1
|
Kmezic I, Gustafsson R, Hansson M, Press R. Beta-trace protein in chronic inflammatory demyelinating polyradiculoneuropathy and Guillain-Barré syndrome - clinical utilization and a new insight into pathophysiological mechanisms. J Neurol Sci 2025; 472:123439. [PMID: 40050150 DOI: 10.1016/j.jns.2025.123439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/04/2025] [Accepted: 02/22/2025] [Indexed: 03/09/2025]
Abstract
INTRODUCTION Beta-trace protein (BTP) is primarily used as a marker for traumatic CSF leakage. Spinal leptomeningeal cells adjacent to the spinal nerve roots are a major producer of this protein, which has prompted interest in its relevance to inflammatory polyradiculoneuropathies. BTP has not previously been investigated in patients with Guillain-Barré syndrome (GBS) and chronic inflammatory demyelinating polyradiculopathy (CIDP). MATERIAL AND METHODS BTP was measured in the cerebrospinal fluid (CSF) and plasma in patients with GBS, CIDP, disease-free controls, non-inflammatory polyneuropathies, and multiple sclerosis. RESULTS Pre-treatment levels of BTP in plasma were increased in patients with CIDP, but not GBS compared to all controls. Pre-treatment levels of BTP in CSF were increased in patients with CIDP compared to all controls but in patients with GBS compared only to disease-free controls. In patients with GBS and CIDP, levels of BTP in CSF were increased compared to disease-free controls irrespective of Q.alb, and the levels correlated with axonal damage markers in CSF and plasma. Elevated pre-treatment levels of BTP in CSF predicted therapy failure 3 months after initiation of immunotherapy in CIDP. CONCLUSIONS BTP in CSF and plasma may be a potential biomarker to aid in the diagnosis of CIDP. BTP in CSF could be a potentially more informative biomarker than albumin in a subgroup of patients with GBS and CIDP who have a normal or mildly elevated Q.alb. BTP in CSF may be predictive for therapy response in patient with CIDP, but needs to be validated in larger studies.
Collapse
Affiliation(s)
- Ivan Kmezic
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Rasmus Gustafsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Hansson
- Department of Clinical Chemistry, Karolinska University Hospital, Stockholm, Sweden; Department of Laboratory Medicine H5, Karolinska Institutet, Stockholm, Sweden
| | - Rayomand Press
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
2
|
Mark IT, Brinjikji W, Cutsforth-Gregory J, Verdoorn JT, Benson JC, Madhavan AA, Meeusen JW. β-Trace Protein as a Potential Biomarker for CSF-Venous Fistulas. AJNR Am J Neuroradiol 2025; 46:416-420. [PMID: 39181694 PMCID: PMC11878952 DOI: 10.3174/ajnr.a8476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 08/14/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND AND PURPOSE Accurately identifying patients with CSF-venous fistulas (CVF) causing spontaneous intracranial hypotension, is a diagnostic dilemma. This conundrum underscores the need for a CVF biomarker to help select who should undergo an invasive myelogram for further diagnostic work-up. β-trace protein (BTP) is the most abundant CNS-derived protein in the CSF and, therefore, is a potential venous biomarker for CVF detection. The purpose of our study was to measure venous BTP levels as a potential CVF biomarker. MATERIALS AND METHODS We prospectively enrolled 14 patients with CVFs and measured the BTP in venous blood samples from the paraspinal veins near the CVF and compared those levels with those in the peripheral blood. Myelograms used initially to identify the CVF were evaluated for technique, CVF laterality, CVF level, and the venous drainage pattern. Patient sex and age and symptom duration were also collected. Brain MR images were reviewed for Bern scores. We also measured the peripheral blood BTP levels in 20 healthy controls. RESULTS In patients with CVF, the mean BTP level near the CVF was 54.5% higher (0.760 [SD, 0.673] mg/L versus 0.492 [SD, 0.095] mg/L; P = .069) compared with peripheral blood. Nine (64.3%) patients with CVFs had a higher paraspinal BTP level than peripheral BTP level. The 20 control patients had a higher mean peripheral BTP level of 0.720 (SD, 0.191) mg/L compared with patients with CVF (P < .001). CONCLUSIONS We found that venous blood at the site of the CVF had higher BTP values compared with peripheral blood in most but not all patients with CVF. This finding may reflect the intermittent leaking nature of CVF. Additionally, we found that patients with CVF had a lower peripheral blood BTP level compared with healthy controls. BTP requires further evaluation as a potential CVF biomarker.
Collapse
Affiliation(s)
- Ian T Mark
- From the Department of Radiolog y (I.T.M., W.B., J.T.V., J.C.B., A.A.M.), Mayo Clinic, Rochester, Minnesota
| | - Waleed Brinjikji
- From the Department of Radiolog y (I.T.M., W.B., J.T.V., J.C.B., A.A.M.), Mayo Clinic, Rochester, Minnesota
| | | | - Jared T Verdoorn
- From the Department of Radiolog y (I.T.M., W.B., J.T.V., J.C.B., A.A.M.), Mayo Clinic, Rochester, Minnesota
| | - John C Benson
- From the Department of Radiolog y (I.T.M., W.B., J.T.V., J.C.B., A.A.M.), Mayo Clinic, Rochester, Minnesota
| | - Ajay A Madhavan
- From the Department of Radiolog y (I.T.M., W.B., J.T.V., J.C.B., A.A.M.), Mayo Clinic, Rochester, Minnesota
| | - Jeff W Meeusen
- Department of Laboratory Medicine and Pathology (J.W.M.), Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
3
|
Costa E Silva VT, Gil LA, Inker LA, Caires RA, Costalonga E, Coura-Filho G, Sapienza MT, Castro G, Estevez-Diz MDP, Zanetta DMT, Antonângelo L, Marçal L, Tighiouart H, Miao S, Mathew P, Levey AS, Burdmann EA. Glomerular Filtration Rate Estimation Using β 2-Microglobulin and β-Trace Protein in Adults With Solid Tumors: A Prospective Cross-Sectional Study. Am J Kidney Dis 2024; 84:339-348.e1. [PMID: 38537905 DOI: 10.1053/j.ajkd.2024.01.532] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 12/06/2023] [Accepted: 01/02/2024] [Indexed: 05/26/2024]
Abstract
RATIONALE & OBJECTIVE β2-Microglobulin (B2M) and β-trace protein (BTP) are novel endogenous filtration markers that may improve the accuracy of estimated glomerular filtration rate (eGFR) beyond creatinine and cystatin C (eGFRcr-cys), but they have not been assessed in patients with cancer. STUDY DESIGN Cross-sectional analysis. SETTING & PARTICIPANTS Prospective cohort of 1,200 patients with active solid tumors recruited between April 2015 and September 2017. EXPOSURE CKD-EPI equations without race combining B2M and/or BTP with creatinine with or without cystatin C (2-, 3-, or 4-marker panel eGFR). OUTCOME Performance of equations compared with eGFRcr-cys and non-GFR determinants of serum B2M and BTP (SB2M, and SBTP, respectively). Measured GFR (mGFR) was determined using the plasma clearance of chromium-51 labeled ethylenediamine tetraacetic acid (51Cr-EDTA). ANALYTICAL APPROACH Bias was defined as the median of the differences between mGFR and eGFR, and 1-P30 was defined as the percentage of estimates that differed by more than 30% from the mGFR (1-P30). Linear regression was used to assess association of clinical and laboratory variables with SB2M, and SBTP after adjustment for mGFR. RESULTS Mean age and mGFR were 58.8±13.2 SD years and 78.4±21.7 SD mL/min/1.73m2, respectively. Performance of the 3-marker and 4-marker panel equations was better than eGFRcr-cys (lesser bias and 1-P30). Performance of 2-marker panel equations was as good as eGFRcr-cys (lesser bias and similar 1-P30). SB2M and SBTP were not strongly influenced by cancer site. LIMITATIONS Participants may have had better clinical performance status than the general population of patients with solid tumors. CONCLUSIONS B2M and BTP can improve the accuracy of eGFR and may be useful as confirmatory tests in patients with solid tumors, either by inclusion in a multimarker panel equation with creatinine and cystatin C, or by substituting for cystatin C in combination with creatinine. PLAIN-LANGUAGE SUMMARY The most accurate method to assess estimate kidney function is estimated glomerular filtration rate (eGFR) using creatinine and cystatin C (eGFRcr-cys). We studied whether using β2-microglobulin (B2M) and/or β-trace protein (BTP) with creatinine with or without cystatin C (2-, 3-, or 4-marker panel eGFR) might be useful in patients with active solid tumors. The performance of the 3-marker and 4-marker panel equations was better than eGFRcr-cys. Performance of 2-marker panel equations was as good as eGFRcr-cys. We conclude that B2M and BTP can improve the accuracy of eGFR and may be useful as a confirmatory test in patients with solid tumors either by inclusion in multimarker panel equation with creatinine and cystatin C or by substituting for cystatin C in combination with creatinine.
Collapse
Affiliation(s)
- Verônica T Costa E Silva
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil; Laboratório de Investigação Médica (LIM) 16, Serviço de Nefrologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
| | - Luiz A Gil
- Laboratório de Investigação Médica (LIM) 66, Serviço de Geriatria, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Lesley A Inker
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Renato A Caires
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Elerson Costalonga
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - George Coura-Filho
- Serviço de Medicina Nuclear, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Marcelo T Sapienza
- Radiology and Oncology Department, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Gilberto Castro
- Serviço de Oncologia Clínica, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Maria D P Estevez-Diz
- Serviço de Oncologia Clínica, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Dirce Maria T Zanetta
- Department of Epidemiology, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Leila Antonângelo
- Laboratório de Investigação Médica (LIM) 03, Division of Clinical Pathology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Lia Marçal
- Laboratório de Investigação Médica (LIM) 03, Division of Clinical Pathology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Hocine Tighiouart
- Institute for Clinical Research and Health Policy Studies, Biostatistics, Epidemiology, and Research Design Center, Tufts Medical Center, Boston, Massachusetts
| | - Shiyuan Miao
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Paul Mathew
- Division of Hematology-Oncology, Tufts Medical Center, Boston, Massachusetts
| | - Andrew S Levey
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Emmanuel A Burdmann
- Laboratório de Investigação Médica (LIM) 12, Serviço de Nefrologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
4
|
Huang PY, Hsu BG, Wang CH, Tsai JP. The Prognostic Role of Serum β-Trace Protein Levels among Patients on Maintenance Hemodialysis. Diagnostics (Basel) 2024; 14:974. [PMID: 38786272 PMCID: PMC11119092 DOI: 10.3390/diagnostics14100974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024] Open
Abstract
Cardiovascular (CV) diseases are the most commonly encountered etiology of mortality in patients having kidney failure. β-Trace protein (BTP) is a biomarker of glomerular filtration function as well as a potential predictor of adverse CV outcomes. This study aimed to determine the prognostic value of BTP in patients on chronic hemodialysis (HD). A total of 96 patients undergoing HD were enrolled. Baseline variables were collected, and the patients were tracked for 3 years. Twenty-five patients died at 3 years. Those who experienced mortality were noted to have higher serum concentrations of BTP and a higher incidence of diabetes mellitus (DM). The area under the receiver operating characteristic curve for serum BTP distinguishing mortality from survival was 0.659 (95% confidence interval [CI], 0.555-0.752; p = 0.027). After the adjustment of variables potentially affecting survival rates, BTP levels above the median (adjusted hazard ratio [aHR]: 2.913, 95% CI, 1.256-6.754; p = 0.013), the presence of DM (aHR: 2.474, 95% CI, 1.041-5.875; p = 0.040), and low serum albumin (aHR: 0.298, 95% CI, 0.110-0.806; p = 0.017) independently correlated with survival in HD patients. Serum BTP is a novel biomarker for predicting overall outcomes in HD patients.
Collapse
Affiliation(s)
- Po-Yu Huang
- Division of Nephrology, Department of Internal Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi 62247, Taiwan;
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan
| | - Bang-Gee Hsu
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan; (B.-G.H.); (C.-H.W.)
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Chih-Hsien Wang
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan; (B.-G.H.); (C.-H.W.)
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Jen-Pi Tsai
- Division of Nephrology, Department of Internal Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi 62247, Taiwan;
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| |
Collapse
|
5
|
Vermassen T, Geboes K, Lumen N, Van Praet C, Rottey S, Delanghe J. Comparison of different estimated glomerular filtration rates for monitoring of kidney function in oncology patients. Clin Kidney J 2024; 17:sfae006. [PMID: 38288036 PMCID: PMC10823486 DOI: 10.1093/ckj/sfae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Indexed: 01/31/2024] Open
Abstract
Background Tyrosine kinase inhibitors (TKIs) are associated with kidney function deterioration. A shift is ongoing towards glomerular filtration rate (GFR) equations based on other protein markers, such as cystatin C (CSTC) and β-trace protein (BTP). We evaluated various GFR equations for monitoring of kidney function in actively treated oncology patients. Methods We monitored 110 patients receiving a TKI. Blood and urine were collected during therapy. Serum analysis included creatinine (Cr), CSTC and BTP; for consequent GFR determination. Urine was analysed for protein, albumin, immunoglobulin G, and α-1-microglobulin. A similar analysis was done in a patient subgroup receiving immune checkpoint inhibitors (ICI) as prior or subsequent line of therapy. Results Cr remained constant during TKI treatment (P = 0.7753), whereas a significant decrease in CSTC (from week 2 onward, P < 0.0001) and BTP (at weeks 2 and 4, P = 0.0100) were noticed. Consequently, GFR estimations, using CSTC and/or BTP as a biochemical parameter, showed an apparent increase in GFR, whereas this was not observed for Cr-related GFR estimations. As a result, the GFR gap (ΔGFR) was significantly different from week 2 onward between Cr-based and CSTC-based GFR and between BTP-based and CSTC-based GFR. Glomerular damage was noticed with significant increase in urine protein-to-creatinine ratio, albumin-to-creatinine ratio and immunoglobulin G (all P < 0.0001). No change in α-1-microglobulin was seen. ICI treatment had no effect on Cr (P = 0.2262), CSTC (P = 0.7341), and BTP concentrations (P = 0.3592). Conclusion GFR equations, in which CSTC is incorporated, fail to correctly estimate the GFR in oncology patients treated with TKIs. As TKI-treated patients show clear signs of glomerular injury, further assessment is needed on how to correctly monitor the kidney function in actively treated oncology patients.
Collapse
Affiliation(s)
- Tijl Vermassen
- Department of Medical Oncology, University Hospital Ghent, Ghent, Belgium
- Biomarkers in Cancer, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Karen Geboes
- Cancer Research Institute Ghent, Ghent, Belgium
- Digestive Oncology, Department of Gastroenterology, Ghent University Hospital, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Nicolaas Lumen
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Urology, University Hospital Ghent, Ghent, Belgium
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Charles Van Praet
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Urology, University Hospital Ghent, Ghent, Belgium
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Sylvie Rottey
- Department of Medical Oncology, University Hospital Ghent, Ghent, Belgium
- Biomarkers in Cancer, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
- Drug Research Unit Ghent, University Hospital Ghent, Ghent, Belgium
| | - Joris Delanghe
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
6
|
Kasozi RN, Meeusen JW, Lieske JC. Estimating glomerular filtration rate with new equations: can one size ever fit all? Crit Rev Clin Lab Sci 2023; 60:549-559. [PMID: 37259709 PMCID: PMC10592396 DOI: 10.1080/10408363.2023.2214812] [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: 02/27/2023] [Revised: 04/25/2023] [Accepted: 05/13/2023] [Indexed: 06/02/2023]
Abstract
Glomerular filtration rate (GFR) is thought to be the best overall indicator of kidney health. On an individual patient basis, a working knowledge of GFR is important to understand the future risk for chronic kidney disease (CKD) progression, enhanced risk for cardiovascular disease and death, and for optimal medical management including the dosing of certain drugs. Although GFR can be directly measured using exogenous compounds that are eliminated by the kidney, these methods are not scalable for repeated and routine use in clinical care. Thus, in most circumstances GFR is estimated, termed estimated GFR (eGFR), using serum biomarkers that are eliminated by the kidney. Of these, serum creatinine, and to a lesser extent cystatin C, are most widely employed. However, the resulting number is simply a population average for an individual of that age and sex with a given serum creatinine and/or cystatin C, while the range of potential GFR values is actually quite large. Thus, it is important to consider characteristics of a given patient that might make this estimate better or worse in a particular case. In some circumstances, cystatin C or creatinine might be the better choice. Ultimately it is difficult, if not impossible, to have an eGFR equation that performs equally well in all populations. Thus, in certain cases it might be appropriate to directly measure GFR for high consequence medical decision-making, such as approval for kidney donation or prior to certain chemotherapeutic regimens. In all cases, the eGFR thresholds of CKD stage should not be viewed as absolute numbers. Thus, clinical care should not be determined solely by CKD stage as determined by eGFR alone, but rather by the combination of an individual patient's likely kidney function together with their current clinical situation.
Collapse
Affiliation(s)
- Ramla N. Kasozi
- Department of Family Medicine, Mayo Clinic, Jacksonville, FL
| | - Jeffrey W. Meeusen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - John C. Lieske
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
- Department of Internal Medicine, Division of Nephrology and Hypertension Mayo Clinic, Rochester, MN
| |
Collapse
|
7
|
Kure N, Krogstrup NV, Oltean M, Jespersen B, Birn H, Nielsen MB. β-Trace Protein and β2-Microglobulin do not Improve Estimation of Glomerular Filtration Rate in Kidney Transplant Recipients Compared With Creatinine and Cystatin C. Transplant Proc 2023; 55:2071-2078. [PMID: 37806869 DOI: 10.1016/j.transproceed.2023.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/16/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Reliable estimates of glomerular filtration rate (eGFR) are important for detecting changes in graft function in kidney transplant recipients. Current eGFR equations are based on plasma creatinine and/or cystatin C; however, these are associated with significant bias. This study investigated if equations based on β-trace protein (BTP) and β2-microglobulin (B2M) performed better than the 2021 Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equations based on creatinine and cystatin C among kidney transplant recipients. METHODS We included samples and data from the clinical trial CONTEXT. Glomerular filtration rate (GFR) was measured by plasma clearance of an exogenous marker. The eGFR was calculated using the CKD-EPI equations for estimating GFR from BTP and/or B2M and the 2021 CKD-EPI creatinine and creatinine-cystatin C equations. The GFR estimates were evaluated 3 (n = 82) and 12 (n = 64) months after transplant using mean bias, precision, and accuracy. Furthermore, we analyzed the ability of the equations to correctly classify the direction of changes in measured GFR from 3 to 12 months. RESULTS Among the BTP- and B2M-based equations, the combined eGFR-BTP-B2M performed best with respect to precision (SD = 7.64 mL/min/1.73 m2) and accuracy (±10% from measured GFR = 36%). The eGFR-BTP-B2M and the eGFR-creatinine-cystatin C (2021) performed similarly when comparing precision, accuracy, and residuals (P = .481). The BTP- and/or B2M-based equations did not perform better than the eGFR-creatinine-cystatin C (2021) in correctly classifying the direction of changes in measured GFR from 3 to 12 months. CONCLUSIONS β-trace protein and/or B2M do not improve the estimation of GFR when compared with creatinine- and cystatin C-based 2021 CKD-EPI equations.
Collapse
Affiliation(s)
- Nathalie Kure
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Nicoline V Krogstrup
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark; Department of Renal Medicine, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mihai Oltean
- The Transplant Institute, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Bente Jespersen
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Henrik Birn
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Marie Bodilsen Nielsen
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
8
|
Chen Q, Jiang H, Ding R, Zhong J, Li L, Wan J, Feng X, Peng L, Yang X, Chen H, Wang A, Jiao J, Yang Q, Chen X, Li X, Shi L, Zhang G, Wang M, Yang H, Li Q. Cell-type-specific molecular characterization of cells from circulation and kidney in IgA nephropathy with nephrotic syndrome. Front Immunol 2023; 14:1231937. [PMID: 37908345 PMCID: PMC10613708 DOI: 10.3389/fimmu.2023.1231937] [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: 05/31/2023] [Accepted: 09/20/2023] [Indexed: 11/02/2023] Open
Abstract
Nephrotic syndrome (NS) is a relatively rare and serious presentation of IgA nephropathy (IgAN) (NS-IgAN). Previous research has suggested that the pathogenesis of NS-IgAN may involve circulating immune imbalance and kidney injury; however, this has yet to be fully elucidated. To investigate the cellular and molecular status of NS-IgAN, we performed single-cell RNA sequencing (scRNA-seq) of peripheral blood mononuclear cells (PBMCs) and kidney cells from pediatric patients diagnosed with NS-IgAN by renal biopsy. Consistently, the proportion of intermediate monocytes (IMs) in NS-IgAN patients was higher than in healthy controls. Furthermore, flow cytometry confirmed that IMs were significantly increased in pediatric patients with NS. The characteristic expression of VSIG4 and MHC class II molecules and an increase in oxidative phosphorylation may be important features of IMs in NS-IgAN. Notably, we found that the expression level of CCR2 was significantly increased in the CMs, IMs, and NCMs of patients with NS-IgAN. This may be related to kidney injury. Regulatory T cells (Tregs) are classified into two subsets of cells: Treg1 (CCR7 high, TCF7 high, and HLA-DR low) and Treg2 (CCR7 low, TCF7 low, and HLA-DR high). We found that the levels of Treg2 cells expressed significant levels of CCR4 and GATA3, which may be related to the recovery of kidney injury. The state of NS in patients was closely related to podocyte injury. The expression levels of CCL2, PRSS23, and genes related to epithelial-mesenchymal transition were significantly increased in podocytes from NS-IgAN patients. These represent key features of podocyte injury. Our analysis suggests that PTGDS is significantly downregulated following injury and may represent a new marker for podocytes. In this study, we systematically analyzed molecular events in the circulatory system and kidney tissue of pediatric patients with NS-IgAN, which provides new insights for targeted therapy in the future.
Collapse
Affiliation(s)
- Qilin Chen
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Huimin Jiang
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Rong Ding
- Nanjing Jiangbei New Area Biopharmaceutical Public Service Platform Co. Ltd, Nanjing, Jiangsu, China
| | - Jinjie Zhong
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Longfei Li
- Nanjing Jiangbei New Area Biopharmaceutical Public Service Platform Co. Ltd, Nanjing, Jiangsu, China
| | - Junli Wan
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Xiaoqian Feng
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Liping Peng
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xia Yang
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Han Chen
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Anshuo Wang
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Jia Jiao
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Qin Yang
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Xuelan Chen
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Xiaoqin Li
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Lin Shi
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Gaofu Zhang
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Mo Wang
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Haiping Yang
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Qiu Li
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
9
|
Iversen E, Walls AB, Petersen A, Jensen PS, Kallemose T, Andersen A, Nielsen RL, Bengaard AK, Juul-Larsen HG, Bornaes O, Damgaard M, Andersen O, Tavenier J, Houlind MB. Estimated glomerular filtration rate based on creatinine, cystatin C, β-trace protein and β2 microglobulin in patients undergoing nontraumatic lower extremity amputation. Br J Clin Pharmacol 2023; 89:1789-1798. [PMID: 36511684 DOI: 10.1111/bcp.15639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 10/28/2022] [Accepted: 12/02/2022] [Indexed: 12/15/2022] Open
Abstract
AIMS The study's aim is to compare current and new equations for estimating glomerular filtration rate (GFR) based on creatinine, cystatin C, β-trace protein (BTP) and β2 microglobulin (B2M) among patients undergoing major amputation. METHODS This is a secondary analysis of data from a prospective cohort study investigating patients undergoing nontraumatic lower extremity amputation. Estimated GFR (eGFR) was calculated using equations based on creatinine (eGFRcre[2009] and eGFRcre[2021]), cystatin C (eGFRcys), the combination of creatinine and cystatin C (eGFRcomb[2012] and eGFRcomb[2021]) or a panel of all 4 filtration markers (eGFRpanel). Primary outcome was changed in eGFR across amputation according to each equation. Two case studies of prior amputation with GFR measured by 99mTc-DTPA clearance are described to illustrate the relative accuracies of each eGFR equation. RESULTS Analysis of the primary outcome included 29 patients (median age 75 years, 31% female). Amputation was associated with a significant decrease in creatinine concentration (-0.09 mg/dL, P = 0.004), corresponding to a significant increase in eGFRcre[2009] (+6.1 mL/min, P = 0.006) and eGFRcre[2021] (+6.3 mL/min, P = 0.006). Change across amputation was not significant for cystatin C, BTP, B2M or equations incorporating these markers (all P > 0.05). In both case studies, eGFRcre[2021] yielded the largest positive bias, eGFRcys yielded the largest negative bias and eGFRcomb[2012] and eGFRcomb[2021] yielded the smallest absolute bias. CONCLUSION Creatinine-based estimates were substantially higher than cystatin C-based estimates before amputation and significantly increased across amputation. Estimates combining creatinine and cystatin were stable across amputation, while the addition of BTP and B2M is unlikely to be clinically relevant.
Collapse
Affiliation(s)
- Esben Iversen
- Department of Clinical Research, Copenhagen University Hospital Amager & Hvidovre, Hvidovre, Denmark
| | - Anne Byriel Walls
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- The Capital Region Pharmacy, Herlev, Denmark
| | - Annamarie Petersen
- Department of Clinical Research, Copenhagen University Hospital Amager & Hvidovre, Hvidovre, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- The Capital Region Pharmacy, Herlev, Denmark
| | - Pia Søe Jensen
- Department of Clinical Research, Copenhagen University Hospital Amager & Hvidovre, Hvidovre, Denmark
- The Research Unit of Orthopedic Nursing, Department of Orthopedic Surgery, Copenhagen University Hospital Amager & Hvidovre, Hvidovre, Denmark
| | - Thomas Kallemose
- Department of Clinical Research, Copenhagen University Hospital Amager & Hvidovre, Hvidovre, Denmark
| | - Aino Andersen
- Department of Clinical Research, Copenhagen University Hospital Amager & Hvidovre, Hvidovre, Denmark
| | - Rikke Lundsgaard Nielsen
- Department of Clinical Research, Copenhagen University Hospital Amager & Hvidovre, Hvidovre, Denmark
| | - Anne Kathrine Bengaard
- Department of Clinical Research, Copenhagen University Hospital Amager & Hvidovre, Hvidovre, Denmark
- The Capital Region Pharmacy, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Helle Gybel Juul-Larsen
- Department of Clinical Research, Copenhagen University Hospital Amager & Hvidovre, Hvidovre, Denmark
| | - Olivia Bornaes
- Department of Clinical Research, Copenhagen University Hospital Amager & Hvidovre, Hvidovre, Denmark
| | - Morten Damgaard
- Department of Clinical Physiology and Nuclear Medicine, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager & Hvidovre, Hvidovre, Denmark
| | - Ove Andersen
- Department of Clinical Research, Copenhagen University Hospital Amager & Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Emergency Department, Copenhagen University Hospital Amager & Hvidovre, Hvidovre, Denmark
| | - Juliette Tavenier
- Department of Clinical Research, Copenhagen University Hospital Amager & Hvidovre, Hvidovre, Denmark
| | - Morten Baltzer Houlind
- Department of Clinical Research, Copenhagen University Hospital Amager & Hvidovre, Hvidovre, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- The Capital Region Pharmacy, Herlev, Denmark
| |
Collapse
|
10
|
Schwab S, Pörner D, Kleine CE, Werberich R, Werberich L, Reinhard S, Bös D, Strassburg CP, von Vietinghoff S, Lutz P, Woitas RP. NT-proBNP as predictor of major cardiac events after renal transplantation in patients with preserved left ventricular ejection fraction. BMC Nephrol 2023; 24:32. [PMID: 36774457 PMCID: PMC9922448 DOI: 10.1186/s12882-023-03082-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/06/2023] [Indexed: 02/13/2023] Open
Abstract
BACKGROUND For the improvement of outcome after renal transplantation it is important to predict future risk of major adverse cardiac events as well as all-cause mortality. We aimed to determine the relationship of pre-transplant NT-proBNP with major adverse cardiac events and all-cause mortality after transplant in patients on the waiting-list with preserved left ventricular ejection fraction. PATIENTS AND METHODS We included 176 patients with end-stage renal disease and preserved left ventricular ejection fraction who received a kidney transplant. MACE was defined as myocardial infarction (ST-segment elevation [STEMI] or non-ST-segment elevation [NSTEMI]), stroke or transient ischemic attack), coronary artery disease requiring intervention or bypass or death from cardiovascular causes. RESULTS MACE occurred in 28/176 patients. Patients with NT-proBNP levels above 4350 pg/ml had 1- and 5-year survival rates of 90.67% and 68.20%, whereas patients with NT-proBNP levels below 4350 pg/ml had 1- and 5-year survival rates of 100% and 90.48% (p < 0.01). 1- and 5-year MACE-free survival rates were calculated as 78.82% and 74.68% for patients with NT-proBNP > 4350 pg/ml and 93.33% and 91.21% for patients with NT-proBNP < 4350 pg/ml (p < 0.01). CONCLUSIONS Pre-transplant NT-proBNP might identify renal transplant candidates at risk for MACE after transplant.
Collapse
Affiliation(s)
- Sebastian Schwab
- Department of Internal Medicine I, Nephrology Section, University of Bonn, Bonn, Germany.
| | - Daniel Pörner
- grid.10388.320000 0001 2240 3300Department of Internal Medicine I, Nephrology Section, University of Bonn, Bonn, Germany
| | - Carola-Ellen Kleine
- grid.10388.320000 0001 2240 3300Department of Internal Medicine I, Nephrology Section, University of Bonn, Bonn, Germany
| | - Roxana Werberich
- grid.10388.320000 0001 2240 3300Department of Internal Medicine I, Nephrology Section, University of Bonn, Bonn, Germany
| | - Louisa Werberich
- grid.10388.320000 0001 2240 3300Department of Internal Medicine I, Nephrology Section, University of Bonn, Bonn, Germany
| | - Stephan Reinhard
- grid.10388.320000 0001 2240 3300Department of Internal Medicine I, Nephrology Section, University of Bonn, Bonn, Germany
| | - Dominik Bös
- Kuratorium for Dialysis, KfH Renal Center, Bonn, Germany
| | - Christian P. Strassburg
- grid.10388.320000 0001 2240 3300Department of Internal Medicine I, Nephrology Section, University of Bonn, Bonn, Germany
| | - Sibylle von Vietinghoff
- grid.10388.320000 0001 2240 3300Department of Internal Medicine I, Nephrology Section, University of Bonn, Bonn, Germany
| | - Philipp Lutz
- grid.10388.320000 0001 2240 3300Department of Internal Medicine I, Nephrology Section, University of Bonn, Bonn, Germany
| | | |
Collapse
|
11
|
Tardif G, Paré F, Gotti C, Roux-Dalvai F, Droit A, Zhai G, Sun G, Fahmi H, Pelletier JP, Martel-Pelletier J. Mass spectrometry-based proteomics identify novel serum osteoarthritis biomarkers. Arthritis Res Ther 2022; 24:120. [PMID: 35606786 PMCID: PMC9125906 DOI: 10.1186/s13075-022-02801-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 05/08/2022] [Indexed: 11/10/2022] Open
Abstract
Background Osteoarthritis (OA) is a slowly developing and debilitating disease, and there are no validated specific biomarkers for its early detection. To improve therapeutic approaches, identification of specific molecules/biomarkers enabling early determination of this disease is needed. This study aimed at identifying, with the use of proteomics/mass spectrometry, novel OA-specific serum biomarkers. As obesity is a major risk factor for OA, we discriminated obesity-regulated proteins to target only OA-specific proteins as biomarkers. Methods Serum from the Osteoarthritis Initiative cohort was used and divided into 3 groups: controls (n=8), OA-obese (n=10) and OA-non-obese (n=10). Proteins were identified and quantified from the liquid chromatography–tandem mass spectrometry analyses using MaxQuant software. Statistical analysis used the Limma test followed by the Benjamini-Hochberg method. To compare the proteomic profiles, the multivariate unsupervised principal component analysis (PCA) followed by the pairwise comparison was used. To select the most predictive/discriminative features, the supervised linear classification model sparse partial least squares regression discriminant analysis (sPLS-DA) was employed. Validation of three differential proteins was performed with protein-specific assays using plasma from a cohort derived from the Newfoundland Osteoarthritis. Results In total, 509 proteins were identified, and 279 proteins were quantified. PCA-pairwise differential comparisons between the 3 groups revealed that 8 proteins were differentially regulated between the OA-obese and/or OA-non-obese with controls. Further experiments using the sPLS-DA revealed two components discriminating OA from controls (component 1, 9 proteins), and OA-obese from OA-non-obese (component 2, 23 proteins). Proteins from component 2 were considered related to obesity. In component 1, compared to controls, 7 proteins were significantly upregulated by both OA groups and 2 by the OA-obese. Among upregulated proteins from both OA groups, some of them alone would not be a suitable choice as specific OA biomarkers due to their rather non-specific role or their strong link to other pathological conditions. Altogether, data revealed that the protein CRTAC1 appears to be a strong OA biomarker candidate. Other potential new biomarker candidates are the proteins FBN1, VDBP, and possibly SERPINF1. Validation experiments revealed statistical differences between controls and OA for FBN1 (p=0.044) and VDPB (p=0.022), and a trend for SERPINF1 (p=0.064). Conclusion Our study suggests that 4 proteins, CRTAC1, FBN1, VDBP, and possibly SERPINF1, warrant further investigation as potential new biomarker candidates for the whole OA population. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02801-1.
Collapse
Affiliation(s)
- Ginette Tardif
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), 900 Saint-Denis, Suite R11.412B, Montreal, QC, H2X 0A9, Canada
| | - Frédéric Paré
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), 900 Saint-Denis, Suite R11.412B, Montreal, QC, H2X 0A9, Canada
| | - Clarisse Gotti
- CHU de Québec Research Center, Laval University, Quebec, QC, G1V 4G2, Canada
| | | | - Arnaud Droit
- CHU de Québec Research Center, Laval University, Quebec, QC, G1V 4G2, Canada
| | - Guangju Zhai
- Division of Biomedical Sciences (Genetics), Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Guang Sun
- Discipline of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Hassan Fahmi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), 900 Saint-Denis, Suite R11.412B, Montreal, QC, H2X 0A9, Canada
| | - Jean-Pierre Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), 900 Saint-Denis, Suite R11.412B, Montreal, QC, H2X 0A9, Canada
| | - Johanne Martel-Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), 900 Saint-Denis, Suite R11.412B, Montreal, QC, H2X 0A9, Canada.
| |
Collapse
|
12
|
MSC-Derived Extracellular Vesicle-Delivered L-PGDS Inhibit Gastric Cancer Progression by Suppressing Cancer Cell Stemness and STAT3 Phosphorylation. Stem Cells Int 2022; 2022:9668239. [PMID: 35087591 PMCID: PMC8789473 DOI: 10.1155/2022/9668239] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/29/2021] [Accepted: 12/14/2021] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cell- (MSC-) derived extracellular vesicles (EVs) serving as delivery system have attracted extensive research interest, especially in cancer therapy. In our previous study, lipocalin-type prostaglandin D2 synthase (L-PGDS) showed inhibitory effects on gastric cancer growth. In this study, we aimed to explore whether MSC-EV-delivered L-PGDS (EVs-L-PGDS) could inhibit gastric cancer progression. EVs-L-PGDS were generated from MSCs transfected with adenovirus encoding L-PGDS. Cell colony-forming, migration, invasion, and flow cytometry assays were used to show the inhibitory effects of EVs on tumor cells in vitro, and the nude mouse subcutaneous tumor model was performed to show the inhibitory effect of EVs on tumor progression in vivo. In vitro, EVs-L-PGDS could be internalized and inhibit the colony-forming, migration, and invasion ability of gastric cancer cell SGC-7901 and promote cell apoptosis. In vivo, EVs-L-PGDS inhibited the tumor growth in nude mouse subcutaneous tumor-bearing model. Compared with the PBS and EVs containing empty vector (EVs-Vector) group, more apoptotic cells and higher L-PGDS expression were detected in tumor tissue of the EVs-L-PGDS treatment group. And these differences are significant. Mechanistically, EVs-L-PGDS reduced the expression of stem cell markers including Oct4, Nanog, and Sox2 and inhibited STAT3 phosphorylation in gastric cancer cell SGC-7901. In conclusion, our results imply that MSC-derived EVs could be utilized as an effective nanovehicle to deliver L-PGDS for gastric cancer treatment, which provides a novel idea for the EV-based cancer therapy.
Collapse
|
13
|
Chang AR, Chen J, Grams ME, Karger AB, Inker LA, Coresh J, Levey AS. β2-Microglobulin and β-Trace Protein in Patients Undergoing Bariatric Surgery: Non-GFR Determinants and Panel-estimated GFR Performance. Kidney Med 2021; 4:100401. [PMID: 35243311 PMCID: PMC8861947 DOI: 10.1016/j.xkme.2021.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Alex R. Chang
- Kidney Health Research Institute, Geisinger, Danville, Pennsylvania
- Department of Population Health Sciences, Geisinger, Danville, Pennsylvania
- Address for Correspondence: Alex R. Chang, MD, MS, Kidney Health Research Institute, Geisinger, 100 N Academy Ave, Danville, PA 17822.
| | - Jingsha Chen
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland
| | - Morgan E. Grams
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland
| | - Amy B. Karger
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Lesley A. Inker
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Josef Coresh
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland
| | - Andrew S. Levey
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| |
Collapse
|
14
|
Lafer I, Michaelis S, Schneider C, Baranyi A, Schnedl WJ, Holasek S, Zelzer S, Niedrist T, Meinitzer A, Enko D. Beta-trace protein concentrations at the blood-cerebrospinal fluid barrier - acute phase affects protein status. EXCLI JOURNAL 2021; 20:1446-1452. [PMID: 34737686 PMCID: PMC8564902 DOI: 10.17179/excli2021-4148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 08/31/2021] [Indexed: 11/10/2022]
Affiliation(s)
- Ingrid Lafer
- Department of Internal Medicine, General Hospital Hochsteiermark, Mürzzuschlag, Austria
| | - Simon Michaelis
- Institute of Clinical Chemistry and Laboratory Medicine, General Hospital Hochsteiermark, Leoben, Austria
| | - Christopher Schneider
- Institute of Clinical Chemistry and Laboratory Medicine, General Hospital Hochsteiermark, Leoben, Austria
| | - Andreas Baranyi
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | | | - Sandra Holasek
- Department of Immunology and Pathophysiology, Medical University of Graz, Otto Loewi Research Center, Graz, Austria
| | - Sieglinde Zelzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Tobias Niedrist
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Andreas Meinitzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Dietmar Enko
- Institute of Clinical Chemistry and Laboratory Medicine, General Hospital Hochsteiermark, Leoben, Austria.,Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| |
Collapse
|
15
|
Baysoy A, Karakoyun I, Arslan FD, Basok BI, Colak A, Duman C. Biological variation data for kidney function related parameter: serum beta trace protein, creatinine and cystatin C from 22 apparently healthy Turkish subjects. Clin Chem Lab Med 2021; 60:584-592. [PMID: 34506692 DOI: 10.1515/cclm-2021-0543] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 08/30/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Biological variation is defined as the variation in analytical concentration between and within individuals, and being aware of this biological variation is important for understanding disease dynamics. The aim of our study is to calculate the within-subject (CVI) and between-subject (CVG) biological variations of serum creatinine, cystatin C and Beta trace protein (BTP), as well as the reference change value (RCV) and individuality indexes (II), which are used to calculate the glomerular filtration rate while evaluating kidney damage. METHODS Blood samples were collected from 22 healthy volunteers for 10 consecutive weeks and stored at -80 °C until the day of analysis. While the analysis for serum creatinine was performed colorimetrically with the kinetic jaffe method, the nephelometric method was employed for cystatin C and BTP measurements. All analyses were carried out in a single session for each test. RESULTS Analytical coefficient of variation (CVA) for serum creatinine, cystatin C and beta trace protein was 5.56, 3.48 and 5.37%, respectively. CVI and CVG: for serum creatinine: 3.31, 14.50%, respectively, for cystatin C: 3.15, 12.24%, respectively, for BTP: 9.91, 14.36%, respectively. RCV and II were calculated as 17.94%, 0.23 for serum creatinine, 13.01%, 0.26 for cystatin C, 31.24%, 0.69 for BTP, respectively. CONCLUSIONS According to the data obtained in our study, serum creatinine and cystatin C show high individuality, therefore we think that the use of RCV instead of reference ranges would be appropriate. Although II is found to be low for BTP, more studies are needed to support this finding.
Collapse
Affiliation(s)
- Anil Baysoy
- Department of Medical Biochemistry, Tepecik Training and Research Hospital, University of Health Sciences, Izmir, Turkey
| | - Inanc Karakoyun
- Department of Medical Biochemistry, Tepecik Training and Research Hospital, University of Health Sciences, Izmir, Turkey
| | - Fatma Demet Arslan
- Department of Medical Biochemistry, Tepecik Training and Research Hospital, University of Health Sciences, Izmir, Turkey
| | - Banu Isbilen Basok
- Department of Medical Biochemistry, Tepecik Training and Research Hospital, University of Health Sciences, Izmir, Turkey
| | - Ayfer Colak
- Department of Medical Biochemistry, Tepecik Training and Research Hospital, University of Health Sciences, Izmir, Turkey
| | - Can Duman
- Department of Medical Biochemistry, University of Demokrasi, Izmir, Turkey
| |
Collapse
|
16
|
Srivastava A, Palaia T, Hall C, Stevenson M, Lee J, Ragolia L. Lipocalin-type Prostaglandin D2 Synthase appears to function as a Novel Adipokine Preventing Adipose Dysfunction in response to a High Fat Diet. Prostaglandins Other Lipid Mediat 2021; 157:106585. [PMID: 34371198 DOI: 10.1016/j.prostaglandins.2021.106585] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/24/2021] [Accepted: 08/03/2021] [Indexed: 12/29/2022]
Abstract
Adipose dysfunction is the primary defect in obesity that contributes to the development of dyslipidemia, insulin resistance, cardiovascular diseases, type 2 diabetes, non-alcoholic fatty liver disease (NAFLD) and some cancers. Previously, we demonstrated the development of NAFLD in lipocalin-type prostaglandin D2 synthase (L-PGDS) knockout mice regardless of diet. In the present study, we examined the role of L-PGDS in adipose in response to a high fat diet. We observed decreased expression of L-PGDS in adipose tissue and concomitant lower plasma levels in a dietary model of obesity as well as in insulin resistant 3T3-L1 adipocytes. We show reduced adiponectin expression and phosphorylation of AMPK in white adipose tissue of L-PGDS KO mice after 14 weeks on a high fat diet as compared to control C57BL/6 mice. We also observe an increased fat content in L-PGDS KO mice as demonstrated by adipocyte hypertrophy and increased expression of lipogenenic genes. We confirmed our in vivo findings in in vitro 3T3-L1 adipocytes, using an enzymatic inhibitor of L-PGDS (AT56). Rosiglitazone treatment drastically increased L-PGDS expression in insulin resistant 3T3-L1 adipocytes and increased adiponectin expression and AMPK phosphorylation in AT56 treated 3T3-L1 adipocytes. We conclude that the absence of L-PGDS has a deleterious effect on adipose tissue functioning, which further reduces insulin sensitivity in adipose tissue. Consequently, we propose L-PGDS appears to function as a potential member of the adipokine secretome involved in the regulation of the obesity-associated metabolic syndrome.
Collapse
Affiliation(s)
- Ankita Srivastava
- Department of Biomedical research, NYU Langone Hospital, Long Island, United States
| | - Thomas Palaia
- Department of Biomedical research, NYU Langone Hospital, Long Island, United States; Department of Foundations of Medicine, NYU Long Island School of Medicine, 101 Mineola Blvd. Suite 4-003, Mineola, NY, 11501, United States
| | - Christopher Hall
- Department of Biomedical research, NYU Langone Hospital, Long Island, United States
| | - Matthew Stevenson
- Department of Biomedical research, NYU Langone Hospital, Long Island, United States
| | - Jenny Lee
- Department of Biomedical research, NYU Langone Hospital, Long Island, United States
| | - Louis Ragolia
- Department of Biomedical research, NYU Langone Hospital, Long Island, United States; Department of Foundations of Medicine, NYU Long Island School of Medicine, 101 Mineola Blvd. Suite 4-003, Mineola, NY, 11501, United States.
| |
Collapse
|
17
|
Ebert N, Bevc S, Bökenkamp A, Gaillard F, Hornum M, Jager KJ, Mariat C, Eriksen BO, Palsson R, Rule AD, van Londen M, White C, Schaeffner E. Assessment of kidney function: clinical indications for measured GFR. Clin Kidney J 2021; 14:1861-1870. [PMID: 34345408 PMCID: PMC8323140 DOI: 10.1093/ckj/sfab042] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Indexed: 12/18/2022] Open
Abstract
In the vast majority of cases, glomerular filtration rate (GFR) is estimated using serum creatinine, which is highly influenced by age, sex, muscle mass, body composition, severe chronic illness and many other factors. This often leads to misclassification of patients or potentially puts patients at risk for inappropriate clinical decisions. Possible solutions are the use of cystatin C as an alternative endogenous marker or performing direct measurement of GFR using an exogenous marker such as iohexol. The purpose of this review is to highlight clinical scenarios and conditions such as extreme body composition, Black race, disagreement between creatinine- and cystatin C-based estimated GFR (eGFR), drug dosing, liver cirrhosis, advanced chronic kidney disease and the transition to kidney replacement therapy, non-kidney solid organ transplant recipients and living kidney donors where creatinine-based GFR estimation may be invalid. In contrast to the majority of literature on measured GFR (mGFR), this review does not include aspects of mGFR for research or public health settings but aims to reach practicing clinicians and raise their understanding of the substantial limitations of creatinine. While including cystatin C as a renal biomarker in GFR estimating equations has been shown to increase the accuracy of the GFR estimate, there are also limitations to eGFR based on cystatin C alone or the combination of creatinine and cystatin C in the clinical scenarios described above that can be overcome by measuring GFR with an exogenous marker. We acknowledge that mGFR is not readily available in many centres but hope that this review will highlight and promote the expansion of kidney function diagnostics using standardized mGFR procedures as an important milestone towards more accurate and personalized medicine.
Collapse
Affiliation(s)
- Natalie Ebert
- Institute of Public Health, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastjan Bevc
- Department of Nephrology, Faculty of Medicine, Clinic for Internal Medicine, University Medical Center Maribor, University of Maribor, Maribor, Slovenia
| | - Arend Bökenkamp
- Department of Pediatric Nephrology, Amsterdam University Medical Center, Emma Kinderziekenhuis, Amsterdam, The Netherlands
| | - Francois Gaillard
- AP-HP, Hôpital Bichat, Service de Néphrologie, Université de Paris, INSERM U1149, Paris, France
| | - Mads Hornum
- Department of Nephrology, Rigshospitalet and Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kitty J Jager
- Department of Medical Informatics, ERA-EDTA Registry, Amsterdam Public Health Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Bjørn Odvar Eriksen
- Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Runolfur Palsson
- Internal Medicine Services, Division of Nephrology, Landspitali–The National University Hospital of Iceland and Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Andrew D Rule
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Marco van Londen
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, Groningen, The Netherlands
| | - Christine White
- Department of Medicine, Division of Nephrology, Queen’s University, Kingston, Canada
| | - Elke Schaeffner
- Institute of Public Health, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
18
|
Zhao L, Zhang J, Lei S, Ren H, Zou Y, Bai L, Zhang R, Xu H, Li L, Zhao Y, Cooper ME, Tong N, Zhang J, Liu F. Combining glomerular basement membrane and tubular basement membrane assessment improves the prediction of diabetic end-stage renal disease. J Diabetes 2021; 13:572-584. [PMID: 33352010 PMCID: PMC8246816 DOI: 10.1111/1753-0407.13150] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/17/2020] [Accepted: 12/20/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND To address the prognostic value of combining tubular basement membrane (TBM) and glomerular basement membrane (GBM) thickness in diabetic nephropathy (DN). METHODS This retrospective study enrolled 110 patients with type 2 diabetes and biopsy-proven DN from 2011 to 2018. The pathological findings were confirmed according to the Renal Pathology Society classifications. GBM and TBM thicknesses were determined using the Haas' direct measurement/arithmetic mean method and orthogonal intercept method, respectively. Cox proportional hazard models were used to investigate the hazard ratios (HRs) for the influence of combined GBM and TBM thickness for predicting end-stage renal disease (ESRD). RESULTS Patients were assigned to three groups according to the median GBM and TBM thickness: GBMlo TBMlo (GBM < 681 nm and TBM < 1200 nm), GBMhi TBMlo /GBMlo TBMhi (GBM ≥ 681 nm and TBM < 1200 nm, or GBM < 681 nm and TBM ≥ 1200 nm), and GBMhi TBMhi (GBM ≥ 681 nm and TBM ≥ 1200 nm). The GBMhi TBMlo /GBMlo TBMhi and GBMhi TBMhi groups displayed poorer renal function, a more severe glomerular classification and interstitial inflammation, and poorer renal survival rates than the GBMlo TBMlo group The GBMhi TBMlo /GBMlo TBMhi and GBMhi TBMhi groups had adjusted HRs of 1.49 (95% confidence interval [CI], 1.21-9.75) and 3.07 (95% CI, 2.87-12.78), respectively, compared with the GBMlo TBMlo group. CONCLUSIONS TBM thickness enhanced GBM thickness for renal prognosis in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Lijun Zhao
- Division of NephrologyWest China Hospital of Sichuan UniversityChengduChina
- Division of General PracticeWest China Hospital of Sichuan UniversityChengduChina
| | - Junlin Zhang
- Division of NephrologyWest China Hospital of Sichuan UniversityChengduChina
| | - Song Lei
- Division of PathologyWest China Hospital of Sichuan UniversityChengduChina
| | - Honghong Ren
- Division of NephrologyWest China Hospital of Sichuan UniversityChengduChina
| | - Yutong Zou
- Division of NephrologyWest China Hospital of Sichuan UniversityChengduChina
| | - Lin Bai
- Histology and Imaging platform, Core Facility of West China HospitalChengduChina
| | - Rui Zhang
- Division of NephrologyWest China Hospital of Sichuan UniversityChengduChina
| | - Huan Xu
- Division of PathologyWest China Hospital of Sichuan UniversityChengduChina
| | - Lin Li
- Division of PathologyWest China Hospital of Sichuan UniversityChengduChina
| | - Yuancheng Zhao
- Division of NephrologyWest China Hospital of Sichuan UniversityChengduChina
| | - Mark E. Cooper
- Division of DiabetesCentral Clinical School, Monash UniversityMelbourneMelbourneAustralia
| | - Nanwei Tong
- Division of EndocrinologyWest China Hospital of Sichuan UniversityChengduChina
| | - Jie Zhang
- Histology and Imaging platform, Core Facility of West China HospitalChengduChina
| | - Fang Liu
- Division of NephrologyWest China Hospital of Sichuan UniversityChengduChina
| |
Collapse
|
19
|
Inker LA, Couture SJ, Tighiouart H, Abraham AG, Beck GJ, Feldman HI, Greene T, Gudnason V, Karger AB, Eckfeldt JH, Kasiske BL, Mauer M, Navis G, Poggio ED, Rossing P, Shlipak MG, Levey AS. A New Panel-Estimated GFR, Including β 2-Microglobulin and β-Trace Protein and Not Including Race, Developed in a Diverse Population. Am J Kidney Dis 2021; 77:673-683.e1. [PMID: 33301877 PMCID: PMC8102017 DOI: 10.1053/j.ajkd.2020.11.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/10/2020] [Indexed: 12/20/2022]
Abstract
RATIONALE AND OBJECTIVE Glomerular filtration rate (GFR) estimation based on creatinine and cystatin C (eGFRcr-cys) is more accurate than estimated GFR (eGFR) based on creatinine or cystatin C alone (eGFRcr or eGFRcys, respectively), but the inclusion of creatinine in eGFRcr-cys requires specification of a person's race. β2-Microglobulin (B2M) and β-trace protein (BTP) are alternative filtration markers that appear to be less influenced by race than creatinine is. STUDY DESIGN Study of diagnostic test accuracy. SETTING AND PARTICIPANTS Development in a pooled population of 7 studies with 5,017 participants with and without chronic kidney disease. External validation in a pooled population of 7 other studies with 2,245 participants. TESTS COMPARED Panel eGFR using B2M and BTP in addition to cystatin C (3-marker panel) or creatinine and cystatin C (4-marker panel) with and without age and sex or race. OUTCOMES GFR measured as the urinary clearance of iothalamate, plasma clearance of iohexol, or plasma clearance of [51Cr]EDTA. RESULTS Mean measured GFRs were 58.1 and 83.2 mL/min/1.73 m2, and the proportions of Black participants were 38.6% and 24.0%, in the development and validation populations, respectively. In development, addition of age and sex improved the performance of all equations compared with equations without age and sex, but addition of race did not further improve the performance. In validation, the 4-marker panels were more accurate than the 3-marker panels (P < 0.001). The 3-marker panel without race was more accurate than eGFRcys (percentage of estimates greater than 30% different from measured GFR [1 - P30] of 15.6% vs 17.4%; P = 0.01), and the 4-marker panel without race was as accurate as eGFRcr-cys (1 - P30 of 8.6% vs 9.4%; P = 0.2). Results were generally consistent across subgroups. LIMITATIONS No representation of participants with severe comorbid illness and from geographic areas outside of North America and Europe. CONCLUSIONS The 4-marker panel eGFR is as accurate as eGFRcr-cys without requiring specification of race. A more accurate race-free eGFR could be an important advance.
Collapse
Affiliation(s)
- Lesley A Inker
- Division of Nephrology, Tufts Medical Center; Tufts Clinical and Translational Science Institute, Tufts University, Boston, MA.
| | - Sara J Couture
- Division of Nephrology, Tufts Medical Center; Tufts Clinical and Translational Science Institute, Tufts University, Boston, MA
| | - Hocine Tighiouart
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center; Tufts Clinical and Translational Science Institute, Tufts University, Boston, MA; Tufts Medical Center; Tufts Clinical and Translational Science Institute, Tufts University, Boston, MA
| | - Alison G Abraham
- Department of Epidemiology, John Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Gerald J Beck
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH
| | - Harold I Feldman
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA
| | - Tom Greene
- Department of Internal Medicine, University of Utah Health, Salt Lake City, UT
| | - Vilmundur Gudnason
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland; Icelandic Heart Association, Kopavogur, Iceland
| | - Amy B Karger
- Departments of Laboratory Medicine and Pathology, University of Minnesota; Department of Medicine, Hennepin County Medical Center, Minneapolis, MN
| | - John H Eckfeldt
- Departments of Laboratory Medicine and Pathology, University of Minnesota; Department of Medicine, Hennepin County Medical Center, Minneapolis, MN
| | - Bertram L Kasiske
- University of Minnesota; Department of Medicine, Hennepin County Medical Center, Minneapolis, MN
| | - Michael Mauer
- Medicine, University of Minnesota; Department of Medicine, Hennepin County Medical Center, Minneapolis, MN
| | - Gerjan Navis
- Faculty of Medical Sciences, University Medical Center Groningen, Groningen, The Netherlands
| | - Emilio D Poggio
- Department of Nephrology and Hypertension, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH
| | - Peter Rossing
- Steno Diabetes Center Copenhagen and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Michael G Shlipak
- Kidney Health Research Collaborative, San Francisco Veterans Affairs Medical Center, University of California, San Francisco, CA
| | - Andrew S Levey
- Division of Nephrology, Tufts Medical Center; Tufts Clinical and Translational Science Institute, Tufts University, Boston, MA
| |
Collapse
|
20
|
Schwab S, Kleine CE, Bös D, Bohmann S, Strassburg CP, Lutz P, Woitas RP. Beta-trace protein as a potential biomarker of residual renal function in patients undergoing peritoneal dialysis. BMC Nephrol 2021; 22:87. [PMID: 33706697 PMCID: PMC7953776 DOI: 10.1186/s12882-021-02287-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/26/2021] [Indexed: 11/17/2022] Open
Abstract
Background Residual renal function is closely linked to quality of life, morbidity and mortality in dialysis patients. Beta-trace protein (BTP), a low molecular weight protein, has been suggested as marker of residual renal function, in particular in patients on hemodialysis. We hypothesized that BTP also serves as a marker of residual renal function in pertioneal dialysis patients. Methods In this study 34 adult patients on peritoneal dialysis were included. BTP, creatinine, cystatin C and urea concentrations were analyzed simultaneously in serum and dialysate to calculate renal and peritoneal removal of the analytes. Results In peritoneal dialysis patients with residual diuresis, mean serum BTP was 8.16 mg/l (SD ± 4.75 mg/l). BTP correlated inversely with residual diuresis (rs = − 0.58, p < 0.001), residual creatinine clearance (ClCr) (rs = − 0.69, p < 0.001) and total urea clearance (Clurea) (rs = − 0.56, p < 0.001). Mean peritoneal removal of BTP was 3.36 L/week/1.73m2 (SD ± 1.38) and mean renal removal 15.14 L/week/1.73m2 (SD ± 12.65) demonstrating a significant renal contribution to the total removal. Finally, serum BTP inversely correlated with alterations in residual diuresis (r = − 0.41, p = 0.035) and renal creatinine clearance over time (r = − 0.79, p = p < 0.001). Conclusion BTP measurement in the serum may be a simple tool to assess residual renal function in peritoneal dialysis patients.
Collapse
Affiliation(s)
- Sebastian Schwab
- Department of Internal Medicine I, University of Bonn, Bonn, Germany. .,Institute of Experimental Immunology, Rheinische-Friedrichs-Wilhelms University of Bonn, Bonn, Germany.
| | | | | | | | | | - Philipp Lutz
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | | |
Collapse
|
21
|
Obert LA, Elmore SA, Ennulat D, Frazier KS. A Review of Specific Biomarkers of Chronic Renal Injury and Their Potential Application in Nonclinical Safety Assessment Studies. Toxicol Pathol 2021; 49:996-1023. [PMID: 33576319 DOI: 10.1177/0192623320985045] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A host of novel renal biomarkers have been developed over the past few decades which have enhanced monitoring of renal disease and drug-induced kidney injury in both preclinical studies and in humans. Since chronic kidney disease (CKD) and acute kidney injury (AKI) share similar underlying mechanisms and the tubulointerstitial compartment has a functional role in the progression of CKD, urinary biomarkers of AKI may provide predictive information in chronic renal disease. Numerous studies have explored whether the recent AKI biomarkers could improve upon the standard clinical biomarkers, estimated glomerular filtration rate (eGFR), and urinary albumin to creatinine ratio, for predicting outcomes in CKD patients. This review is an introduction to alternative assays that can be utilized in chronic (>3 months duration) nonclinical safety studies to provide information on renal dysfunction and to demonstrate specific situations where these assays could be utilized in nonclinical drug development. Novel biomarkers such as symmetrical dimethyl arginine, dickkopf homolog 3, and cystatin C predict chronic renal injury in animals, act as surrogates for GFR, and may predict changes in GFR in patients over time, ultimately providing a bridge from preclinical to clinical renal monitoring.
Collapse
Affiliation(s)
- Leslie A Obert
- 549350GlaxoSmithKline (GSK), Nonclinical Safety, Collegeville, PA, USA
| | - Susan A Elmore
- Cellular and Molecular Pathology Branch, National Toxicology Program (NTP), 6857National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Daniela Ennulat
- 549350GlaxoSmithKline (GSK), Nonclinical Safety, Collegeville, PA, USA
| | | |
Collapse
|
22
|
Levey AS, Titan SM, Powe NR, Coresh J, Inker LA. Kidney Disease, Race, and GFR Estimation. Clin J Am Soc Nephrol 2020; 15:1203-1212. [PMID: 32393465 PMCID: PMC7409747 DOI: 10.2215/cjn.12791019] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Assessment of GFR is central to clinical practice, research, and public health. Current Kidney Disease Improving Global Outcomes guidelines recommend measurement of serum creatinine to estimate GFR as the initial step in GFR evaluation. Serum creatinine is influenced by creatinine metabolism as well as GFR; hence, all equations to estimate GFR from serum creatinine include surrogates for muscle mass, such as age, sex, race, height, or weight. The guideline-recommended equation in adults (the 2009 Chronic Kidney Disease Epidemiology Collaboration creatinine equation) includes a term for race (specified as black versus nonblack), which improves the accuracy of GFR estimation by accounting for differences in non-GFR determinants of serum creatinine by race in the study populations used to develop the equation. In that study, blacks had a 16% higher average measured GFR compared with nonblacks with the same age, sex, and serum creatinine. The reasons for this difference are only partly understood, and the use of race in GFR estimation has limitations. Some have proposed eliminating the race coefficient, but this would induce a systematic underestimation of measured GFR in blacks, with potential unintended consequences at the individual and population levels. We propose a more cautious approach that maintains and improves accuracy of GFR estimates and avoids disadvantaging any racial group. We suggest full disclosure of use of race in GFR estimation, accommodation of those who decline to identify their race, and shared decision making between health care providers and patients. We also suggest mindful use of cystatin C as a confirmatory test as well as clearance measurements. It would be preferable to avoid specification of race in GFR estimation if there was a superior, evidence-based substitute. The goal of future research should be to develop more accurate methods for GFR estimation that do not require use of race or other demographic characteristics.
Collapse
Affiliation(s)
- Andrew S Levey
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts;
| | - Silvia M Titan
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Neil R Powe
- Department of Medicine, Priscilla Chan and Mark Zuckerberg San Francisco General Hospital and University of California, San Francisco, California; and
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Lesley A Inker
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| |
Collapse
|
23
|
Bargnoux AS, Buthiau D, Morena M, Rodriguez A, Noguera-Gonzalez ME, Gilbert O, Le Quintrec M, Kuster N, Cristol JP. Estimation of residual renal function using beta-trace protein: Impact of dialysis procedures. Artif Organs 2020; 44:647-654. [PMID: 31951029 DOI: 10.1111/aor.13641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/07/2020] [Accepted: 01/07/2020] [Indexed: 12/11/2022]
Abstract
Beta-trace protein (BTP), a low molecular weight protein of 23-29 kDa, has been proposed as a promising biomarker to estimate residual renal function (RRF) in patients on maintenance hemodialysis (HD). Indeed, BTP is cleared by native kidney but not during conventional HD session. By contrast, the removal rate of BTP using convective processes (mainly hemodiafiltration [HDF]) and peritoneal dialysis (PD) has been little or not investigated. Therefore, an aim of this study was to evaluate the impact of dialysis procedures (high-flux HD, on-line post-dilution HDF and PD) on BTP removal in comparison with beta-2 microglobulin (B2M) and cystatin C (CYSC) removals after a single session. In addition, the ability of BTP to predict RRF in PD was assessed. This observational cross-sectional study included a total of 82 stable chronic kidney disease patients, 53 patients were on maintenance dialysis (with n = 26 in HD and n = 27 in HDF) and 29 were on PD. Serum concentrations of BTP, B2M, and CYSC were measured (a) before and after a single dialysis session in HD and HDF anuric patients to calculate reduction percentages, (b) in serum, 24-hour-dialysate and 24-hour-urine in PD patients to compute total, peritoneal, and urinary clearance. RRF was estimated using four equations developed for dialysis patients without urine collection and compared to the mean of the urea and creatinine clearances in PD. The concentrations of the three studied molecules were significantly reduced (P < .001) after dialysis session with significantly higher reduction ratio using HDF compared to HD modality (P < .001): BTP 49.3% vs 17.5%; B2M 82.3% vs 69.7%; CYSC 77.4% vs 66% in HDF and HD, respectively. In non-anuric PD patients, B2M and CYSC were partly removed by peritoneal clearance (72.3% and 57.6% for B2M and CYSC, respectively). By contrast, BTP removal by the peritoneum was negligible and a low bias for the BTP-based equation to estimate RRF (-1.4 mL/min/1.73 m2 ) was calculated. BTP is significantly removed by high-flux HD or HDF, thereby compromising its use to estimate RRF. By contrast, BTP appears as a promising biomarker to estimate RRF in PD patients since it is not affected by peritoneal clearance, unlike B2M and CYSC, and it is well correlated to RRF.
Collapse
Affiliation(s)
- Anne-Sophie Bargnoux
- Département de Biochimie et Hormonologie, PhyMedExp, INSERM, CNRS, Université de Montpellier, CHU de Montpellier, Montpellier Cedex 5, France
| | - Delphine Buthiau
- Département de Biochimie et Hormonologie, Université de Montpellier, CHU de Montpellier, Montpellier Cedex 5, France
| | - Marion Morena
- Département de Biochimie et Hormonologie, PhyMedExp, INSERM, CNRS, Université de Montpellier, CHU de Montpellier, Montpellier Cedex 5, France
| | - Annie Rodriguez
- Département de Biochimie et Hormonologie, Université de Montpellier, CHU de Montpellier, Montpellier Cedex 5, France.,AIDER, Montpellier, France
| | - Maria-Eugenia Noguera-Gonzalez
- Département de Néphrologie, Dialyse et Transplantation, CHU de Montpellier, Université de Montpellier, Montpellier Cedex 5, France
| | | | - Moglie Le Quintrec
- Département de Néphrologie, Dialyse et Transplantation, CHU de Montpellier, Université de Montpellier, Montpellier Cedex 5, France
| | - Nils Kuster
- Département de Biochimie et Hormonologie, PhyMedExp, INSERM, CNRS, Université de Montpellier, CHU de Montpellier, Montpellier Cedex 5, France
| | - Jean-Paul Cristol
- Département de Biochimie et Hormonologie, PhyMedExp, INSERM, CNRS, Université de Montpellier, CHU de Montpellier, Montpellier Cedex 5, France
| |
Collapse
|
24
|
Chen N, Shi H, Zhang L, Zuo L, Xie J, Xie D, Karger AB, Miao S, Ren H, Zhang W, Wang W, Pan Y, Minji W, Sui Z, Okparavero A, Simon A, Chaudhari J, Eckfeldt JH, Inker LA, Levey AS. GFR Estimation Using a Panel of Filtration Markers in Shanghai and Beijing. Kidney Med 2020; 2:172-180. [PMID: 32734236 PMCID: PMC7380432 DOI: 10.1016/j.xkme.2019.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
RATIONALE & OBJECTIVES Estimated glomerular filtration rate (eGFR) using creatinine and cystatin C (eGFRcr-cys) may be less accurate compared to measured GFR (mGFR) in China than in North America, Europe, and Australia due to variation across regions in their non-GFR determinants. The non-GFR determinants of β2-microglobulin (B2M) and β-trace protein (BTP) differ from those of creatinine and cystatin C. Thus, the average eGFR using all 4 markers (eGFRavg) could be more accurate than eGFRcr-cys in China. STUDY DESIGN Diagnostic test study. SETTING & PARTICIPANTS 1,066 participants in Shanghai and Beijing with creatinine and cystatin C and 666 participants with all 4 filtration markers. TESTS COMPARED Index tests were previously developed equations for eGFR using creatinine, cystatin C, B2M, and BTP and combinations. The reference test was mGFR using plasma clearance of iohexol. We compared the performance of eGFRavg to eGFRcr-cys using the proportion of participants with errors in eGFR >30% of mGFR (1 - P30) and root mean square error (RMSE) of the regression of eGFR on mGFR on the logarithmic scale. We also compared classification and reclassification of mGFR categories using eGFRavg compared to eGFRcr-cys. OUTCOMES Accuracy was significantly better for eGFRavg (1 - P30 of 10.4% and RMSE of 0.214) compared to eGFRcr-cys (1 - P30 of 13.8% and RMSE of 0.232; P = 0.004 and P = 0.006, respectively). However, improvements in accuracy did not generally translate into significant improvement in classification or reclassification of mGFR categories. LIMITATIONS Study population may not be generalizable to clinical settings other than large urban medical centers in China. CONCLUSIONS A panel of endogenous filtration markers including B2M and BTP in addition to creatinine and cystatin C may improve GFR estimation in China. Further study is necessary to determine whether GFR estimation using B2M and BTP can be improved and whether these improvements lead to useful clinical applications.
Collapse
Affiliation(s)
- Nan Chen
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Shi
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Luxia Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Peking University, Center for Data Science in Health and Medicine, Beijing, China
| | - Li Zuo
- Department of Nephrology, Peking University People's Hospital, Beijing, China
| | - Jingyuan Xie
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Danshu Xie
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Amy B. Karger
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Shiyuan Miao
- Division of Nephrology, Tufts Medical Center, Boston, MA
| | - Hong Ren
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Zhang
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiming Wang
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yujing Pan
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Wei Minji
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
| | - Zhun Sui
- Department of Nephrology, Peking University People's Hospital, Beijing, China
| | | | - Andrew Simon
- Division of Nephrology, Tufts Medical Center, Boston, MA
| | - Juhi Chaudhari
- Division of Nephrology, Tufts Medical Center, Boston, MA
| | - John H. Eckfeldt
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | | | | |
Collapse
|
25
|
den Bakker E, Gemke RJ, van Wijk JA, Hubeek I, Stoffel-Wagner B, Bökenkamp A. Evidence for shrunken pore syndrome in children. Scandinavian Journal of Clinical and Laboratory Investigation 2019; 80:32-38. [DOI: 10.1080/00365513.2019.1692231] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Emil den Bakker
- Department of Pediatrics, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Reinoud Jbj Gemke
- Department of Pediatrics, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Joanna Ae van Wijk
- Department of Pediatrics, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Isabelle Hubeek
- Department of Clinical Chemistry, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Birgit Stoffel-Wagner
- Department of Clinical Chemistry and Clinical Pharmacology, University Clinics, Bonn, Germany
| | - Arend Bökenkamp
- Department of Pediatrics, Amsterdam University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
26
|
Schwab S, Bös D, Hundt F, Kleine CE, Strassburg CP, Woitas RP. β-Trace protein in hemodialysis - comparison of different therapy modalities and high flux dialyzers. Clin Chem Lab Med 2019; 56:265-267. [PMID: 29715180 DOI: 10.1515/cclm-2018-0178] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 03/28/2018] [Indexed: 11/15/2022]
Affiliation(s)
- Sebastian Schwab
- Department of Internal Medicine I, Medical Clinic I - University of Bonn, Bonn, Germany, Phone: +49 228 287 11038, Fax: +49 228 287 11052.,Institute of Experimental Immunology, Rheinische-Friedrichs-Wilhelms University of Bonn, Bonn, Germany
| | - Dominik Bös
- Department of Nephrology, Medical Clinic I - University of Bonn, Bonn, Germany.,KfH, Renal Center, Bonn, Germany
| | - Felix Hundt
- Department of Nephrology, Medical Clinic I - University of Bonn, Bonn, Germany.,KfH, Renal Center, Bonn, Germany
| | - Carola Ellen Kleine
- KfH, Renal Center, Bonn, Germany.,University of California Irvine, Orange, CA, USA
| | | | - Rainer Peter Woitas
- Department of Nephrology, Medical Clinic I - University of Bonn, Bonn, Germany.,KfH, Renal Center, Bonn, Germany
| |
Collapse
|
27
|
|
28
|
Steubl D, Fan L, Michels WM, Inker LA, Tighiouart H, Dekker FW, Krediet RT, Simon AL, Foster MC, Karger AB, Eckfeldt JH, Li H, Tang J, He Y, Xie M, Xiong F, Li H, Zhang H, Hu J, Liao Y, Ye X, Shafi T, Chen W, Yu X, Levey AS. Development and Validation of Residual Kidney Function Estimating Equations in Dialysis Patients. Kidney Med 2019; 1:104-114. [PMID: 32734191 PMCID: PMC7380427 DOI: 10.1016/j.xkme.2019.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
RATIONALE & OBJECTIVE Measurement of residual kidney function is recommended for the adjustment of the dialysis prescription, but timed urine collections are difficult and prone to errors. Equations to calculate residual kidney function from serum concentrations of endogenous filtration markers and demographic parameters would simplify monitoring of residual kidney function. However, few equations to estimate residual kidney function using serum concentrations of small solutes and low-molecular-weight proteins have been developed and externally validated. STUDY DESIGN Study of diagnostic test accuracy. SETTING & PARTICIPANTS 823 Chinese peritoneal dialysis (PD) patients (development cohort) and 826 PD and hemodialysis patients from the Netherlands NECOSAD study (validation cohort). TESTS COMPARED Equations to estimate residual kidney function (estimated clearance [eCl]) using serum creatinine, urea nitrogen, cystatin C, β2-microglobulin (B2M), β-trace protein (BTP), and combinations, as well as demographic variables (age, sex, height, and weight). Equations were developed using multivariable linear regression analysis in the development cohort and then tested in the validation cohort. Equations were compared with published validated equations. OUTCOMES Residual kidney function measured as urinary clearance (mCl) of urea nitrogen (mClUN) and average of creatinine and urea nitrogen clearance (mClUN-cr). RESULTS In external validation, bias (difference between mCl and eCl) was within ± 1.0 unit for all equations. Accuracy (percent of differences within ± 2.0 units) was significantly better for eClBTP, eClB2M, and eClBTP-B2M than eClUN-cr for both mClUN (78%, 80%, and 81% vs 72%; P < 0.05 for all) and mClUN-cr (72%, 78%, and 79% vs 68%; P < 0.05 for all). The area under the curve for predicting mClUN > 2.0 mL/min was highest for eClB2M (0.853) and eClBTP-B2M (0.848). Results were similar for other validated equations. LIMITATIONS Development cohort only consisted of PD patients, no gold-standard method for residual kidney function measurement. CONCLUSIONS These results confirm the validity and extend the generalizability of residual kidney function estimating equations from serum concentrations of low-molecular-weight proteins without urine collection.
Collapse
Affiliation(s)
- Dominik Steubl
- Division of Nephrology, Tufts Medical Center, Boston, MA
- Abteilung für Nephrologie, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Li Fan
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, NHC Key Laboratory of Nephrology (Sun Yat-sen University), Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Wieneke M. Michels
- Division of Nephrology, Department of Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | | | - Hocine Tighiouart
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center
- Tufts Clinical and Translational Science Institute, Tufts University, Boston, MA
| | - Friedo W. Dekker
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Raymond T. Krediet
- Division of Nephrology, Department of Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | | | | | - Amy B. Karger
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - John H. Eckfeldt
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Hongyan Li
- Department of Nephrology, Huadu District People's Hospital of Guangzhou, Huadu
| | - Jiamin Tang
- Department of Nephrology, Huadu District People's Hospital of Guangzhou, Huadu
| | - Yongcheng He
- Department of Nephrology, Shenzhen Second People's Hospital and the First Affiliated Hospital of Shenzhen University, Shenzhen
| | - Minyan Xie
- Department of Nephrology, Guangzhou Panyu Central Hospital, Panyu
| | - Fei Xiong
- Department of Nephrology, Wuhan No.1 Hospital and Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan
| | - Hongbo Li
- Department of Nephrology, Wuhan No.1 Hospital and Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan
| | - Hao Zhang
- Department of Nephrology, Third Xiangya Hospital of Central South University, Changsha
| | - Jing Hu
- Department of Nephrology, Third Xiangya Hospital of Central South University, Changsha
| | - Yunhua Liao
- Department of Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xudong Ye
- Department of Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Tariq Shafi
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins University
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, NHC Key Laboratory of Nephrology (Sun Yat-sen University), Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Xueqing Yu
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, NHC Key Laboratory of Nephrology (Sun Yat-sen University), Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
- Institute of Nephrology, Guangdong Medical University, Zhanjiang, China
| | | |
Collapse
|
29
|
Abstract
Chronic kidney disease is currently assessed by estimated glomerular filtration rate, a mathematical construct based on creatinine or creatinine and cystatin concentration. Creatinine-based equations have improved with standardization efforts and the Modification of Diet in Renal Disease Study (MDRD) and CKD-Epidemiology Collaboration Study (CKD-EPI). Because the measurement of creatinine is subject to interference from non-GFR determinants, alternative markers have long been sought. These have included cystatin C and low molecular weight proteins like β2-microglobulin and beta trace protein. Tubular disease often occurs before glomerular filtration is impaired and investigators have investigated the excretion of other low molecular weight proteins such as Neutrophil Gelatinase-Associated Lipocalin (NGAL) and Kidney Injury Molecule-1 and N-acetyl-β-d-glucosaminidase. While preliminary, there is some evidence linking these analytes with GFR, disease stage and mortality. Although asymmetrical dimethyl arginine, an inhibitor of nitric oxide, has been shown to be associated with progression of renal disease, symmetric dimethyl arginine may be a better marker. Recent work has also explored the potential of microRNA (miRNA) analysis and metabolomics studies to further elucidate this complex pathophysiologic disease process. Investigators hope to improve our ability to detect CKD by the use of test panels, i.e., various marker combinations thereof. Unfortunately, most of these markers lack standardization unlike traditional measures that rely on creatinine and cystatin C measurement.
Collapse
|
30
|
Song WL, Ricciotti E, Liang X, Grosser T, Grant GR, FitzGerald GA. Lipocalin-Like Prostaglandin D Synthase but Not Hemopoietic Prostaglandin D Synthase Deletion Causes Hypertension and Accelerates Thrombogenesis in Mice. J Pharmacol Exp Ther 2018; 367:425-432. [PMID: 30305427 PMCID: PMC6226547 DOI: 10.1124/jpet.118.250936] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 09/17/2018] [Indexed: 12/13/2022] Open
Abstract
Prostaglandin (PG) D2 is formed by two distinct PGD synthases (PGDS): lipocalin-type PGDS (L-PGDS), which acts as a PGD2-producing enzyme and as extracellular lipophilic transporter, and hematopoietic PGDS (H-PGDS), a σ glutathione-S-transferase. PGD2 plays an important role in the maintenance of vascular function; however, the relative contribution of L-PGDS- and H-PGDS-dependent formation of PGD2 in this setting is unknown. To gain insight into the function played by these distinct PGDS, we assessed systemic blood pressure (BP) and thrombogenesis in L-Pgds and H-Pgds knockout (KO) mice. Deletion of L-Pgds depresses urinary PGD2 metabolite (PGDM) by ∼35%, whereas deletion of H-Pgds does so by ∼90%. Deletion of L-Pgds, but not H-Pgds, elevates BP and accelerates the thrombogenic occlusive response to a photochemical injury to the carotid artery. HQL-79, a H-PGDS inhibitor, further depresses PGDM in L-Pgds KO mice, but has no effect on BP or on the thrombogenic response. Gene expression profiling reveals that pathways relevant to vascular function are dysregulated in the aorta of L-Pgds KOs. These results indicate that the functional impact of L-Pgds deletion on vascular homeostasis may result from an autocrine effect of L-PGDS-dependent PGD2 on the vasculature and/or the L-PGDS function as lipophilic carrier protein.
Collapse
Affiliation(s)
- Wen-Liang Song
- Department of Systems Pharmacology and Translational Therapeutics (W.-L.S., E.R., X.L., T.G., G.A.F.), Institute for Translational Medicine and Therapeutics (W.-L.S., E.R., X.L., T.G., G.R.G., G.A.F.), and Perelman School of Medicine and Department of Genetics (G.R.G.), University of Pennsylvania, Philadelphia, Pennsylvania
| | - Emanuela Ricciotti
- Department of Systems Pharmacology and Translational Therapeutics (W.-L.S., E.R., X.L., T.G., G.A.F.), Institute for Translational Medicine and Therapeutics (W.-L.S., E.R., X.L., T.G., G.R.G., G.A.F.), and Perelman School of Medicine and Department of Genetics (G.R.G.), University of Pennsylvania, Philadelphia, Pennsylvania
| | - Xue Liang
- Department of Systems Pharmacology and Translational Therapeutics (W.-L.S., E.R., X.L., T.G., G.A.F.), Institute for Translational Medicine and Therapeutics (W.-L.S., E.R., X.L., T.G., G.R.G., G.A.F.), and Perelman School of Medicine and Department of Genetics (G.R.G.), University of Pennsylvania, Philadelphia, Pennsylvania
| | - Tilo Grosser
- Department of Systems Pharmacology and Translational Therapeutics (W.-L.S., E.R., X.L., T.G., G.A.F.), Institute for Translational Medicine and Therapeutics (W.-L.S., E.R., X.L., T.G., G.R.G., G.A.F.), and Perelman School of Medicine and Department of Genetics (G.R.G.), University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gregory R Grant
- Department of Systems Pharmacology and Translational Therapeutics (W.-L.S., E.R., X.L., T.G., G.A.F.), Institute for Translational Medicine and Therapeutics (W.-L.S., E.R., X.L., T.G., G.R.G., G.A.F.), and Perelman School of Medicine and Department of Genetics (G.R.G.), University of Pennsylvania, Philadelphia, Pennsylvania
| | - Garret A FitzGerald
- Department of Systems Pharmacology and Translational Therapeutics (W.-L.S., E.R., X.L., T.G., G.A.F.), Institute for Translational Medicine and Therapeutics (W.-L.S., E.R., X.L., T.G., G.R.G., G.A.F.), and Perelman School of Medicine and Department of Genetics (G.R.G.), University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
31
|
Comparison of the new and traditional CKD-EPI GFR estimation equations with urinary inulin clearance: A study of equation performance. Clin Chim Acta 2018; 488:189-195. [PMID: 30445029 DOI: 10.1016/j.cca.2018.11.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/06/2018] [Accepted: 11/12/2018] [Indexed: 01/13/2023]
Abstract
BACKGROUND Diagnosis, prognostication and treatment in chronic kidney disease is often informed by an estimate of the glomerular filtration rate (GFR). Commonly used GFR estimation (eGFR) equations are based on serum creatinine (Cr) concentrations and display suboptimal precision and accuracy. Newer equations incorporating additional endogenous markers such as β-Trace Protein (BTP), β2-Microglobulin (B2M) and cystatin C (cysC) have been developed but require validation. METHODS This prospective cohort study evaluated the performance of 6 eGFR equations developed by the chronic kidney disease - epidemiology collaboration group (CKD-EPI) against urinary inulin clearance GFR in patients recruited from outpatient nephrology clinics. RESULTS Mean biases were negligible and similar between equations. The eGFR-EPI Cr/cysC had the best precision and accuracy of all the equations and the best agreement with inulin mGFR when classifying participants into GFR categories. The BTP and B2M equations displayed the worst precisions and accuracies and showed the least consistent performance across levels of GFR. Thus, the eGFR-EPI Cr/cysC is the least biased, most precise and has the highest accuracy as compared to other eGFR-EPI equations. CONCLUSIONS The BTP and B2M equations are the worst performing of the eGFR-EPI equations, and no benefit is observed with the addition of BTP or B2M to Cr/cysC.
Collapse
|
32
|
Enko D, Meinitzer A, Scharnagl H, Stojakovic T, Kleber ME, Delgado GE, Zelzer S, Drechsler C, Krämer BK, Wanner C, März W, Woitas RP. Prospective cohort studies of beta-trace protein and mortality in haemodialysis patients and patients undergoing coronary angiography. Nephrol Dial Transplant 2018; 33:1984-1991. [PMID: 29474602 DOI: 10.1093/ndt/gfy025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 01/18/2018] [Indexed: 11/13/2022] Open
Abstract
Background Beta-trace protein (BTP) is a low-molecular-weight glycoprotein, which may serve as an endogenous biomarker of kidney function and cardiovascular risk. Methods We examined cardiovascular and all-cause mortality according to BTP concentrations in 2962 individuals referred for coronary angiography from the Ludwigshafen Risk and Cardiovascular Health study and in 907 patients with Type 2 diabetes mellitus undergoing haemodialysis from the German Diabetes and Dialysis (4D) study. Results Haemodialysis patients had considerably higher median (interquartile range) BTP concentrations [6.00 (4.49-7.96) mg/L] and experienced a 4-fold increased mortality rate compared with coronary angiography patients [BTP concentration: 0.55 (0.44-0.67) mg/L]. After adjustment for age, sex, cardiovascular risk factors and creatinine, 4D patients in the highest quartile (>7.96 mg/L) had a 1.6-fold increased rate of all-cause mortality [hazard ratio (HR) 1.62, 95% confidence interval (CI) 1.19-2.20] compared with the lowest quartile (<4.49 mg/L) (P = 0.002) In patients undergoing coronary angiography, the adjusted HRs (95% CI) for all-cause and cardiovascular mortality were 1.23 (1.0-1.51) and 1.27 (0.99-1.63) in the highest (>0.67 mg/L) compared with the lowest (<0.44 mg/L) quartile (P = 0.043 and 0.062). In both cohorts, the BTP/creatinine ratio was a stronger predictor of all-cause and cardiovascular mortality compared with BTP. Conclusion BTP was associated with all-cause mortality independently of renal function in haemodialysis patients. The BTP/creatinine ratio was more predictive for all-cause and cardiovascular mortality in haemodialysis patients and individuals referred for angiography compared with BTP as single marker.
Collapse
Affiliation(s)
- Dietmar Enko
- Institute of Clinical Chemistry and Laboratory Medicine, General Hospital Steyr, Steyr, Austria.,Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Andreas Meinitzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Tatjana Stojakovic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Marcus E Kleber
- Vth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Graciela E Delgado
- Vth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Sieglinde Zelzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Christiane Drechsler
- Division of Nephrology, University of Würzburg, University Hospital, Würzburg, Germany
| | - Bernhard K Krämer
- Vth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Christoph Wanner
- Division of Nephrology, University of Würzburg, University Hospital, Würzburg, Germany
| | - Winfried März
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.,Vth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Synlab Academy, Synlab Holding Germany GmbH, Mannheim and Augsburg, Germany
| | - Rainer P Woitas
- Division of Nephrology, Department of Internal Medicine I, Medical University of Bonn, Bonn.,KfH Renal Center Bonn, Bonn, Germany
| |
Collapse
|
33
|
Corsonello A, Tap L, Roller-Wirnsberger R, Wirnsberger G, Zoccali C, Kostka T, Guligowska A, Mattace-Raso F, Gil P, Fuentes LG, Meltzer I, Yehoshua I, Formiga-Perez F, Moreno-González R, Weingart C, Freiberger E, Ärnlöv J, Carlsson AC, Bustacchini S, Lattanzio F. Design and methodology of the screening for CKD among older patients across Europe (SCOPE) study: a multicenter cohort observational study. BMC Nephrol 2018; 19:260. [PMID: 30309342 PMCID: PMC6180570 DOI: 10.1186/s12882-018-1030-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/31/2018] [Indexed: 12/25/2022] Open
Abstract
Background Decline of renal function is common in older persons and the prevalence of chronic kidney disease (CKD) is rising with ageing. CKD affects different outcomes relevant to older persons, additionally to morbidity and mortality which makes CKD a relevant health burden in this population. Still, accurate laboratory measurement of kidney function is under debate, since current creatinine-based equations have a certain degree of inaccuracy when used in the older population. The aims of the study are as follows: to assess kidney function in a cohort of 75+ older persons using existing methodologies for CKD screening; to investigate existing and innovative biomarkers of CKD in this cohort, and to align laboratory and biomarker results with medical and functional data obtained from this cohort. The study was registered at ClinicalTrials.gov, identifier NCT02691546, February 25th 2016. Methods/design An observational, multinational, multicenter, prospective cohort study in community dwelling persons aged 75 years and over, visiting the outpatient clinics of participating institutions. The study will enroll 2450 participants and is carried out in Austria, Germany, Israel, Italy, the Netherlands, Poland and Spain. Participants will undergo clinical and laboratory evaluations at baseline and after 12 and 24 months- follow-up. Clinical evaluation also includes a comprehensive geriatric assessment (CGA). Local laboratory will be used for ‘basic’ parameters (including serum creatinine and albumin-to-creatinine ratio), whereas biomarker assessment will be conducted centrally. An intermediate telephone follow-up will be carried out at 6 and 18 months. Discussion Combining the use of CGA and the investigation of novel and existing independent biomarkers within the SCOPE study will help to provide evidence in the development of European guidelines and recommendations in the screening and management of CKD in older people. Trial registration This study was registered prospectively on the 25th February 2016 at clinicaltrials.gov (NCT02691546).
Collapse
Affiliation(s)
- Andrea Corsonello
- Italian National Research Center on Aging (INRCA), Ancona, Fermo and Cosenza, Italy
| | - Lisanne Tap
- Section of Geriatric Medicine, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Regina Roller-Wirnsberger
- Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.
| | - Gerhard Wirnsberger
- Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Carmine Zoccali
- CNR-IFC, Clinical Epidemiology and Pathophysiology of Hypertension and Renal Diseases, Ospedali Riuniti, Reggio Calabria, Italy
| | - Tomasz Kostka
- Department of Geriatrics, Healthy Ageing Research Centre, Medical University of Lodz, Lodz, Poland
| | - Agnieszka Guligowska
- Department of Geriatrics, Healthy Ageing Research Centre, Medical University of Lodz, Lodz, Poland
| | - Francesco Mattace-Raso
- Section of Geriatric Medicine, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Pedro Gil
- Department of Geriatric Medicine, Hospital Clinico San Carlos, Madrid, Spain
| | | | - Itshak Meltzer
- The Recanati School for Community Health Professions at the faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Ilan Yehoshua
- Maccabi Healthcare Services Southern Region, Tel Aviv, Israel
| | - Francesc Formiga-Perez
- Geriatric Unit, Internal Medicine Department and Nephrology Department, Bellvitge University Hospital - IDIBELL - L'Hospitalet de Llobregat, Barcelona, Spain
| | - Rafael Moreno-González
- Geriatric Unit, Internal Medicine Department and Nephrology Department, Bellvitge University Hospital - IDIBELL - L'Hospitalet de Llobregat, Barcelona, Spain
| | - Christian Weingart
- Department of General Internal Medicine and Geriatrics, Krankenhaus Barmherzige Brüder Regensburg and Institute for Biomedicine of Aging, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ellen Freiberger
- Department of General Internal Medicine and Geriatrics, Krankenhaus Barmherzige Brüder Regensburg and Institute for Biomedicine of Aging, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Johan Ärnlöv
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.,School of Health and Social Studies, Dalarna University, Falun, Sweden.,Division of Family Medicine, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Axel C Carlsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.,Division of Family Medicine, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Silvia Bustacchini
- Italian National Research Center on Aging (INRCA), Ancona, Fermo and Cosenza, Italy
| | - Fabrizia Lattanzio
- Italian National Research Center on Aging (INRCA), Ancona, Fermo and Cosenza, Italy
| | | |
Collapse
|
34
|
den Bakker E, Gemke R, Pottel H, van Wijk JAE, Hubeek I, Stoffel-Wagner B, Bökenkamp A. Estimation of GFR in children using rescaled beta-trace protein. Clin Chim Acta 2018; 486:259-264. [PMID: 30121167 DOI: 10.1016/j.cca.2018.08.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/13/2018] [Accepted: 08/14/2018] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Beta-trace protein (BTP) is a low molecular weight protein, produced mainly in the cerebrospinal fluid. It has been proposed as a marker for kidney function. Recently, a new method for GFR estimation using mean normal values to rescale GFR marker concentrations has been described for creatinine and cystatin C, two commonly used endogenous markers for kidney function. The aim of this study is to apply this approach to BTP in children. METHOD We retrospectively analyzed serum concentrations of creatinine, cystatin C and BTP measured during inulin clearance tests in children. BTP was measured using a particle-enhanced immunonephelometric assay (Siemens Healthcare). A novel BTP-based eGFR equation was developed using published normal values for children: eGFRBTP[ml/min/1.73m2] = 107.3/BTP/QBTP with QBTP = 0.69. Performance of this equation was compared to the established creatinine-based full age spectrum equation FASage and the cystatin C-based FAScys equations as well as the BTP-based Benlamri equation in terms of bias, % prediction error and P30 and P10 accuracy rates. RESULTS 322 inulin clearance tests were studied. Overall, our novel equation performed comparably to the creatinine-based FASage and the BTP-based Benlamri equations but was less accurate than FAScys (P30: 79.2 vs 86.3%, p = .008). Combining markers significantly enhanced performance compared to the single marker equations, with the exception of FAScys. CONCLUSION Rescaled BTP concentrations are a simple method for estimating GFR in children. However, the additional value of BTP for the estimation of GFR compared to rescaled creatinine and cystatin C still remains to be demonstrated.
Collapse
Affiliation(s)
- Emil den Bakker
- Department of Pediatrics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Reinoud Gemke
- Department of Pediatrics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Hans Pottel
- Department of Public Health and Primary Care, KU Leuven, Kortrijk, Belgium
| | - Joanna A E van Wijk
- Department of Pediatrics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Isabelle Hubeek
- Department of Clinical Chemistry, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Birgit Stoffel-Wagner
- Department of Clinical Chemistry and Clinical Pharmacology, University Clinics, Bonn, Germany
| | - Arend Bökenkamp
- Department of Pediatrics, Amsterdam University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
35
|
Li Y, Xia W, Zhao F, Wen Z, Zhang A, Huang S, Jia Z, Zhang Y. Prostaglandins in the pathogenesis of kidney diseases. Oncotarget 2018; 9:26586-26602. [PMID: 29899878 PMCID: PMC5995175 DOI: 10.18632/oncotarget.25005] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 03/14/2018] [Indexed: 12/11/2022] Open
Abstract
Prostaglandins (PGs) are important lipid mediators produced from arachidonic acid via the sequential catalyzation of cyclooxygenases (COXs) and specific prostaglandin synthases. There are five subtypes of PGs, namely PGE2, PGI2, PGD2, PGF2α, and thromboxane A2 (TXA2). PGs exert distinct roles by combining to a diverse family of membrane-spanning G protein-coupled prostanoid receptors. The distribution of these PGs, their specific synthases and receptors vary a lot in the kidney. This review summarized the recent findings of PGs together with the COXs and their specific synthases and receptors in regulating renal function and highlighted the insights into their roles in the pathogenesis of various kidney diseases.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Weiwei Xia
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Fei Zhao
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Zhaoying Wen
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Aihua Zhang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Songming Huang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Zhanjun Jia
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Yue Zhang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| |
Collapse
|
36
|
Chakraborty D, Akbari A, Knoll GA, Flemming JA, Lowe C, Akbari S, White CA. Serum BTP concentrations are not affected by hepatic dysfunction. BMC Nephrol 2018; 19:87. [PMID: 29653513 PMCID: PMC5899361 DOI: 10.1186/s12882-018-0881-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 03/21/2018] [Indexed: 02/06/2023] Open
Abstract
Background Beta Trace Protein (BTP) is a promising marker of glomerular filtration rate (GFR). Equations to estimate GFR using BTP have been proposed. Very little is known about BTP’s production and metabolism. It has been hypothesized that the liver metabolizes certain BTP isoforms. As such, hepatic dysfunction may influence serum levels independently of GFR. This would impact on the accuracy and precision of GFR estimates using BTP. The purpose of this study was to assess the impact of cirrhosis on serum BTP concentrations. Methods BTP, cystatin C (cysC) and creatinine (Cr) were measured in 99 cirrhotic subjects and in matched controls. BTP/cysC and Cr/cysC ratios were compared between cases and controls. This was repeated after stratification by Child Pugh category. Comparisons of ratios between Child Pugh category A and combined B and C case subjects were also performed. Results There were no differences in BTP/cysC ratios between cases and controls for the entire cohort (0.80 vs 0.79) or for any of the Child Pugh categories (p > 0.10). There were significant differences between cases (1.09) and controls (0.73) for the BTP/Cr ratios (p < 0.001). The BTP/Cr ratio was higher in those with more advanced cirrhosis as compared to those with less severe cirrhosis (1.20 vs 1.03, p < 0.01). There were no differences in BTP/cysC ratios between those with less severe and more advanced cirrhosis (p = 0.25). Conclusions This study suggests that hepatic dysfunction does not influence serum BTP levels and argues against a significant role for the liver in BTP metabolism. Confirmation in a larger group of patients with advanced cirrhosis is required.
Collapse
Affiliation(s)
- Debarati Chakraborty
- Division of Nephrology, Department of Medicine, Queen's University, Etherington Hall, 94 Stuart Street, Kingston, ON, K7L 2N6, Canada
| | - Ayub Akbari
- Division of Nephrology, Department of Medicine, University of Ottawa, Ottawa, ON, Canada.,Kidney Research Centre, Ottawa Health Research Institute, Ottawa, ON, Canada.,Clinical Epidemiology Program, Ottawa Health Research Institute, Ottawa, ON, Canada
| | - Greg A Knoll
- Division of Nephrology, Department of Medicine, University of Ottawa, Ottawa, ON, Canada.,Kidney Research Centre, Ottawa Health Research Institute, Ottawa, ON, Canada.,Clinical Epidemiology Program, Ottawa Health Research Institute, Ottawa, ON, Canada
| | - Jennifer A Flemming
- Division of Gastroenterology, Department of Medicine, Queen's University, Kingston, ON, Canada.,Department of Public Health Sciences, Queen's University, Kingston, ON, Canada
| | - Catherine Lowe
- Division of Gastroenterology, Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Shareef Akbari
- Kidney Research Centre, Ottawa Health Research Institute, Ottawa, ON, Canada
| | - Christine A White
- Division of Nephrology, Department of Medicine, Queen's University, Etherington Hall, 94 Stuart Street, Kingston, ON, K7L 2N6, Canada.
| |
Collapse
|
37
|
Bacci MR, Alves BDCA, Cavallari MR, Azzalis LA, Rozier-Alves RMD, Perez MM, Chehter EZ, Pereira EC, Fonseca FLA. Urinary beta-trace protein gene expression analysis in type 2 diabetes mellitus patients. ACTA ACUST UNITED AC 2018; 15:441-444. [PMID: 29364366 PMCID: PMC5875157 DOI: 10.1590/s1679-45082017ao4012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 08/09/2017] [Indexed: 11/22/2022]
Abstract
Objective To evaluate the gene expression of beta-trace protein in urine of diabetic patients, with no reduction in glomerular filtration rate, which was defined as below 60mL/min/1.73m2. Methods Type 2 diabetes mellitus patients were recruited, and a group of non-diabetic individuals served as control. Beta-trace protein gene expression was analyzed by quantitative PCR. Blood samples were collected to establish glucose levels and baseline kidney function. Accuracy was analyzed using ROC curves. Results Ninety type 2 diabetes mellitus patients and 20 non-diabetic individuals were recruited. The area under the curve was 0.601, sensitivity of 20% and specificity of 89.47%. Among diabetic participants, 18% showed an expression above the cutoff point. Conclusion These results of accuracy of beta-trace protein gene expression in urine of diabetic patients are promising, although they did not achieve a higher area under the curve level.
Collapse
|
38
|
Davenport A. Measuring residual renal function for hemodialysis adequacy: Is there an easier option? Hemodial Int 2018; 21 Suppl 2:S41-S46. [PMID: 29064172 DOI: 10.1111/hdi.12592] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 08/01/2017] [Indexed: 02/04/2023]
Abstract
Most patients starting hemodialysis (HD) have residual renal function. As such, there has been increased interest in starting patients with less frequent and shorter dialysis session times. However, for this incremental approach to be successful, patients require regular monitoring of residual renal function, so that as residual renal function declines, the amount of HD is appropriately increased. Currently most dialysis centers rely on interdialytic urine collections. However, many patients find these inconvenient and there may be marked intrapatient variability due to compliance issues. Thus, alternative markers of residual renal function are required for routine clinical practice. Currently three middle sized molecules; cystatin C, β2 microglobulin, and βtrace protein have been investigated as potential endogenous markers of glomerular filtration. Although none is ideal, combinations of these markers have been proposed to provide a more accurate estimation of glomerular clearance, and in particular cut offs for minimal residual renal function. However, in patients with low levels of residual renal function it remains unclear as to whether the benefits of residual renal function equally apply to glomerular filtration or tubular function.
Collapse
Affiliation(s)
- Andrew Davenport
- UCL Centre for Nephrology, Royal Free Hospital, University College London, Rowland Hill Street, London, NW3 2PF
| |
Collapse
|
39
|
Pottel H, Schaeffner E, Ebert N. Evaluating the diagnostic value of rescaled β-trace protein in combination with serum creatinine and serum cystatin C in older adults. Clin Chim Acta 2018; 480:206-213. [PMID: 29476732 DOI: 10.1016/j.cca.2018.02.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 02/20/2018] [Accepted: 02/20/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND Beta trace protein (BTP) is a novel renal biomarker that has emerged as potential alternative or addition to serum creatinine (Scr) and serum cystatin C (ScysC). We analyzed BTP's diagnostic ability to detect impaired kidney function by rescaling it and we tested whether rescaling BTP allowed us to expand the Full-Age-Spectrum (FAS)-equation to BTP. METHODS 566 participants aged ≥70 years with measured glomerular filtration rate (mGFR), Scr, ScysC and BTP from the population-based Berlin Initiative Study (BIS) were considered. We developed a single and combined FAS-equation using rescaled BTP (BTP/0.60) and calculated its sensitivity (S) and specificity (Sp) to identify kidney disease using a fixed (60 mL/min/1.73 m2) and age-dependent threshold for mGFR. RESULTS Rescaled BTP shared the same reference interval with rescaled Scr and ScysC and showed acceptable diagnostic performance (S = 73.1%, Sp = 86.5%), comparable to Scr (S = 71.0%, Sp = 90.5%) and ScysC (S = 80.7%, Sp = 92.9%). Rescaled BTP can be used in the FAS-equation with comparable performance as Scr and ScysC, but the Scr/ScysC/BTP-combined FAS-eq. (P10 = 57.8%, P30 = 96.6%) did not outperform the Scr/ScysC-combined FAS-eq. (P10 = 57.1%, P30 = 96.3%). CONCLUSIONS Rescaled BTP is a valid alternative to Scr or ScysC to diagnose kidney function. The FAS-concept can be applied to BTP or the combination of BTP, Scr and ScysC.
Collapse
Affiliation(s)
- Hans Pottel
- Department of Public Health and Primary Care, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium.
| | - Elke Schaeffner
- Charité Universitätsmedizin Berlin, Institute of Public Health, Berlin, Germany
| | - Natalie Ebert
- Charité Universitätsmedizin Berlin, Institute of Public Health, Berlin, Germany
| |
Collapse
|
40
|
den Bakker E, Gemke RJBJ, Bökenkamp A. Endogenous markers for kidney function in children: a review. Crit Rev Clin Lab Sci 2018; 55:163-183. [DOI: 10.1080/10408363.2018.1427041] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Emil den Bakker
- Department of Pediatric Nephrology, VU Medical Centre, Amsterdam, The Netherlands
| | | | - Arend Bökenkamp
- Department of Pediatric Nephrology, VU Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
41
|
Motawi TK, Shehata NI, ElNokeety MM, El-Emady YF. Potential serum biomarkers for early detection of diabetic nephropathy. Diabetes Res Clin Pract 2018; 136:150-158. [PMID: 29253627 DOI: 10.1016/j.diabres.2017.12.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 10/28/2017] [Accepted: 12/12/2017] [Indexed: 12/09/2022]
Abstract
AIM Diabetic nephropathy (DN) is considered as one of the diabetic complications affecting up to 40% of patients with type 1 or type 2 diabetes. In clinical practice, the frequently used markers of renal disease and progression are serum creatinine, estimated glomerular filtration rate (eGFR) and albuminuria. The aim of this study is to determine new biomarkers in human serum which are promising for early detection of DN. METHODS This study included 50 patients with type 2 diabetes mellitus (T2DM) and 25 clinically healthy individuals. The patients were divided into two groups; group I included 25 T2DM patients with normoalbuminuria, and group II consisted of 25 T2DM patients with microalbuminuria. In all groups, neutrophil gelatinase-associated lipocalin (NGAL), β-trace protein (βTP) and microRNA- 130b (miR-130b) were estimated. RESULTS The serum levels of NGAL and βTP were significantly elevated in T2DM patients with microalbuminuria (group II) compared with T2DM patients with normoalbuminuria (group I) and control subjects but there was no significant difference between group I and control subjects. Serum miR-130b level was significantly decreased in patients with T2DM (groups I and II) compared with healthy control subjects, with a higher decrease in their levels in group II compared with group I. CONCLUSION Our results suggest that serum NGAL and βTP as tubular and glomerular markers respectively, together with serum miR-130b may be independent and reliable biomarkers for early detection of DN in patients with T2DM.
Collapse
Affiliation(s)
- Tarek Kamal Motawi
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Kasr Al Ainy, St. Cairo 11562, Egypt
| | - Nagwa Ibrahim Shehata
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Kasr Al Ainy, St. Cairo 11562, Egypt
| | - Mahmoud Mohamed ElNokeety
- Department of Internal Medicine, Faculty of Medicine, Cairo University, Kasr Al Ainy, St. Cairo 11562, Egypt
| | - Yasmin Farid El-Emady
- The Holding Company for Biological Products & Vaccines (VACSERA), 51 Wezaret ElZeraa St., Agouza, Giza 12622, Egypt.
| |
Collapse
|
42
|
Shafi T, Levey AS. Measurement and Estimation of Residual Kidney Function in Patients on Dialysis. Adv Chronic Kidney Dis 2018; 25:93-104. [PMID: 29499893 PMCID: PMC5841591 DOI: 10.1053/j.ackd.2017.09.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 09/11/2017] [Indexed: 12/17/2022]
Abstract
Residual kidney function (RKF) in patients on dialysis is strongly associated with survival and better quality of life. Assessment of kidney function underlies the management of patients with chronic kidney disease before dialysis initiation. However, methods to assess RKF after dialysis initiation are just now being refined. In this review, we discuss the definition of RKF and methods for measurement and estimation of RKF, highlighting the unique aspects of dialysis that impact these assessments.
Collapse
Affiliation(s)
- Tariq Shafi
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD; Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, MD; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; and Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, MA.
| | - Andrew S Levey
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD; Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, MD; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; and Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, MA
| |
Collapse
|
43
|
van Craenenbroeck AH, Bragfors-Helin AC, Qureshi AR, Lindholm B, Sjöberg B, Anderstam B, Heimburger O, Stenvinkel P, Bárány P. Plasma Beta-Trace Protein as a Marker of Residual Renal Function: The Effect of Different Hemodialysis Modalities and Intra-Individual Variability over Time. Kidney Blood Press Res 2017; 42:877-885. [PMID: 29161688 DOI: 10.1159/000484537] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 09/21/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Beta-trace protein (BTP) is a low-molecular-weight molecule, which may be used to assess residual renal function (RRF) in dialysis patients. Here we evaluated the influence of hemodialysis (HD) and hemodiafiltration (HDF) on plasma BTP, and analyzed the inter- and intra-individual variability of plasma BTP over time in HD and peritoneal dialysis (PD) patients. METHODS In 12 prevalent HD patients, the effect of a single session of low-flux HD, high-flux HD and HDF on plasma BTP was studied. Blood samples were taken at baseline, after 120 and 240 minutes, and at the start of the next dialysis session. In 13 HD patients and 10 PD patients, inter- and intra-individual variability over three months was studied (monthly and weekly, respectively). Plasma BTP was measured using a nephelometric method. RESULTS No significant decrease in plasma BTP was seen following a session of low-flux HD. Both high-flux HD and HDF resulted in a significant decrease immediately after dialysis (22% and 61% median decrease, respectively). A significant reduction of the molecule persisted only in HDF and a significant decrease (-15%) was still found immediately before the start of the next dialysis session. In both HD and PD patients, the reproducibility over time was excellent with intra-class correlation coefficient of 0.96 (0.93-0.99) and 0.92 (0.86-0.99) respectively. In a small cohort of PD patients, fair agreement existed between mGFR (average of renal urea and creatinine clearance from a 24 hours urine collection) and the BTP-based GFR estimation. CONCLUSION BTP is a stable marker and a promising tool for RRF estimations in PD and HD patients. In patients receiving HDF, plasma levels of BTP should be interpreted with caution.
Collapse
Affiliation(s)
- Amaryllis H van Craenenbroeck
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.,Department of Nephrology, Antwerp University Hospital, Antwerp, Belgium.,University of Antwerp, Antwerp, Belgium
| | - Ann-Christin Bragfors-Helin
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Abdul Rashid Qureshi
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Lindholm
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Bodil Sjöberg
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Björn Anderstam
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Olof Heimburger
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Peter Stenvinkel
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Peter Bárány
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
44
|
Schaeffner E. Determining the Glomerular Filtration Rate—An Overview. J Ren Nutr 2017; 27:375-380. [DOI: 10.1053/j.jrn.2017.07.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 07/19/2017] [Indexed: 11/11/2022] Open
|
45
|
Ebert N, Koep C, Schwarz K, Martus P, Mielke N, Bartel J, Kuhlmann M, Gaedeke J, Toelle M, van der Giet M, Schuchardt M, Schaeffner E. Beta Trace Protein does not outperform Creatinine and Cystatin C in estimating Glomerular Filtration Rate in Older Adults. Sci Rep 2017; 7:12656. [PMID: 28978997 PMCID: PMC5627233 DOI: 10.1038/s41598-017-12645-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 09/13/2017] [Indexed: 11/19/2022] Open
Abstract
Despite intense research the optimal endogenous biomarker for glomerular filtration rate (GFR) estimation has not been identified yet. We analyzed if ß-trace protein (BTP) improved GFR estimation in elderly. 566 participants aged 70+ from the population-based Berlin Initiative Study were included in a cross-sectional validation study. BTP, standardized creatinine and cystatin C were measured in participants with iohexol clearance measurement as gold standard method for measured GFR (mGFR). In a double logarithmic linear model prediction of mGFR by BTP was assessed. Analyses with BTP only and combined with creatinine and cystatin C were performed. Additionally, performance of GFR estimating equations was compared to mGFR. We found that the combination of all three biomarkers showed the best prediction of mGFR (r2 = 0.83), whereat the combination of creatinine and cystatin C provided only minimally diverging results (r2 = 0.82). Single usage of BTP showed worst prediction (r2 = 0.67) within models with only one biomarker. Subgroup analyses (arterial hypertension, diabetes, body mass index ≤23 and >30) demonstrated a slight additional benefit of including BTP into the prediction model for diabetic, hypertensive and lean patients. Among BTP-containing GFR equations the Inker BTP-based equation showed superior performance. Especially the use of cystatin C renders the addition of BTP unnecessary.
Collapse
Affiliation(s)
- Natalie Ebert
- Institute of Public Health, Charité University Medicine, Berlin, Germany.
| | - Camilla Koep
- Institute of Public Health, Charité University Medicine, Berlin, Germany
| | - Kristin Schwarz
- Institute of Public Health, Charité University Medicine, Berlin, Germany
| | - Peter Martus
- Institute of Clinical Epidemiology and Medical Biostatistics, Eberhard Karls University, Tübingen, Germany
| | - Nina Mielke
- Institute of Public Health, Charité University Medicine, Berlin, Germany
| | | | - Martin Kuhlmann
- Department of Nephrology, Vivantes Klinikum im Friedrichshain, Berlin, Germany
| | - Jens Gaedeke
- Division of Nephrology, Charité University Medicine, Campus Mitte, Berlin, Germany
| | - Markus Toelle
- Division of Nephrology, Charité University Medicine Campus Benjamin Franklin, Berlin, Germany
| | - Markus van der Giet
- Division of Nephrology, Charité University Medicine Campus Benjamin Franklin, Berlin, Germany
| | - Mirjam Schuchardt
- Division of Nephrology, Charité University Medicine Campus Benjamin Franklin, Berlin, Germany
| | - Elke Schaeffner
- Institute of Public Health, Charité University Medicine, Berlin, Germany
| |
Collapse
|
46
|
Foster MC, Levey AS, Inker LA, Shafi T, Fan L, Gudnason V, Katz R, Mitchell GF, Okparavero A, Palsson R, Post WS, Shlipak MG. Non-GFR Determinants of Low-Molecular-Weight Serum Protein Filtration Markers in the Elderly: AGES-Kidney and MESA-Kidney. Am J Kidney Dis 2017; 70:406-414. [PMID: 28549536 PMCID: PMC5572311 DOI: 10.1053/j.ajkd.2017.03.021] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/27/2017] [Indexed: 11/11/2022]
Abstract
BACKGROUND Studies in chronic kidney disease populations suggest that the non-glomerular filtration rate (GFR) determinants of serum levels of the low-molecular-weight protein filtration markers cystatin C, β2-microglobulin (B2M), and beta-trace protein (BTP) are less affected by age, sex, and ethnicity than those of creatinine. STUDY DESIGN Cross-sectional study. SETTING & PARTICIPANTS Predominantly elderly participants selected from the Age, Gene/Environment Susceptibility Kidney Study (AGES-Kidney; N=683; mean [SD] age, 79 [4] years; GFR, 62 [17]mL/min/1.73 m2) and from the Multi-Ethnic Study of Atherosclerosis Kidney Study (MESA-Kidney; N=273; mean [SD] age, 70.5 [9] years; GFR, 73 [19]mL/min/1.73 m2). PREDICTORS Demographic and clinical factors hypothesized to be associated with conditions affecting non-GFR determinants of the filtration markers. OUTCOMES Measured GFRs and estimated GFRs (eGFRs) based on creatinine, cystatin C, B2M, and BTP levels (eGFRcr, eGFRcys, eGFRB2M, and eGFRBTP, respectively). Residual associations of factors with eGFR after accounting for measured GFR as the parameter of interest. RESULTS eGFRcys, eGFRB2M, and eGFRBTP had significantly less strong residual associations with age and sex than eGFRcr in both AGES-Kidney and MESA-Kidney and were not associated with ethnicity (black vs white) in MESA-Kidney. After adjusting for age, sex, and ethnicity, residual associations with most clinical factors were smaller than observed with age and sex. eGFRcys and eGFRB2M, but not eGFRBTP, had significant residual associations with C-reactive protein levels in both studies. LIMITATIONS Small sample size may limit power to detect associations. Participants may be healthier than the general population. CONCLUSIONS Similar to previous studies in chronic kidney disease, in community-dwelling elders, cystatin C, B2M, and BTP levels are less affected than creatinine level by age and sex and are not affected by ethnicity. Both cystatin C and B2M levels may be affected by inflammation. These findings are important for the development and use of GFR estimating equations based on low-molecular-weight serum proteins throughout the range in GFRs.
Collapse
Affiliation(s)
- Meredith C Foster
- Division of Nephrology, Tufts Medical Center, Boston, MA; Program in Health Services and Systems Research, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Andrew S Levey
- Division of Nephrology, Tufts Medical Center, Boston, MA.
| | - Lesley A Inker
- Division of Nephrology, Tufts Medical Center, Boston, MA
| | | | - Li Fan
- Division of Nephrology, Tufts Medical Center, Boston, MA; Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University; Key Laboratory of Nephrology, Ministry of Health of China; Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland; Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Ronit Katz
- Kidney Research Institute, Department of Medicine, University of Washington, Seattle, WA
| | | | | | - Runolfur Palsson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland; Division of Nephrology, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland
| | | | | |
Collapse
|
47
|
Brunner HI, Bennett MR, Gulati G, Abulaban K, Klein-Gitelman MS, Ardoin SP, Tucker LB, Rouster-Stevens KA, Witte D, Ying J, Devarajan P. Urine Biomarkers to Predict Response to Lupus Nephritis Therapy in Children and Young Adults. J Rheumatol 2017; 44:1239-1248. [PMID: 28620062 PMCID: PMC6719540 DOI: 10.3899/jrheum.161128] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To delineate urine biomarkers that forecast response to therapy of lupus nephritis (LN). METHODS Starting from the time of kidney biopsy, patients with childhood-onset systemic lupus erythematosus who were diagnosed with LN were studied serially. Levels of 15 biomarkers were measured in random spot urine samples, including adiponectin, α-1-acid glycoprotein (AGP), ceruloplasmin, hemopexin, hepcidin, kidney injury molecule 1, monocyte chemotactic protein-1, lipocalin-like prostaglandin D synthase (LPGDS), transforming growth factor-β (TGF-β), transferrin, and vitamin D binding protein (VDBP). RESULTS Among 87 patients (mean age 15.6 yrs) with LN, there were 37 treatment responders and 50 nonresponders based on the American College of Rheumatology criteria. At the time of kidney biopsy, levels of TGF-β (p < 0.0001) and ceruloplasmin (p = 0.006) were significantly lower among responders than nonresponders; less pronounced differences were present for AGP, hepcidin, LPGDS, transferrin, and VDBP (all p < 0.05). By Month 3, responders experienced marked decreases of adiponectin, AGP, transferrin, and VDBP (all p < 0.01) and mean levels of these biomarkers were all outstanding (area under the receiver-operating characteristic curve ≥ 0.9) for discriminating responders from nonresponders. Patient demographics and extrarenal disease did not influence differences in biomarker levels between response groups. CONCLUSION Low urine levels of TGF-β and ceruloplasmin at baseline and marked reduction of AGP, LPGDS, transferrin, or VDBP and combinations of other select biomarkers by Month 3 are outstanding predictors for achieving remission of LN. If confirmed, these results can be used to help personalize LN therapy.
Collapse
Affiliation(s)
- Hermine I Brunner
- From the Division of Rheumatology, and the Division of Nephrology and Hypertension, and the Division of Pathology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine; Division of Allergy and Rheumatology, Department of Medicine, and the Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA; Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; DeVos Children's Hospital, Grand Rapids, Michigan; Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio; Emory University, Division of Rheumatology, Department of Pediatrics, Atlanta, Georgia, USA.
- H.I. Brunner, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; M.R. Bennett, PhD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; G. Gulati, MD, Division of Allergy and Rheumatology, Department of Medicine, University of Cincinnati; K. Abulaban, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, and DeVos Children's Hospital; M.S. Klein-Gitelman, MD, Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine; S.P. Ardoin, MD, Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center; L.B. Tucker, MBBS, Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital; K.A. Rouster-Stevens, MD, Division of Rheumatology, Emory University, Department of Pediatrics; D. Witte, MD, Division of Pathology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; J. Ying, PhD; Department of Environmental Health, University of Cincinnati; P. Devarajan, MD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine.
| | - Michael R Bennett
- From the Division of Rheumatology, and the Division of Nephrology and Hypertension, and the Division of Pathology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine; Division of Allergy and Rheumatology, Department of Medicine, and the Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA; Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; DeVos Children's Hospital, Grand Rapids, Michigan; Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio; Emory University, Division of Rheumatology, Department of Pediatrics, Atlanta, Georgia, USA
- H.I. Brunner, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; M.R. Bennett, PhD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; G. Gulati, MD, Division of Allergy and Rheumatology, Department of Medicine, University of Cincinnati; K. Abulaban, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, and DeVos Children's Hospital; M.S. Klein-Gitelman, MD, Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine; S.P. Ardoin, MD, Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center; L.B. Tucker, MBBS, Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital; K.A. Rouster-Stevens, MD, Division of Rheumatology, Emory University, Department of Pediatrics; D. Witte, MD, Division of Pathology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; J. Ying, PhD; Department of Environmental Health, University of Cincinnati; P. Devarajan, MD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Gaurav Gulati
- From the Division of Rheumatology, and the Division of Nephrology and Hypertension, and the Division of Pathology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine; Division of Allergy and Rheumatology, Department of Medicine, and the Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA; Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; DeVos Children's Hospital, Grand Rapids, Michigan; Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio; Emory University, Division of Rheumatology, Department of Pediatrics, Atlanta, Georgia, USA
- H.I. Brunner, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; M.R. Bennett, PhD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; G. Gulati, MD, Division of Allergy and Rheumatology, Department of Medicine, University of Cincinnati; K. Abulaban, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, and DeVos Children's Hospital; M.S. Klein-Gitelman, MD, Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine; S.P. Ardoin, MD, Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center; L.B. Tucker, MBBS, Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital; K.A. Rouster-Stevens, MD, Division of Rheumatology, Emory University, Department of Pediatrics; D. Witte, MD, Division of Pathology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; J. Ying, PhD; Department of Environmental Health, University of Cincinnati; P. Devarajan, MD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Khalid Abulaban
- From the Division of Rheumatology, and the Division of Nephrology and Hypertension, and the Division of Pathology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine; Division of Allergy and Rheumatology, Department of Medicine, and the Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA; Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; DeVos Children's Hospital, Grand Rapids, Michigan; Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio; Emory University, Division of Rheumatology, Department of Pediatrics, Atlanta, Georgia, USA
- H.I. Brunner, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; M.R. Bennett, PhD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; G. Gulati, MD, Division of Allergy and Rheumatology, Department of Medicine, University of Cincinnati; K. Abulaban, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, and DeVos Children's Hospital; M.S. Klein-Gitelman, MD, Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine; S.P. Ardoin, MD, Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center; L.B. Tucker, MBBS, Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital; K.A. Rouster-Stevens, MD, Division of Rheumatology, Emory University, Department of Pediatrics; D. Witte, MD, Division of Pathology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; J. Ying, PhD; Department of Environmental Health, University of Cincinnati; P. Devarajan, MD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Marisa S Klein-Gitelman
- From the Division of Rheumatology, and the Division of Nephrology and Hypertension, and the Division of Pathology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine; Division of Allergy and Rheumatology, Department of Medicine, and the Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA; Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; DeVos Children's Hospital, Grand Rapids, Michigan; Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio; Emory University, Division of Rheumatology, Department of Pediatrics, Atlanta, Georgia, USA
- H.I. Brunner, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; M.R. Bennett, PhD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; G. Gulati, MD, Division of Allergy and Rheumatology, Department of Medicine, University of Cincinnati; K. Abulaban, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, and DeVos Children's Hospital; M.S. Klein-Gitelman, MD, Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine; S.P. Ardoin, MD, Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center; L.B. Tucker, MBBS, Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital; K.A. Rouster-Stevens, MD, Division of Rheumatology, Emory University, Department of Pediatrics; D. Witte, MD, Division of Pathology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; J. Ying, PhD; Department of Environmental Health, University of Cincinnati; P. Devarajan, MD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Stacy P Ardoin
- From the Division of Rheumatology, and the Division of Nephrology and Hypertension, and the Division of Pathology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine; Division of Allergy and Rheumatology, Department of Medicine, and the Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA; Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; DeVos Children's Hospital, Grand Rapids, Michigan; Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio; Emory University, Division of Rheumatology, Department of Pediatrics, Atlanta, Georgia, USA
- H.I. Brunner, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; M.R. Bennett, PhD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; G. Gulati, MD, Division of Allergy and Rheumatology, Department of Medicine, University of Cincinnati; K. Abulaban, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, and DeVos Children's Hospital; M.S. Klein-Gitelman, MD, Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine; S.P. Ardoin, MD, Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center; L.B. Tucker, MBBS, Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital; K.A. Rouster-Stevens, MD, Division of Rheumatology, Emory University, Department of Pediatrics; D. Witte, MD, Division of Pathology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; J. Ying, PhD; Department of Environmental Health, University of Cincinnati; P. Devarajan, MD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Lori B Tucker
- From the Division of Rheumatology, and the Division of Nephrology and Hypertension, and the Division of Pathology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine; Division of Allergy and Rheumatology, Department of Medicine, and the Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA; Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; DeVos Children's Hospital, Grand Rapids, Michigan; Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio; Emory University, Division of Rheumatology, Department of Pediatrics, Atlanta, Georgia, USA
- H.I. Brunner, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; M.R. Bennett, PhD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; G. Gulati, MD, Division of Allergy and Rheumatology, Department of Medicine, University of Cincinnati; K. Abulaban, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, and DeVos Children's Hospital; M.S. Klein-Gitelman, MD, Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine; S.P. Ardoin, MD, Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center; L.B. Tucker, MBBS, Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital; K.A. Rouster-Stevens, MD, Division of Rheumatology, Emory University, Department of Pediatrics; D. Witte, MD, Division of Pathology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; J. Ying, PhD; Department of Environmental Health, University of Cincinnati; P. Devarajan, MD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Kelly A Rouster-Stevens
- From the Division of Rheumatology, and the Division of Nephrology and Hypertension, and the Division of Pathology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine; Division of Allergy and Rheumatology, Department of Medicine, and the Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA; Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; DeVos Children's Hospital, Grand Rapids, Michigan; Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio; Emory University, Division of Rheumatology, Department of Pediatrics, Atlanta, Georgia, USA
- H.I. Brunner, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; M.R. Bennett, PhD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; G. Gulati, MD, Division of Allergy and Rheumatology, Department of Medicine, University of Cincinnati; K. Abulaban, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, and DeVos Children's Hospital; M.S. Klein-Gitelman, MD, Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine; S.P. Ardoin, MD, Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center; L.B. Tucker, MBBS, Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital; K.A. Rouster-Stevens, MD, Division of Rheumatology, Emory University, Department of Pediatrics; D. Witte, MD, Division of Pathology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; J. Ying, PhD; Department of Environmental Health, University of Cincinnati; P. Devarajan, MD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine
| | - David Witte
- From the Division of Rheumatology, and the Division of Nephrology and Hypertension, and the Division of Pathology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine; Division of Allergy and Rheumatology, Department of Medicine, and the Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA; Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; DeVos Children's Hospital, Grand Rapids, Michigan; Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio; Emory University, Division of Rheumatology, Department of Pediatrics, Atlanta, Georgia, USA
- H.I. Brunner, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; M.R. Bennett, PhD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; G. Gulati, MD, Division of Allergy and Rheumatology, Department of Medicine, University of Cincinnati; K. Abulaban, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, and DeVos Children's Hospital; M.S. Klein-Gitelman, MD, Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine; S.P. Ardoin, MD, Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center; L.B. Tucker, MBBS, Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital; K.A. Rouster-Stevens, MD, Division of Rheumatology, Emory University, Department of Pediatrics; D. Witte, MD, Division of Pathology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; J. Ying, PhD; Department of Environmental Health, University of Cincinnati; P. Devarajan, MD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Jun Ying
- From the Division of Rheumatology, and the Division of Nephrology and Hypertension, and the Division of Pathology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine; Division of Allergy and Rheumatology, Department of Medicine, and the Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA; Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; DeVos Children's Hospital, Grand Rapids, Michigan; Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio; Emory University, Division of Rheumatology, Department of Pediatrics, Atlanta, Georgia, USA
- H.I. Brunner, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; M.R. Bennett, PhD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; G. Gulati, MD, Division of Allergy and Rheumatology, Department of Medicine, University of Cincinnati; K. Abulaban, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, and DeVos Children's Hospital; M.S. Klein-Gitelman, MD, Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine; S.P. Ardoin, MD, Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center; L.B. Tucker, MBBS, Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital; K.A. Rouster-Stevens, MD, Division of Rheumatology, Emory University, Department of Pediatrics; D. Witte, MD, Division of Pathology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; J. Ying, PhD; Department of Environmental Health, University of Cincinnati; P. Devarajan, MD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Prasad Devarajan
- From the Division of Rheumatology, and the Division of Nephrology and Hypertension, and the Division of Pathology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine; Division of Allergy and Rheumatology, Department of Medicine, and the Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA; Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; DeVos Children's Hospital, Grand Rapids, Michigan; Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio; Emory University, Division of Rheumatology, Department of Pediatrics, Atlanta, Georgia, USA
- H.I. Brunner, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; M.R. Bennett, PhD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; G. Gulati, MD, Division of Allergy and Rheumatology, Department of Medicine, University of Cincinnati; K. Abulaban, MD, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, and DeVos Children's Hospital; M.S. Klein-Gitelman, MD, Department of Pediatrics, Division of Rheumatology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine; S.P. Ardoin, MD, Division of Rheumatology, Department of Internal Medicine, Ohio State University Wexner Medical Center; L.B. Tucker, MBBS, Division of Rheumatology, Department of Pediatrics, British Columbia Children's Hospital; K.A. Rouster-Stevens, MD, Division of Rheumatology, Emory University, Department of Pediatrics; D. Witte, MD, Division of Pathology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine; J. Ying, PhD; Department of Environmental Health, University of Cincinnati; P. Devarajan, MD, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine
| |
Collapse
|
48
|
White CA, Akbari A, Eckfeldt JH, Chakraborty D, McCudden C, Flemming JA, Lowe C, Labrecque P, Parent JL, Fergusson D, Gill JS, Knoll GA. β-Trace Protein Assays: A Comparison Between Nephelometric and ELISA Methodologies. Am J Kidney Dis 2017; 69:866-868. [DOI: 10.1053/j.ajkd.2017.02.371] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 02/17/2017] [Indexed: 11/11/2022]
|
49
|
Baranyi A, Amouzadeh-Ghadikolai O, Lewinski DV, Breitenecker RJ, Stojakovic T, März W, Robier C, Rothenhäusler HB, Mangge H, Meinitzer A. Beta-trace Protein as a new non-invasive immunological Marker for Quinolinic Acid-induced impaired Blood-Brain Barrier Integrity. Sci Rep 2017; 7:43642. [PMID: 28276430 PMCID: PMC5343478 DOI: 10.1038/srep43642] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 01/26/2017] [Indexed: 12/27/2022] Open
Abstract
Quinolinic acid, a macrophage/microglia-derived excitotoxin fulfills a plethora of functions such as neurotoxin, gliotoxin, and proinflammatory mediator, and it alters the integrity and cohesion of the blood-brain barrier in several pathophysiological states. Beta-trace protein (BTP), a monomeric glycoprotein, is known to indicate cerebrospinal fluid leakage. Thus, the prior aim of this study was to investigate whether BTP might non-invasively indicate quinolinic acid-induced impaired blood-brain barrier integrity. The research hypotheses were tested in three subsamples with different states of immune activation (patients with HCV-infection and interferon-α, patients with major depression, and healthy controls). BTP has also been described as a sensitive marker in detecting impaired renal function. Thus, the renal function has been considered. Our study results revealed highest quinolinic acid and highest BTP- levels in the subsample of patients with HCV in comparison with the other subsamples with lower or no immune activation (quinolinic acid: F = 21.027, p < 0.001 [ANOVA]; BTP: F = 6.792, p < 0.01 [ANOVA]). In addition, a two-step hierarchical linear regression model showed that significant predictors of BTP levels are quinolinic acid, glomerular filtration rate and age. The neurotoxin quinolinic acid may impair blood-brain barrier integrity. BTP might be a new non-invasive biomarker to indicate quinolinic acid-induced impaired blood-brain barrier integrity.
Collapse
Affiliation(s)
- Andreas Baranyi
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria.,Institute for International Management Practice, ARU Cambridge, Cambridge, UK
| | | | - Dirk von Lewinski
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Robert J Breitenecker
- Department of Innovation Management and Entrepreneurship, Alpen-Adria-Universität Klagenfurt, Klagenfurt, Austria
| | - Tatjana Stojakovic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Winfried März
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.,Synlab Academy, Synlab Services LLC, Mannheim, Germany.,Medical Clinic V (Nephrology, Hypertensiology, Endocrinology), Medical Faculty Mannheim, Ruperto Carola University Heidelberg, Mannheim, Germany
| | - Christoph Robier
- Hospital of the Brothers of St. John of God, Graz, Austria.,Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Hans-Bernd Rothenhäusler
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Harald Mangge
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Andreas Meinitzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| |
Collapse
|
50
|
Liu X, Foster MC, Tighiouart H, Anderson AH, Beck GJ, Contreras G, Coresh J, Eckfeldt JH, Feldman HI, Greene T, Hamm LL, He J, Horwitz E, Lewis J, Ricardo AC, Shou H, Townsend RR, Weir MR, Inker LA, Levey AS. Non-GFR Determinants of Low-Molecular-Weight Serum Protein Filtration Markers in CKD. Am J Kidney Dis 2016; 68:892-900. [PMID: 27663042 PMCID: PMC5123901 DOI: 10.1053/j.ajkd.2016.07.021] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 07/18/2016] [Indexed: 01/12/2023]
Abstract
BACKGROUND Unlike the case with creatinine, conditions affecting the non-glomerular filtration rate (GFR) determinants of low-molecular-weight serum proteins, β-trace protein (BTP), β2-microglobulin (B2M), and cystatin C, are not well characterized. STUDY DESIGN Pooled cross-sectional analysis of 3 studies. SETTING & PARTICIPANTS 3,156 persons with chronic kidney disease from the MDRD (Modification of Diet in Renal Disease) Study, AASK (African American Study of Kidney Disease and Hypertension), and CRIC (Chronic Renal Insufficiency Cohort) Study. PREDICTORS Demographic and clinical factors hypothesized to be associated with non-GFR determinants of the filtration markers, selected from literature review and physiologic and clinical considerations. OUTCOMES Serum creatinine, BTP, B2M, and cystatin C levels. RESULTS In multivariable-adjusted errors-in-variables regression models that included adjustment for measured GFR (mGFR) and mGFR measurement error, creatinine level had stronger associations with male sex, black race, and higher urine creatinine excretion than the other filtration markers. BTP was associated less strongly with age, similar in direction with sex, and opposite in direction with race than creatinine level. Like cystatin C, B2M level was associated less strongly with age, sex, and race than creatinine level. BTP, B2M, and cystatin C levels were associated more strongly than creatinine level with other factors, including urine protein excretion and weight for BTP, smoking and urine protein excretion for B2M, and smoking for cystatin C. LIMITATIONS Findings may not be generalizable to populations without chronic kidney disease, and residual confounding with GFR due to incomplete adjustment for GFR measurement error. CONCLUSIONS Like creatinine, serum levels of low-molecular-weight proteins are affected by conditions other than GFR. Knowledge of these conditions can aid the interpretation of GFR estimates and risk using these markers and guide the use of these filtration markers in developing GFR estimating equations.
Collapse
Affiliation(s)
- Xun Liu
- Division of Nephrology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guanghzou, China
| | | | | | - Amanda H Anderson
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | | | | - Josef Coresh
- Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Harold I Feldman
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | | - L Lee Hamm
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | | | | | - Ana C Ricardo
- Division of Nephrology, Department of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Haochang Shou
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Raymond R Townsend
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Matthew R Weir
- Division of Nephrology, University of Maryland School of Medicine, Baltimore, MD
| | | | | |
Collapse
|