediatrics, Toronto Hospital for Sick Children, Toronto, Canada
| | | | - Yaqi Jia
- Department of Internal Medicine, Section of Nephrology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Paul L Kimmel
- National Institute of Diabetes and Digestive Kidney Diseases (NIDDK), Bethesda, MD, USA
| | - James S Kaufman
- Division of Nephrology, New York University Grossman School of Medicine and VA New York Harbor Healthcare System, New York, NY, USA
| | - Prasad Devarajan
- Department of Nephrology and Hypertension, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Chirag R Parikh
- Department of Internal Medicine, Section of Nephrology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jason H Greenberg
- Clinical and Translational Research Accelerator, Yale University, New Haven, CT, USA.
- Department of Pediatrics, Section of Nephrology, Yale University, New Haven, CT, USA.
| |
Collapse
|
20
|
Jiang K, Greenberg JH, Abraham A, Xu Y, Schelling JR, Feldman HI, Schrauben SJ, Waikar SS, Shlipak MG, Wettersten N, Coca SG, Vasan RS, Gutierrez OM, Ix JH, Warady BA, Kimmel PL, Bonventre JV, Parikh CR, Mitsnefes MM, Denburg MR, Furth S. Associations of Biomarkers of Kidney Tubule Health, Injury, and Inflammation with Left Ventricular Hypertrophy in Children with CKD. KIDNEY360 2023; 4:1039-1047. [PMID: 37303083 PMCID: PMC10476681 DOI: 10.34067/kid.0000000000000183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 05/23/2023] [Indexed: 06/13/2023]
Abstract
Key Points Higher plasma and urine kidney injury molecule-1, urine monocyte chemoattractant protein-1, and lower urine alpha-1-microglobulin were associated with left ventricular hypertrophy, even after adjustment for confounders. Biomarkers of tubular injury, dysfunction, and inflammation may indicate the severity of kidney pathology and are associated with left ventricular hypertrophy. Background Left ventricular hypertrophy (LVH) is common in children with CKD and is associated with an increased risk of cardiovascular disease and mortality. We have shown that several plasma and urine biomarkers are associated with increased risk of CKD progression. As CKD is associated with LVH, we sought to investigate the association between the biomarkers and LVH. Methods In the CKD in Children Cohort Study, children aged 6 months to 16 years with an eGFR of 30–90 ml/min per 1.73 m2 were enrolled at 54 centers in the United States and Canada. We measured plasma biomarkers kidney injury molecule-1 (KIM-1), tumor necrosis factor receptor-1, tumor necrosis factor receptor-2, soluble urokinase-type plasminogen activator receptor and urine KIM-1, monocyte chemoattractant protein-1 (MCP-1), YKL-40, alpha-1-microglobulin (alpha-1m), and epidermal growth factor in stored plasma and urine collected 5 months after enrollment. Echocardiograms were performed 1 year after enrollment. We assessed the cross-sectional association between the log2 biomarker levels and LVH (left ventricular mass index greater than or equal to the 95th percentile) using a Poisson regression model, adjusted for age, sex, race, body mass index, hypertension, glomerular diagnosis, urine protein-to-creatinine ratio, and eGFR at study entry. Results Among the 504 children, LVH prevalence was 12% (n =59) 1 year after enrollment. In a multivariable-adjusted model, higher plasma and urine KIM-1 and urine MCP-1 concentrations were associated with a higher prevalence of LVH (plasma KIM-1 prevalence ratio [PR] per log2: 1.27, 95% confidence interval [CI], 1.02 to 1.58; urine KIM-1 PR: 1.21, 95% CI, 1.11 to 1.48; and urine MCP-1 PR: 1.18, 95% CI, 1.04 to 1.34). After multivariable adjustment for covariates, lower urine alpha-1m was also associated with a higher prevalence of LVH (PR: 0.90, 95% CI, 0.82 to 0.99). Conclusions Higher plasma and urine KIM-1, urine MCP-1, and lower urine alpha-1m were each associated with LVH prevalence in children with CKD. These biomarkers may better inform risk and help elucidate the pathophysiology of LVH in pediatric CKD.
Collapse
Affiliation(s)
- Kuan Jiang
- Yale School of Medicine, New Haven, Connecticut
| | | | - Alison Abraham
- University of Colorado, Anschutz Medical Campus, Denver, Colorado
- Johns Hopkins University, Baltimore, Maryland
| | - Yunwen Xu
- Johns Hopkins University, Baltimore, Maryland
| | | | - Harold I. Feldman
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sarah J. Schrauben
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | - Steven G. Coca
- Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | - Joachim H. Ix
- University of California San Diego, San Diego, California
| | | | | | | | | | | | | | - Susan Furth
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
21
|
Yang M, Lopez LN, Brewer M, Delgado R, Menshikh A, Clouthier K, Zhu Y, Vanichapol T, Yang H, Harris R, Gewin L, Brooks C, Davidson A, de Caestecker MP. Inhibition of Retinoic Acid Signaling in Proximal Tubular Epithelial cells Protects against Acute Kidney Injury by Enhancing Kim-1-dependent Efferocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.15.545113. [PMID: 37398101 PMCID: PMC10312711 DOI: 10.1101/2023.06.15.545113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Retinoic acid receptor (RAR) signaling is essential for mammalian kidney development, but in the adult kidney is restricted to occasional collecting duct epithelial cells. We now show there is widespread reactivation of RAR signaling in proximal tubular epithelial cells (PTECs) in human sepsis-associated acute kidney injury (AKI), and in mouse models of AKI. Genetic inhibition of RAR signaling in PTECs protects against experimental AKI but is associated with increased expression of the PTEC injury marker, Kim-1. However, Kim-1 is also expressed by de-differentiated, proliferating PTECs, and protects against injury by increasing apoptotic cell clearance, or efferocytosis. We show that the protective effect of inhibiting PTEC RAR signaling is mediated by increased Kim-1 dependent efferocytosis, and that this is associated with de-differentiation, proliferation, and metabolic reprogramming of PTECs. These data demonstrate a novel functional role that reactivation of RAR signaling plays in regulating PTEC differentiation and function in human and experimental AKI. Graphical abstract
Collapse
|
22
|
Kadatane SP, Satariano M, Massey M, Mongan K, Raina R. The Role of Inflammation in CKD. Cells 2023; 12:1581. [PMID: 37371050 DOI: 10.3390/cells12121581] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/02/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic kidney disease (CKD) affects many adults worldwide. Persistent low-grade inflammation is a substantial factor in its development and progression and has correlated with increased mortality and cardiovascular problems. This low-grade inflammation is a product of dysregulation of the normal balance between pro- and anti-inflammatory markers. Various factors such as increased innate immune system activation, reactive oxygen species production, periodontal disease, dysregulation of anti-inflammatory systems and intestinal dysbiosis result in the dysregulation of this balance. Furthermore, this low-grade inflammation has down-effects such as hypertension, renal fibrosis and acceleration of renal function decline. Moreover, low-grade inflammation over time has been linked to malignancy in CKD. As CKD progresses, many patients require dialysis, which has a negative bidirectional relationship with persistent inflammation. Treatment options for inflammation in CKD are vast, including cytokine inhibitors, statins and diets. However, more research is needed to create a standardized management plan. In this review, we will examine the normal physiology of the kidney and its relationship with the immune system. We will then delve into the pathology behind persistent inflammation, the various causes of inflammation, the downstream effects of inflammation, dialysis and potential treatments for inflammation in CKD.
Collapse
Affiliation(s)
| | - Matthew Satariano
- Department of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Michael Massey
- Department of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Kai Mongan
- Department of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Rupesh Raina
- Akron Nephrology Associates/Cleveland Clinic Akron General Medical Center, Akron, OH 44302, USA
- Department of Nephrology, Akron Children's Hospital, Akron, OH 44308, USA
| |
Collapse
|
23
|
Schlosser P, Grams ME, Rhee EP. Proteomics: Progress and Promise of High-Throughput Proteomics in Chronic Kidney Disease. Mol Cell Proteomics 2023; 22:100550. [PMID: 37076045 PMCID: PMC10326701 DOI: 10.1016/j.mcpro.2023.100550] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/20/2023] [Accepted: 03/28/2023] [Indexed: 04/21/2023] Open
Abstract
Current proteomic tools permit the high-throughput analysis of the blood proteome in large cohorts, including those enriched for chronic kidney disease (CKD) or its risk factors. To date, these studies have identified numerous proteins associated with cross-sectional measures of kidney function, as well as with the longitudinal risk of CKD progression. Representative signals that have emerged from the literature include an association between levels of testican-2 and favorable kidney prognosis and an association between levels of TNFRSF1A and TNFRSF1B and worse kidney prognosis. For these and other associations, however, understanding whether the proteins play a causal role in kidney disease pathogenesis remains a fundamental challenge, especially given the strong impact that kidney function can have on blood protein levels. Prior to investing in dedicated animal models or randomized trials, methods that leverage the availability of genotyping in epidemiologic cohorts-including Mendelian randomization, colocalization analyses, and proteome-wide association studies-can add evidence for causal inference in CKD proteomics research. In addition, integration of large-scale blood proteome analyses with urine and tissue proteomics, as well as improved assessment of posttranslational protein modifications (e.g., carbamylation), represent important future directions. Taken together, these approaches seek to translate progress in large-scale proteomic profiling into the promise of improved diagnostic tools and therapeutic target identification in kidney disease.
Collapse
Affiliation(s)
- Pascal Schlosser
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
| | - Morgan E Grams
- Division of Precision Medicine, Department of Medicine, New York University, New York, New York, USA
| | - Eugene P Rhee
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Sandokji I, Xu Y, Denburg M, Furth S, Abraham AG, Greenberg JH. Current and Novel Biomarkers of Progression Risk in Children with Chronic Kidney Disease. Nephron Clin Pract 2023; 148:1-10. [PMID: 37232009 PMCID: PMC10840447 DOI: 10.1159/000530918] [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: 09/15/2022] [Accepted: 02/18/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND Due to the complexity of chronic kidney disease (CKD) pathophysiology, biomarkers representing different mechanistic pathways have been targeted for the study and development of novel biomarkers. The discovery of clinically useful CKD biomarkers would allow for the identification of those children at the highest risk of kidney function decline for timely interventions and enrollment in clinical trials. SUMMARY Glomerular filtration rate and proteinuria are traditional biomarkers to classify and prognosticate CKD progression in clinical practice but have several limitations. Over the recent decades, novel biomarkers have been identified from blood or urine with metabolomic screening studies, proteomic screening studies, and an improved knowledge of CKD pathophysiology. This review highlights promising biomarkers associated with the progression of CKD that could potentially serve as future prognostic markers in children with CKD. KEY MESSAGES Further studies are needed in children with CKD to validate putative biomarkers, particularly candidate proteins and metabolites, for improving clinical management.
Collapse
Affiliation(s)
- Ibrahim Sandokji
- Department of Pediatrics, Taibah University College of Medicine, Medina, Saudi Arabia,
| | - Yunwen Xu
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michelle Denburg
- Division of Nephrology, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Susan Furth
- Division of Nephrology, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alison G Abraham
- Department of Epidemiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jason H Greenberg
- Department of Pediatrics, Section of Nephrology, Clinical and Translational Research Accelerator, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
25
|
Chen J, Tang TT, Cao JY, Li ZL, Zhong X, Wen Y, Shen AR, Liu BC, Lv LL. KIM-1 augments hypoxia-induced tubulointerstitial inflammation through uptake of small extracellular vesicles by tubular epithelial cells. Mol Ther 2023; 31:1437-1450. [PMID: 35982620 PMCID: PMC10188645 DOI: 10.1016/j.ymthe.2022.08.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/29/2022] [Accepted: 08/12/2022] [Indexed: 12/19/2022] Open
Abstract
Tubular epithelial cells (TECs) exposed to hypoxia incite tubulointerstitial inflammation (TII), while the exact mechanism is unclear. In this study, we identified that hypoxia evoked tubule injury as evidenced by tubular hypoxia-inducible factor-1α and kidney injury molecule-1 (KIM-1) expression and that renal small extracellular vesicle (sEV) production was increased with the development of TII after ischemia-reperfusion injury (IRI). Intriguingly, KIM-1-positive tubules were surrounded by macrophages and co-localized with sEVs. In vitro, KIM-1 expression and sEV release were increased in hypoxic TECs and the hypoxia-induced inflammatory response was ameliorated when KIM-1 or Rab27a, a master regulator of sEV secretion, was silenced. Furthermore, KIM-1 was identified to mediate hypoxic TEC-derived sEV (Hypo-sEV) uptake by TECs. Phosphatidylserine (PS), a ligand of KIM-1, was present in Hypo-sEVs as detected by nanoflow cytometry. Correspondingly, the inflammatory response induced by exogenous Hypo-sEVs was attenuated when KIM-1 was knocked down. In vivo, exogenous-applied Hypo-sEVs localized to KIM-1-positive tubules and exacerbated TII in IRI mice. Our study demonstrated that KIM-1 expressed by injured tubules mediated sEV uptake via recognizing PS, which participated in the amplification of tubule inflammation induced by hypoxia, leading to the development of TII in ischemic acute kidney injury.
Collapse
Affiliation(s)
- Jun Chen
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, 87 Ding Jia Qiao Road, Nanjing 210009, China
| | - Tao-Tao Tang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, 87 Ding Jia Qiao Road, Nanjing 210009, China
| | - Jing-Yuan Cao
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, 87 Ding Jia Qiao Road, Nanjing 210009, China
| | - Zuo-Lin Li
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, 87 Ding Jia Qiao Road, Nanjing 210009, China
| | - Xin Zhong
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, 87 Ding Jia Qiao Road, Nanjing 210009, China
| | - Yi Wen
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, 87 Ding Jia Qiao Road, Nanjing 210009, China
| | - An-Ran Shen
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, 87 Ding Jia Qiao Road, Nanjing 210009, China
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, 87 Ding Jia Qiao Road, Nanjing 210009, China.
| | - Lin-Li Lv
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, 87 Ding Jia Qiao Road, Nanjing 210009, China.
| |
Collapse
|
26
|
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
|
27
|
Hanly PJ, Unruh ML. Continuous Positive Airway Pressure Therapy for Chronic Kidney Disease in Patients with Obstructive Sleep Apnea: The Jury Is Still Out. Am J Respir Crit Care Med 2023; 207:657-659. [PMID: 36480960 PMCID: PMC10037486 DOI: 10.1164/rccm.202211-2171ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Patrick J Hanly
- Sleep Centre Foothills Medical Centre Calgary, Alberta, Canada and Cumming School of Medicine University of Calgary Calgary, Alberta, Canada
| | - Mark L Unruh
- Department of Internal Medicine University of New Mexico Albuquerque, New Mexico and Section of Nephrology New Mexico VA Health Care System Albuquerque, New Mexico
| |
Collapse
|
28
|
McCoy IE, Hsu JY, Bonventre JV, Parikh CR, Go AS, Liu KD, Ricardo AC, Srivastava A, Cohen DL, He J, Chen J, Rao PS, Muiru AN, Hsu CY. Absence of long-term changes in urine biomarkers after AKI: findings from the CRIC study. BMC Nephrol 2022; 23:311. [PMID: 36100915 PMCID: PMC9472364 DOI: 10.1186/s12882-022-02937-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] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 08/31/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Mechanisms by which AKI leads to CKD progression remain unclear. Several urine biomarkers have been identified as independent predictors of progressive CKD. It is unknown whether AKI may result in long-term changes in these urine biomarkers, which may mediate the effect of AKI on CKD progression. METHODS We selected 198 episodes of hospitalized AKI (defined as peak/nadir inpatient serum creatinine values ≥ 1.5) among adult participants in the Chronic Renal Insufficiency Cohort (CRIC) Study. We matched the best non-AKI hospitalization (unique patients) for each AKI hospitalization using pre-hospitalization characteristics including eGFR and urine protein/creatinine ratio. Biomarkers were measured in banked urine samples collected at annual CRIC study visits. RESULTS Urine biomarker measurements occurred a median of 7 months before and 5 months after hospitalization. There were no significant differences in the change in urine biomarker-to-creatinine ratio between the AKI and non-AKI groups: KIM-1/Cr + 9% vs + 7%, MCP-1/Cr + 4% vs + 1%, YKL-40/Cr + 7% vs -20%, EGF/Cr -11% vs -8%, UMOD/Cr -2% vs -7% and albumin/Cr + 17% vs + 13% (all p > 0.05). CONCLUSION In this cohort of adults with CKD, AKI did not associate with long-term changes in urine biomarkers.
Collapse
Affiliation(s)
- Ian E McCoy
- Division of Nephrology, University of California San Francisco, Box 0532, 500 Parnassus Ave., MUW418, 94143-0532, San Francisco, CA, USA.
| | - Jesse Y Hsu
- Division of Biostatistics, University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph V Bonventre
- Division of Renal Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Chirag R Parikh
- Division of Nephrology, Johns Hopkins University, Baltimore, MD, USA
| | - Alan S Go
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Kathleen D Liu
- Division of Nephrology, University of California San Francisco, Box 0532, 500 Parnassus Ave., MUW418, 94143-0532, San Francisco, CA, USA
| | - Ana C Ricardo
- Division of Nephrology, University of Illinois, Chicago, IL, USA
| | - Anand Srivastava
- Division of Nephrology and Hypertension, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Debbie L Cohen
- Division of Nephrology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jiang He
- Department of Epidemiology, Tulane University, New Orleans, Louisiana, USA
| | - Jing Chen
- Department of Epidemiology, Tulane University, New Orleans, Louisiana, USA
- Division of Nephrology, Tulane University, New Orleans, Louisiana, USA
| | - Panduranga S Rao
- Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Anthony N Muiru
- Division of Nephrology, University of California San Francisco, Box 0532, 500 Parnassus Ave., MUW418, 94143-0532, San Francisco, CA, USA
| | - Chi-Yuan Hsu
- Division of Nephrology, University of California San Francisco, Box 0532, 500 Parnassus Ave., MUW418, 94143-0532, San Francisco, CA, USA
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| |
Collapse
|
29
|
Zeni L, Norden AGW, Prandi E, Canepa C, Burling K, Simpson K, Felappi B, Plebani A, Cancarini G, Ferraro PM, Fraser D, Unwin RJ. Exploration of a panel of urine biomarkers of kidney disease in two paediatric cohorts with Type 1 diabetes mellitus of differing duration. Diabetol Metab Syndr 2022; 14:71. [PMID: 35550634 PMCID: PMC9097324 DOI: 10.1186/s13098-022-00839-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/26/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND The pathogenesis of diabetic kidney disease (DKD) is complex and involves both glomerular and tubular dysfunction. A global assessment of kidney function is necessary to stage DKD, a progressive kidney disease that is likely to begin in childhood. The present study evaluated whether kidney injury biomarkers identified as early DKD biomarkers in adults have any prognostic value in the very early stages of childhood diabetes. METHODS We measured urine free Retinol-binding protein 4 (UfRBP4), albumin (UAlb), Kidney injury molecule-1 (KIM-1) and the microRNAs miR-155, miR-126 and miR-29b in two cohorts of paediatric T1DM patients without evidence of DKD, but with diabetes of short-duration, ≤ 2.5 years (SD, n = 25) or of long-duration, ≥ 10 years (LD, n = 29); non-diabetic siblings (H, n = 26) were recruited as controls. A p value < 0.05 was considered significant for all results. RESULTS UfRBP4 and UAlb were not significantly different across the three groups. No differences were found in KIM-1 excretion between any of the three groups. UfRBP4 was correlated with UAlb in all three groups (r 0.49; p < 0.001), whereas KIM-1 showed no correlation with albumin excretion. Among microRNAs, miR-29b was higher in all diabetic children compared with the H control group (p = 0.03), whereas miR-155 and miR-126 were not significantly different. No differences were found between the SD and LD groups for all three microRNAs. No associations were identified between these biomarkers with sex, age, BMI, eGFR, T1DM duration or glycaemic control. CONCLUSIONS UfRBP4, KIM-1, miR-155, and miR-126 were unaffected by the presence and duration of diabetes, whereas miR-29b showed a modest elevation in diabetics, regardless of duration. These data support the specificity of a panel of urine biomarkers as DKD biomarkers, rather than any relationship to diabetes per se or its duration, and not as early DKD biomarkers in a paediatric setting.
Collapse
Affiliation(s)
- Letizia Zeni
- Nephrology Unit, Azienda Socio-Sanitaria Territoriale degli Spedali Civili di Brescia, Piazzale Spedali Civili, 1-25123, Brescia, Italy.
| | - Anthony G W Norden
- Department of Renal Medicine, Royal Free Hospital Trust, University College London, Rowland Hill Street, London, NW3 2PF, UK
| | - Elena Prandi
- Pediatrics Clinic, Department of Clinical and Experimental Sciences, University of Brescia and ASST Spedali Civili of Brescia, Piazzale Spedali Civili, 1-25123, Brescia, Italy
| | - Carolina Canepa
- Department of Nephrology, Royal Berkshire Hospital, Reading, UK
| | - Keith Burling
- Core Biochemical Assay Laboratory, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK
| | - Katherine Simpson
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Barbara Felappi
- Pediatrics Clinic, Department of Clinical and Experimental Sciences, University of Brescia and ASST Spedali Civili of Brescia, Piazzale Spedali Civili, 1-25123, Brescia, Italy
| | - Alessandro Plebani
- Pediatrics Clinic, Department of Clinical and Experimental Sciences, University of Brescia and ASST Spedali Civili of Brescia, Piazzale Spedali Civili, 1-25123, Brescia, Italy
| | - Giovanni Cancarini
- Nephrology, Department of Medical and Surgical Specialties, Radiological Science and Public Health, University of Brescia, Brescia, Italy
| | - Pietro Manuel Ferraro
- U.O.S. Terapia Conservativa della Malattia Renale Cronica, U.O.C. Nefrologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Donald Fraser
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Robert J Unwin
- Department of Renal Medicine, Royal Free Hospital Trust, University College London, Rowland Hill Street, London, NW3 2PF, UK
| |
Collapse
|
30
|
Tamma G, Di Mise A, Ranieri M, Centrone M, Venneri M, D'Agostino M, Ferrulli A, Šimunič B, Narici M, Pisot R, Valenti G. Early Biomarkers of Altered Renal Function and Orthostatic Intolerance During 10-day Bedrest. Front Physiol 2022; 13:858867. [PMID: 35514354 PMCID: PMC9065601 DOI: 10.3389/fphys.2022.858867] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Exposure to actual or simulated microgravity results in alterations of renal function, fluid redistribution, and bone loss, which is coupled to a rise of urinary calcium excretion. We provided evidence that high calcium delivery to the collecting duct reduces local Aquaporin 2 (AQP2)-mediated water reabsorption under vasopressin action, thus limiting the maximal urinary concentration to reduce calcium saturation. To investigate early renal adaptation into simulated microgravity, we investigated the effects of 10 days of strict bedrest in 10 healthy volunteers. We report here that 10 days of inactivity are associated with a transient, significant decrease (day 5) in vasopressin (copeptin) paralleled by a decrease in AQP2 excretion, consistent with an increased central volume to the heart, resulting in reduced water reabsorption. Moreover, bedrest caused a significant increase in calciuria secondary to bone demineralization paralleled by a decrease in PTH. Urinary osteopontin, a glycoprotein exerting a protective effect on stone formation, was significantly reduced during bedrest. Moreover, a significant increase in adrenomedullin (day 5), a peptide with vasodepressor properties, was observed at day 5, which may contribute to the known reduced orthostatic capacity post-bedrest. We conclude that renal function is altered in simulated microgravity and is associated with an early increase in the risk of stone formation and reduced orthostatic capacity post-bedrest within a few days of inactivity.
Collapse
Affiliation(s)
- Grazia Tamma
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Annarita Di Mise
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Marianna Ranieri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Mariangela Centrone
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Maria Venneri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Mariagrazia D'Agostino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Angela Ferrulli
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Boštjan Šimunič
- Institute of Kinesiology Research, Science and Research Centre, Koper, Slovenia
| | - Marco Narici
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Rado Pisot
- Institute of Kinesiology Research, Science and Research Centre, Koper, Slovenia
| | - Giovanna Valenti
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| |
Collapse
|
31
|
Alvarez-Llamas G, Santiago-Hernandez A, Ruilope LM. Evidence of chronic kidney injury in patients not meeting KDIGO criteria for CKD. Clin Kidney J 2022; 15:1217-1220. [PMID: 35756746 PMCID: PMC9217645 DOI: 10.1093/ckj/sfac007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Indexed: 11/25/2022] Open
Abstract
Subjects not meeting KDIGO criteria for chronic kidney disease (CKD), i.e. normoalbuminuric (urinary albumin:creatinine ratio, UACR <30 mg/g) individuals with an estimated glomerular filtration rate >60 mL/min/1.73 m2, are considered at no increased cardiovascular or kidney risk associated with kidney disease, but the incidence of subclinical atherosclerosis, cardiovascular events and CKD progression is already increased in the high-normal UACR range (10–30 mg/g). Earlier intervention in this subclinical pre-CKD stage may diminish cardiorenal risk. However, tools to predict albuminuria development and to identify those subjects who will benefit most from intervention are limited. Recent data have identified urine molecular changes within the normoalbuminuria condition, consisting of an altered urinary peptidome, proteome and metabolome, which represent subclinical organ damage and processes such as inflammation, oxidative stress, tricarboxylic acids cycle deregulation, impaired fatty acids β-oxidation or defective tubular reabsorption.
Collapse
Affiliation(s)
- Gloria Alvarez-Llamas
- Immunology Department, IIS-Fundación Jiménez Díaz-UAM, Madrid, Spain
- REDINREN, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Luis M Ruilope
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
- CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain
| |
Collapse
|
32
|
Abstract
Serum creatinine and level of proteinuria, as biomarkers of chronic kidney disease (CKD) progression, inadequately explain the variability of glomerular filtration rate decline, and are late markers of glomerular filtration rate decline. Recent studies have identified plasma and urine biomarkers at higher levels in children with CKD and also associate independently with CKD progression, even after adjustment for serum creatinine and proteinuria. These novel biomarkers represent diverse biologic pathways of tubular injury, tubular dysfunction, inflammation, and tubular health, and can be used as a liquid biopsy to better characterize CKD in children. In this review, we highlight the biomarker findings from the Chronic Kidney Disease in Children cohort, a large longitudinal study of children with CKD, and compare results with those from other pediatric CKD cohorts. The biomarkers in focus in this review include plasma kidney injury molecule-1, monocyte chemoattractant protein-1, fibroblast growth factor-23, tumor necrosis factor receptor-1, tumor necrosis factor receptor-2, soluble urokinase plasminogen activator receptor, and chitinase-3-like protein 1, as well as urine epidermal growth factor, α-1 microglobulin, kidney injury molecule-1, monocyte chemoattractant protein-1, and chitinase-3-like protein 1. Blood and urine biomarkers improve our ability to prognosticate CKD progression and may improve our understanding of CKD pathophysiology. Further research is required to establish how these biomarkers can be used in the clinical setting to improve the clinical management of CKD.
Collapse
Affiliation(s)
- Ibrahim Sandokji
- Section of Nephrology, Clinical and Translational Research Accelerator, Department of Pediatrics, Yale University School of Medicine, New Haven, CT; Department of Pediatrics, Taibah University College of Medicine, Medina, Saudi Arabia
| | - Jason H Greenberg
- Section of Nephrology, Clinical and Translational Research Accelerator, Department of Pediatrics, Yale University School of Medicine, New Haven, CT.
| |
Collapse
|