1
|
Shafiee S, Dastmalchi S, Gharekhani A, Shayanfar A. Analysis of indoxyl sulfate in biological fluids with emphasis on sample preparation techniques: A comprehensive analytical review. Heliyon 2024; 10:e35032. [PMID: 39157307 PMCID: PMC11328088 DOI: 10.1016/j.heliyon.2024.e35032] [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: 09/05/2023] [Revised: 07/16/2024] [Accepted: 07/22/2024] [Indexed: 08/20/2024] Open
Abstract
The uremic toxin indoxyl sulfate (IS) has been related to the development of various medical conditions notably chronic kidney disease (CKD). Hence, quantification of this biomarker in biological fluids may be a diagnostic tool to evaluate renal system functionality. Numerous analytical methods including liquid chromatography, gas chromatography, spectroscopy, and electrochemical techniques have since been used to analyze IS in different biological fluids. The current review highlights the relevant studies that assessed IS with a special focus on sample preparation, which is essential to reduce or eliminate the effect of endogenous components from the matrix in bioanalysis.
Collapse
Affiliation(s)
- Samira Shafiee
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Siavoush Dastmalchi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afshin Gharekhani
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shayanfar
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
2
|
André C, Bodeau S, Kamel S, Bennis Y, Caillard P. The AKI-to-CKD Transition: The Role of Uremic Toxins. Int J Mol Sci 2023; 24:16152. [PMID: 38003343 PMCID: PMC10671582 DOI: 10.3390/ijms242216152] [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: 09/14/2023] [Revised: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
After acute kidney injury (AKI), renal function continues to deteriorate in some patients. In a pro-inflammatory and profibrotic environment, the proximal tubules are subject to maladaptive repair. In the AKI-to-CKD transition, impaired recovery from AKI reduces tubular and glomerular filtration and leads to chronic kidney disease (CKD). Reduced kidney secretion capacity is characterized by the plasma accumulation of biologically active molecules, referred to as uremic toxins (UTs). These toxins have a role in the development of neurological, cardiovascular, bone, and renal complications of CKD. However, UTs might also cause CKD as well as be the consequence. Recent studies have shown that these molecules accumulate early in AKI and contribute to the establishment of this pro-inflammatory and profibrotic environment in the kidney. The objective of the present work was to review the mechanisms of UT toxicity that potentially contribute to the AKI-to-CKD transition in each renal compartment.
Collapse
Affiliation(s)
- Camille André
- Department of Clinical Pharmacology, Amiens Medical Center, 80000 Amiens, France; (S.B.); (Y.B.)
- GRAP Laboratory, INSERM UMR 1247, University of Picardy Jules Verne, 80000 Amiens, France
| | - Sandra Bodeau
- Department of Clinical Pharmacology, Amiens Medical Center, 80000 Amiens, France; (S.B.); (Y.B.)
- MP3CV Laboratory, UR UPJV 7517, University of Picardy Jules Verne, 80000 Amiens, France; (S.K.); (P.C.)
| | - Saïd Kamel
- MP3CV Laboratory, UR UPJV 7517, University of Picardy Jules Verne, 80000 Amiens, France; (S.K.); (P.C.)
- Department of Clinical Biochemistry, Amiens Medical Center, 80000 Amiens, France
| | - Youssef Bennis
- Department of Clinical Pharmacology, Amiens Medical Center, 80000 Amiens, France; (S.B.); (Y.B.)
- MP3CV Laboratory, UR UPJV 7517, University of Picardy Jules Verne, 80000 Amiens, France; (S.K.); (P.C.)
| | - Pauline Caillard
- MP3CV Laboratory, UR UPJV 7517, University of Picardy Jules Verne, 80000 Amiens, France; (S.K.); (P.C.)
- Department of Nephrology, Dialysis and Transplantation, Amiens Medical Center, 80000 Amiens, France
| |
Collapse
|
3
|
Okazaki M, Obi Y, Shafi T, Rhee CM, Kovesdy CP, Kalantar-Zadeh K. Residual Kidney Function and Cause-Specific Mortality Among Incident Hemodialysis Patients. Kidney Int Rep 2023; 8:1989-2000. [PMID: 37849997 PMCID: PMC10577493 DOI: 10.1016/j.ekir.2023.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 07/16/2023] [Accepted: 07/24/2023] [Indexed: 10/19/2023] Open
Abstract
Introduction The survival benefit of residual kidney function (RKF) in patients on hemodialysis is presumably due to enhanced fluid management and solute clearance. However, data are lacking on the association of renal urea clearance (CLurea) with specific causes of death. Methods We conducted a longitudinal cohort study of 39,623 adults initiating thrice-weekly in-center hemodialysis from 2007 to 2011 and had data on renal CLurea and urine volume. Multivariable cause-specific proportional hazards model was used to examine the associations between baseline RKF and cause-specific mortality, including sudden cardiac death (SCD), non-SCD cardiovascular death (CVD), and non-CVD. Restricted cubic splines were fitted for change in RKF over 6 months after initiating hemodialysis. Results Among 39,623 patients with data on baseline renal CLurea and urine volume, there was a significant trend toward a higher mortality risk across lower RKF levels, irrespective of cause of death in a case-mix adjustment model (Ptrend < 0.05). Adjustment for ultrafiltration rate (UFR) slightly attenuated the association between low renal CLurea and high cause-specific mortality, whereas adjustment for highest potassium did not have substantial effect. Among 12,169 patients with data on change in RKF, a 6-month decline in renal CLurea showed graded associations with SCD, non-SCD CVD, and non-CVD risk, whereas the graded associations between faster 6-month decline in urine output and higher death risk were clear only for SCD and non-CVD. Conclusion Lower RKF and loss of RKF were associated with higher cause-specific mortality among patients initiating thrice-weekly in-center hemodialysis.
Collapse
Affiliation(s)
- Masaki Okazaki
- Harold Simmons Center for Kidney Disease Research and Epidemiology, Division of Nephrology, Hypertension and Kidney Transplantation, University of California Irvine School of Medicine, Orange, California, USA
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshitsugu Obi
- Division of Nephrology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Tariq Shafi
- Division of Nephrology, Houston Methodist Hospital, Houston, Texas, USA
| | - Connie M. Rhee
- Harold Simmons Center for Kidney Disease Research and Epidemiology, Division of Nephrology, Hypertension and Kidney Transplantation, University of California Irvine School of Medicine, Orange, California, USA
- Division of Nephrology, Hypertension, and Kidney Transplantation, University of California Irvine Medical Center, Orange, California, USA
| | - Csaba P. Kovesdy
- Division of Nephrology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Nephrology Section, Memphis Veterans Affairs Medical Center, Memphis, Tennessee, USA
| | - Kamyar Kalantar-Zadeh
- Harold Simmons Center for Kidney Disease Research and Epidemiology, Division of Nephrology, Hypertension and Kidney Transplantation, University of California Irvine School of Medicine, Orange, California, USA
- Los Angeles County Harbor-UCLA Medical Center, and The Lundquist Institute, Torrance, California, USA
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles (UCLA), Los Angeles, California, USA
- Nephrology Section, Tibor Rubin Veterans Affairs Medical Center, Long Beach, California, USA
| |
Collapse
|
4
|
Ahmed S, de Vries JC, Lu J, Stuart MHV, Mihăilă SM, Vernooij RWM, Masereeuw R, Gerritsen KGF. Animal Models for Studying Protein-Bound Uremic Toxin Removal-A Systematic Review. Int J Mol Sci 2023; 24:13197. [PMID: 37686004 PMCID: PMC10487432 DOI: 10.3390/ijms241713197] [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: 07/25/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Protein-bound uremic toxins (PBUTs) are associated with the progression of chronic kidney disease (CKD) and its associated morbidity and mortality. The conventional dialysis techniques are unable to efficiently remove PBUTs due to their plasma protein binding. Therefore, novel approaches are being developed, but these require validation in animals before clinical trials can begin. We conducted a systematic review to document PBUT concentrations in various models and species. The search strategy returned 1163 results for which abstracts were screened, resulting in 65 full-text papers for data extraction (rats (n = 41), mice (n = 17), dogs (n = 3), cats (n = 4), goats (n = 1), and pigs (n = 1)). We performed descriptive and comparative analyses on indoxyl sulfate (IS) concentrations in rats and mice. The data on large animals and on other PBUTs were too heterogeneous for pooled analysis. Most rodent studies reported mean uremic concentrations of plasma IS close to or within the range of those during kidney failure in humans, with the highest in tubular injury models in rats. Compared to nephron loss models in rats, a greater rise in plasma IS compared to creatinine was found in tubular injury models, suggesting tubular secretion was more affected than glomerular filtration. In summary, tubular injury rat models may be most relevant for the in vivo validation of novel PBUT-lowering strategies for kidney failure in humans.
Collapse
Affiliation(s)
- Sabbir Ahmed
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (S.A.); (J.L.); (S.M.M.); (R.M.)
| | - Joost C. de Vries
- Department of Nephrology and Hypertension, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (J.C.d.V.); (M.H.V.S.); (R.W.M.V.)
| | - Jingyi Lu
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (S.A.); (J.L.); (S.M.M.); (R.M.)
| | - Milan H. Verrijn Stuart
- Department of Nephrology and Hypertension, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (J.C.d.V.); (M.H.V.S.); (R.W.M.V.)
| | - Silvia M. Mihăilă
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (S.A.); (J.L.); (S.M.M.); (R.M.)
| | - Robin W. M. Vernooij
- Department of Nephrology and Hypertension, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (J.C.d.V.); (M.H.V.S.); (R.W.M.V.)
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (S.A.); (J.L.); (S.M.M.); (R.M.)
| | - Karin G. F. Gerritsen
- Department of Nephrology and Hypertension, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (J.C.d.V.); (M.H.V.S.); (R.W.M.V.)
| |
Collapse
|
5
|
Krieter DH, Rüth M, Lemke HD, Wanner C. Clinical performance comparison of two medium cut-off dialyzers. Ther Apher Dial 2023; 27:284-292. [PMID: 36858049 DOI: 10.1111/1744-9987.13919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Medium-cut-off (MCO) dialyzers may beneficially impact outcomes in patients on hemodialysis. METHODS In a randomized, controlled trial in maintenance hemodialysis patients, the new Nipro ELISIO-17HX MCO dialyzer was compared to the Baxter Theranova 400 filter regarding middle molecule removal. Furthermore, the suitability of two assays for free lambda-light chain (λFLC) detection (Freelite vs. N-Latex) was verified. RESULTS ELISIO-HX achieved slightly lower reduction ratios for β2 -microglobulin (71.8 ± 6.0 vs. 75.3 ± 5.8%; p = 0.001), myoglobin (54.7 ± 8.6 vs. 64.9 ± 8.7%; p < 0.001), and kappa-FLC (62.1 ± 8.8 vs. 56.3 ± 7.7%; p = 0.021). λFLC reduction ratios were more conclusive with the Freelite assay and not different between ELISIO-HX and Theranova (28.4 ± 3.9 vs. 38.7 ± 13.4%; p = 0.069). The albumin loss of Theranova was considerably higher (2.14 ± 0.45 vs. 0.77 ± 0.25 g; p = 0.001) and the Global Removal ScoreLoss alb largely inferior (30.6 ± 7.4 vs. 82.4 ± 29.2%/g; p = 0.006) to ELISIO-HX. CONCLUSIONS The new ELISIO-HX expands the choice of dialyzers for MCO hemodialysis.
Collapse
Affiliation(s)
- Detlef H Krieter
- Division of Nephrology, University Hospital Würzburg, Würzburg, Germany
| | | | | | - Christoph Wanner
- Division of Nephrology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
6
|
Liabeuf S, Drueke T, Massy Z. Rôle des toxines urémiques dans la genèse des complications de la maladie rénale chronique. BULLETIN DE L'ACADÉMIE NATIONALE DE MÉDECINE 2023. [DOI: 10.1016/j.banm.2022.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
7
|
Koniman R, Teo SH, Kaushik M, Nagarajan C, Tan MSY, Tan HK, Ramirez MEG, Lim CC. The use of medium cutoff dialyzers in patients with multiple myeloma and acute kidney injury requiring hemodialysis: A systematic review. Semin Dial 2023; 36:12-17. [PMID: 35840146 DOI: 10.1111/sdi.13115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 06/14/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND Patients with multiple myeloma and high serum levels of circulating free light chains (FLC) have increased risk of acute kidney injury (AKI) secondary to cast nephropathy and is associated with poor survival. Despite removal of FLC by medium cutoff (MCO) dialyzer, the role of MCO hemodialysis (HD) in the treatment of cast nephropathy and its clinical benefits remain unknown. METHODS A systematic review was conducted to establish the effectiveness of MCO dialyzer and clinical outcomes, compared to other forms of dialyzers in the removal of FLC, in myeloma patients with AKI. The primary outcome was effectiveness of MCO-HD in reducing serum FLC. The secondary outcomes were HD independence, estimated glomerular filtrate rate, mortality rates, length of hospitalization, rebound of serum FLC before the next dialysis, removal of other molecules during dialysis, and adverse events. RESULTS We identified three case series, with a total of 17 patients. There were no randomized controlled trials (RCTs) or cohort studies. These case series showed that MCO dialyzer was effective in the removal of FLC and led to a reduction in FLC concentration post-dialysis. The majority of the case series did not have comparator arm and renal and/or other clinical outcomes. CONCLUSION MCO dialyzer appeared to be effective in the removal of FLC based on the existing limited data. However, more data, particularly large-scale RCTs, are needed to assess the use of MCO dialyzer in reducing serum FLC and its effect on clinical outcomes in patients with multiple myeloma and AKI.
Collapse
Affiliation(s)
- Riece Koniman
- Department of Renal Medicine, Singapore General Hospital, Singapore, Singapore
| | - Su Hooi Teo
- Department of Renal Medicine, Singapore General Hospital, Singapore, Singapore
| | - Manish Kaushik
- Department of Renal Medicine, Singapore General Hospital, Singapore, Singapore
| | | | - Melinda Si Yun Tan
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Han Khim Tan
- Department of Renal Medicine, Singapore General Hospital, Singapore, Singapore
| | | | - Cynthia Ciwei Lim
- Department of Renal Medicine, Singapore General Hospital, Singapore, Singapore
| | | |
Collapse
|
8
|
Brookes EM, Power DA. Elevated serum urea-to-creatinine ratio is associated with adverse inpatient clinical outcomes in non-end stage chronic kidney disease. Sci Rep 2022; 12:20827. [PMID: 36460694 PMCID: PMC9718835 DOI: 10.1038/s41598-022-25254-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
To better understand the role of the urea-to-creatinine ratio in chronic kidney disease patients, we assessed the epidemiology of the urea-to-creatinine ratio among hospitalised chronic kidney disease patients, and the association between the urea-to-creatinine ratio and inpatient clinical outcomes. This retrospective cohort study (n = 11,156) included patients with at least two eGFR values < 60 mL/min/1.73m2 measured greater than 90-days apart and admitted to a tertiary hospital between 2014 and 2019. Dialysis and renal transplant patients were excluded. Adjusted odds ratios for factors associated with an elevated urea-to-creatinine ratio were calculated. Multivariate regression was conducted to identify the relationship between elevated UCR and inpatient mortality, intensive care admission, hospital readmission and hospital length-of-stay. Urea-to-creatinine ratio > 100 was present in 27.67% of hospital admissions. Age ≥ 65 years, female gender, gastrointestinal tract bleeding, heart failure, acute kidney injury and lower serum albumin were associated with elevated urea-to-creatinine ratio. Higher urea-to-creatinine ratio level was associated with greater rates of inpatient mortality, hospital readmission within 30-days and longer hospital length-of-stay. Despite this, there was no statistically significant association between higher urea-to-creatinine ratio and intensive care unit admission. Elevated urea-to-creatinine ratio is associated with poor clinical outcomes in chronic kidney disease inpatients. This warrants further investigation to understand the pathophysiological basis for this relationship and to identify effective interventions.
Collapse
Affiliation(s)
- Elizabeth M. Brookes
- grid.1008.90000 0001 2179 088XMelbourne Medical School, The University of Melbourne, Parkville, VIC Australia
| | - David A. Power
- grid.410678.c0000 0000 9374 3516The Department of Nephrology, Austin Health, Heidelberg, VIC Australia ,grid.1008.90000 0001 2179 088XThe Department of Medicine, The University of Melbourne, Victoria, Australia
| |
Collapse
|
9
|
Fluorophenylalkyl-substituted cyanoguanidine derivatives as bacteria-selective MATE transporter inhibitors for the treatment of antibiotic-resistant infections. Bioorg Med Chem 2022; 74:117042. [DOI: 10.1016/j.bmc.2022.117042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/21/2022] [Accepted: 09/28/2022] [Indexed: 11/19/2022]
|
10
|
van Ham WB, Cornelissen CM, van Veen TAB. Uremic toxins in chronic kidney disease highlight a fundamental gap in understanding their detrimental effects on cardiac electrophysiology and arrhythmogenesis. Acta Physiol (Oxf) 2022; 236:e13888. [PMID: 36148604 PMCID: PMC9787632 DOI: 10.1111/apha.13888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/09/2022] [Accepted: 09/18/2022] [Indexed: 01/29/2023]
Abstract
Chronic kidney disease (CKD) and cardiovascular disease (CVD) have an estimated 700-800 and 523 million cases worldwide, respectively, with CVD being the leading cause of death in CKD patients. The pathophysiological interplay between the heart and kidneys is defined as the cardiorenal syndrome (CRS), in which worsening of kidney function is represented by increased plasma concentrations of uremic toxins (UTs), culminating in dialysis patients. As there is a high incidence of CVD in CKD patients, accompanied by arrhythmias and sudden cardiac death, knowledge on electrophysiological remodeling would be instrumental for understanding the CRS. While the interplay between both organs is clearly of importance in CRS, the involvement of UTs in pro-arrhythmic remodeling is only poorly investigated, especially regarding the mechanistic background. Currently, the clinical approach against potential arrhythmic events is mainly restricted to symptom treatment, stressing the need for fundamental research on UT in relation to electrophysiology. This review addresses the existing knowledge of UTs and cardiac electrophysiology, and the experimental research gap between fundamental research and clinical research of the CRS. Clinically, mainly absorbents like ibuprofen and AST-120 are studied, which show limited safe and efficient usability. Experimental research shows disturbances in cardiac electrical activation and conduction after inducing CKD or exposure to UTs, but are scarcely present or focus solely on already well-investigated UTs. Based on UTs data derived from CKD patient cohort studies, a clinically relevant overview of physiological and pathological UTs concentrations is created. Using this, future experimental research is stimulated to involve electrophysiologically translatable animals, such as rabbits, or in vitro engineered heart tissues.
Collapse
Affiliation(s)
- Willem B. van Ham
- Department of Medical Physiology, Division Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Carlijn M. Cornelissen
- Department of Medical Physiology, Division Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Toon A. B. van Veen
- Department of Medical Physiology, Division Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| |
Collapse
|
11
|
Gou X, Ran F, Yang J, Ma Y, Wu X. Construction and Evaluation of a Novel Organic Anion Transporter 1/3 CRISPR/Cas9 Double-Knockout Rat Model. Pharmaceutics 2022; 14:2307. [PMID: 36365126 PMCID: PMC9697873 DOI: 10.3390/pharmaceutics14112307] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/13/2022] [Accepted: 10/25/2022] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND Organic anion transporter 1 (OAT1) and OAT3 have an overlapping spectrum of substrates such that one can exert a compensatory effect when the other is dysfunctional. As a result, the knockout of either OAT1 or OAT3 is not reflected in a change in the excretion of organic anionic substrates. To date, only the mOAT1 and mOAT3 individual knockout mouse models have been available. METHODS In this study, we successfully generated a Slc22a6/Slc22a8 double-knockout (KO) rat model using CRISPR/Cas9 technology and evaluated its biological properties. RESULTS The double-knockout rat model did not expression mRNA for rOAT1 or rOAT3 in the kidneys. Consistently, the renal excretion of p-aminohippuric acid (PAH), the classical substrate of OAT1/OAT3, was substantially decreased in the Slc22a6/Slc22a8 double-knockout rats. The relative mRNA level of Slco4c1 was up-regulated in KO rats. No renal pathological phenotype was evident. The renal elimination of the organic anionic drug furosemide was nearly abolished in the Slc22a6/Slc22a8 knockout rats, but elimination of the organic cationic drug metformin was hardly affected. CONCLUSIONS These results demonstrate that this rat model is a useful tool for investigating the functions of OAT1/OAT3 in metabolic diseases, drug metabolism and pharmacokinetics, and OATs-mediated drug interactions.
Collapse
Affiliation(s)
- Xueyan Gou
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730013, China
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Fenglin Ran
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jinru Yang
- School of First Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Yanrong Ma
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730013, China
| | - Xin’an Wu
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730013, China
| |
Collapse
|
12
|
Tang M, Kalim S. Novel Approaches for the Removal of Uremic Solutes. Clin J Am Soc Nephrol 2022; 17:1113-1115. [PMID: 35835517 PMCID: PMC9435973 DOI: 10.2215/cjn.06860622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Mengyao Tang
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, and Harvard Medical School, Boston, Massachusetts
| | - Sahir Kalim
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
13
|
Bonomini M, Piscitani L, Di Liberato L, Sirolli V. Biocompatibility of Surface-Modified Membranes for Chronic Hemodialysis Therapy. Biomedicines 2022; 10:biomedicines10040844. [PMID: 35453594 PMCID: PMC9025662 DOI: 10.3390/biomedicines10040844] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 02/06/2023] Open
Abstract
Hemodialysis is a life-sustaining therapy for millions of people worldwide. However, despite considerable technical and scientific improvements, results are still not fully satisfactory in terms of morbidity and mortality. The membrane contained in the hemodialyzer is undoubtedly the main determinant of the success and quality of hemodialysis therapy. Membrane properties influence solute removal and the interactions with blood components that define the membrane’s biocompatibility. Bioincompatibility is considered a potential contributor to several uremic complications. Thus, the development of more biocompatible polymers used as hemodialyzer membrane is of utmost importance for improving results and clinical patient outcomes. Many different surface-modified membranes for hemodialysis have been manufactured over recent years by varying approaches in the attempt to minimize blood incompatibility. Their main characteristics and clinical results in hemodialysis patients were reviewed in the present article.
Collapse
Affiliation(s)
- Mario Bonomini
- Nephrology and Dialysis Unit, Department of Medicine, G. d’Annunzio University, Chieti-Pescara, SS. Annunziata Hospital, Via dei Vestini, 66013 Chieti, Italy; (L.D.L.); (V.S.)
- Correspondence:
| | - Luca Piscitani
- Nephrology and Dialysis Unit, Department of Medicine, San Salvatore Hospital, Via Vetoio, 67100 L’Aquila, Italy;
| | - Lorenzo Di Liberato
- Nephrology and Dialysis Unit, Department of Medicine, G. d’Annunzio University, Chieti-Pescara, SS. Annunziata Hospital, Via dei Vestini, 66013 Chieti, Italy; (L.D.L.); (V.S.)
| | - Vittorio Sirolli
- Nephrology and Dialysis Unit, Department of Medicine, G. d’Annunzio University, Chieti-Pescara, SS. Annunziata Hospital, Via dei Vestini, 66013 Chieti, Italy; (L.D.L.); (V.S.)
| |
Collapse
|
14
|
Fructooligosaccharide decreases the production of uremic toxin precursor through modulating gut microbes mediated tyrosine metabolism pathway. FUTURE FOODS 2021. [DOI: 10.1016/j.fufo.2021.100069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
15
|
Rosner MH, Reis T, Husain-Syed F, Vanholder R, Hutchison C, Stenvinkel P, Blankestijn PJ, Cozzolino M, Juillard L, Kashani K, Kaushik M, Kawanishi H, Massy Z, Sirich TL, Zuo L, Ronco C. Classification of Uremic Toxins and Their Role in Kidney Failure. Clin J Am Soc Nephrol 2021; 16:1918-1928. [PMID: 34233920 PMCID: PMC8729494 DOI: 10.2215/cjn.02660221] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Advances in our understanding of uremic retention solutes, and improvements in hemodialysis membranes and other techniques designed to remove uremic retention solutes, offer opportunities to readdress the definition and classification of uremic toxins. A consensus conference was held to develop recommendations for an updated definition and classification scheme on the basis of a holistic approach that incorporates physicochemical characteristics and dialytic removal patterns of uremic retention solutes and their linkage to clinical symptoms and outcomes. The major focus is on the removal of uremic retention solutes by hemodialysis. The identification of representative biomarkers for different classes of uremic retention solutes and their correlation to clinical symptoms and outcomes may facilitate personalized and targeted dialysis prescriptions to improve quality of life, morbidity, and mortality. Recommendations for areas of future research were also formulated, aimed at improving understanding of uremic solutes and improving outcomes in patients with CKD.
Collapse
Affiliation(s)
- Mitchell H. Rosner
- Division of Nephrology, University of Virginia Health System, Charlottesville, Virginia
| | - Thiago Reis
- Department of Nephrology, University of Brazil, Brasília, Brazil
- National Academy of Medicine, Rio de Janeiro, Brazil
| | - Faeq Husain-Syed
- Department of Internal Medicine II, Justus-Liebig-University Giessen, Giessen, Germany
| | - Raymond Vanholder
- Department of Internal Medicine and Pediatrics, University Hospital, Ghent, Belgium
| | - Colin Hutchison
- Faculty of Medicine, University of Queensland, Herston, Australia
- Department of Medicine, Hawke's Bay District Health Board, Hastings, New Zealand
| | - Peter Stenvinkel
- Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Peter J. Blankestijn
- Department of Nephrology and Hypertension, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Mario Cozzolino
- Renal Division, Università degli Studi di Milano, Milan, Italy
| | - Laurent Juillard
- University of Lyon, Villeurbanne, France
- Hôpital E. Herriot, Lyon, France
| | - Kianoush Kashani
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Manish Kaushik
- Department of Renal Medicine, Singapore General Hospital, Singapore, Singapore
| | - Hideki Kawanishi
- Department of Artificial Organs, Tsuchiya General Hospital, Hiroshima, Japan
| | - Ziad Massy
- INSERM U1018, Villejuif, France
- Service de Néphrologie et Dialyse, Hôpital Universitaire Ambroise Paré, Boulogne-Billancourt, France
| | - Tammy Lisa Sirich
- Nephrology Section, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
- Division of Nephrology, Stanford University School of Medicine, Stanford, California
| | - Li Zuo
- Department of Nephrology, Peking University People's Hospital, Beijing, China
| | - Claudio Ronco
- Department of Medicine, University of Padova, Padova, Italy
- Department of Nephrology, Dialysis and Transplantation, International Renal Research Institute of Vicenza, Vicenza, Italy
| |
Collapse
|
16
|
Abstract
Uremic encephalopathy encompasses a wide range of central nervous system abnormalities associated with poor kidney function occurring with either progressive chronic kidney disease or acute kidney injury. The syndrome is likely caused by retention of uremic solutes, alterations in hormonal metabolism, changes in electrolyte and acid-base homeostasis, as well as changes in vascular reactivity, blood-brain barrier transport, and inflammation. There are no defining clinical, laboratory, or imaging findings, and the diagnosis is often made retrospectively when symptoms improve after dialysis or transplantation. The diagnosis is also made difficult because of the many confounding and overlapping conditions seen in patients with chronic kidney disease and acute kidney injury. Thus, institution of kidney replacement therapy should be considered as a trial to improve symptoms in the right clinical context. Neurological symptoms that do not improve after improvement in clearance should prompt a search for other explanations. Further knowledge linking possible uremic retention solutes with neurological symptoms is needed to better understand this syndrome as well as to develop more tailored treatments that aim to improve cognitive function.
Collapse
|
17
|
Passage Number-Induced Replicative Senescence Modulates the Endothelial Cell Response to Protein-Bound Uremic Toxins. Toxins (Basel) 2021; 13:toxins13100738. [PMID: 34679030 PMCID: PMC8538293 DOI: 10.3390/toxins13100738] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 12/17/2022] Open
Abstract
Endothelial aging may be induced early in pathological situations. The uremic toxins indoxyl sulfate (IS) and p-cresol (PC) accumulate in the plasma of chronic kidney disease (CKD) patients, causing accelerated endothelial aging, increased cardiovascular events and mortality. However, the mechanisms by which uremic toxins exert their deleterious effects on endothelial aging are not yet fully known. Thus, the aim of the present study is to determine the effects of IS and PC on endothelial damage and early senescence in cultured human umbilical vein endothelial cells (HUVECs). Hence, we establish an in vitro model of endothelial damage mediated by different passages of HUVECs and stimulated with different concentrations of IS and PC to evaluate functional effects on the vascular endothelium. We observe that cell passage-induced senescence is associated with apoptosis, ROS production and decreased endothelial proliferative capacity. Similarly, we observe that IS and PC cause premature aging in a dose-dependent manner, altering HUVECs' regenerative capacity, and decreasing their cell migration and potential to form vascular structures in vitro. In conclusion, IS and PC cause accelerated aging in HUVECs, thus contributing to endothelial dysfunction associated with CKD progression.
Collapse
|
18
|
Comparative analysis of therapeutic effects between medium cut-off and high flux dialyzers using metabolomics and proteomics: exploratory, prospective study in hemodialysis. Sci Rep 2021; 11:17335. [PMID: 34462546 PMCID: PMC8405670 DOI: 10.1038/s41598-021-96974-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 08/10/2021] [Indexed: 12/03/2022] Open
Abstract
In this single-center prospective study of 20 patients receiving maintenance hemodialysis (HD), we compared the therapeutic effects of medium cut-off (MCO) and high flux (HF) dialyzers using metabolomics and proteomics. A consecutive dialyzer membrane was used for 15-week study periods: 1st HF dialyzer, MCO dialyzer, 2nd HF dialyzer, for 5 weeks respectively. 1H-nuclear magnetic resonance was used to identify the metabolites and liquid chromatography-tandem mass spectrometry (LC–MS/MS) analysis was used to identify proteins. To compare the effects of the HF and MCO dialyzers, orthogonal projection to latent structure discriminant analysis (OPLS-DA) was performed. OPLS-DA showed that metabolite characteristics could be significantly classified by 1st HF and MCO dialyzers. The Pre-HD metabolites with variable importance in projection scores ≥ 1.0 in both 1st HF versus MCO and MCO versus 2nd HF were succinate, glutamate, and histidine. The pre-HD levels of succinate and histidine were significantly lower, while those of glutamate were significantly higher in MCO period than in the HF period. OPLS-DA of the proteome also substantially separated 1st HF and MCO periods. Plasma pre-HD levels of fibronectin 1 were significantly higher, and those of complement component 4B and retinol-binding protein 4 were significantly lower in MCO than in the 1st HF period. Interestingly, as per Ingenuity Pathway Analysis, an increase in epithelial cell proliferation and a decrease in endothelial cell apoptosis occurred during the MCO period. Overall, our results suggest that the use of MCO dialyzers results in characteristic metabolomics and proteomics profiles during HD compared with HF dialyzers, which might be related to oxidative stress, insulin resistance, complement-coagulation axis, inflammation, and nutrition.
Collapse
|
19
|
Lu PH, Yu MC, Wei MJ, Kuo KL. The Therapeutic Strategies for Uremic Toxins Control in Chronic Kidney Disease. Toxins (Basel) 2021; 13:573. [PMID: 34437444 PMCID: PMC8402511 DOI: 10.3390/toxins13080573] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/04/2021] [Accepted: 08/16/2021] [Indexed: 12/27/2022] Open
Abstract
Uremic toxins (UTs) are mainly produced by protein metabolized by the intestinal microbiota and converted in the liver or by mitochondria or other enzymes. The accumulation of UTs can damage the intestinal barrier integrity and cause vascular damage and progressive kidney damage. Together, these factors lead to metabolic imbalances, which in turn increase oxidative stress and inflammation and then produce uremia that affects many organs and causes diseases including renal fibrosis, vascular disease, and renal osteodystrophy. This article is based on the theory of the intestinal-renal axis, from bench to bedside, and it discusses nonextracorporeal therapies for UTs, which are classified into three categories: medication, diet and supplement therapy, and complementary and alternative medicine (CAM) and other therapies. The effects of medications such as AST-120 and meclofenamate are described. Diet and supplement therapies include plant-based diet, very low-protein diet, probiotics, prebiotics, synbiotics, and nutraceuticals. The research status of Chinese herbal medicine is discussed for CAM and other therapies. This review can provide some treatment recommendations for the reduction of UTs in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Ping-Hsun Lu
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 23142, Taiwan; (P.-H.L.); (M.-C.Y.); (M.-J.W.)
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 97048, Taiwan
| | - Min-Chien Yu
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 23142, Taiwan; (P.-H.L.); (M.-C.Y.); (M.-J.W.)
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 97048, Taiwan
| | - Meng-Jiun Wei
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 23142, Taiwan; (P.-H.L.); (M.-C.Y.); (M.-J.W.)
| | - Ko-Lin Kuo
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 23142, Taiwan
- School of Medicine, Buddhist Tzu Chi University, Hualien 97048, Taiwan
| |
Collapse
|
20
|
Abe M, Masakane I, Wada A, Nakai S, Kanda E, Nitta K, Nakamoto H. High-performance dialyzers and mortality in maintenance hemodialysis patients. Sci Rep 2021; 11:12272. [PMID: 34112908 PMCID: PMC8192518 DOI: 10.1038/s41598-021-91751-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/31/2021] [Indexed: 12/13/2022] Open
Abstract
Few data are available regarding the association of dialyzer type with prognosis. In Japan, dialyzers are classified as types I, II, III, IV, and V based on β2-microglobulin clearance rates of < 10, < 30, < 50, < 70, and ≥ 70 mL/min, respectively. We investigated the relationship of the 5 dialyzer types with 1-year mortality. This nationwide cohort study used data collected at the end of 2008 and 2009 by the Japanese Society for Dialysis Therapy Renal Data Registry. We enrolled 203,008 patients on maintenance hemodialysis who underwent hemodialysis for at least 1 year and were managed with any of the 5 dialyzer types. To evaluate the association of dialyzer type with 1-year all-cause mortality, Cox proportional hazards models and propensity score-matched analyses were performed. After adjustment of the data with clinicodemographic factors, the type I, II, and III groups showed significantly higher hazard ratios (HRs) than the type IV dialyzers (reference). After adjustment for Kt/V and β2-microglobulin levels, the HRs were significantly higher in the type I and II groups. After further adjustment for nutrition- and inflammation-related factors, the HRs were not significantly different between the type IV and type I and II groups. However, type V dialyzers consistently showed a significantly lower HR. With propensity score matching, the HR for the type V dialyzer group was significantly lower than that for the type IV dialyzer group. Additional long-term trials are required to determine whether type V dialyzers, which are high-performance dialyzers, can improve prognosis.
Collapse
Affiliation(s)
- Masanori Abe
- The Committee of Renal Data Registry, The Japanese Society for Dialysis Therapy, Tokyo, Japan. .,Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan.
| | - Ikuto Masakane
- The Committee of Renal Data Registry, The Japanese Society for Dialysis Therapy, Tokyo, Japan.,Yabuki Hospital, Yamagata, Japan
| | - Atsushi Wada
- The Committee of Renal Data Registry, The Japanese Society for Dialysis Therapy, Tokyo, Japan.,Department of Nephrology, Kitasaito Hospital, Asahikawa, Japan
| | - Shigeru Nakai
- The Committee of Renal Data Registry, The Japanese Society for Dialysis Therapy, Tokyo, Japan.,Department of Clinical Engineering, Fujita Health University, Aichi, Japan
| | - Eiichiro Kanda
- The Committee of Renal Data Registry, The Japanese Society for Dialysis Therapy, Tokyo, Japan.,Medical Science, Kawasaki Medical School, Okayama, Japan
| | - Kosaku Nitta
- The Committee of Renal Data Registry, The Japanese Society for Dialysis Therapy, Tokyo, Japan.,Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - Hidetomo Nakamoto
- The Committee of Renal Data Registry, The Japanese Society for Dialysis Therapy, Tokyo, Japan.,Department of General Internal Medicine, Saitama Medical University, Saitama, Japan
| |
Collapse
|
21
|
Glorieux G, Vanholder R, Van Biesen W, Pletinck A, Schepers E, Neirynck N, Speeckaert M, De Bacquer D, Verbeke F. Free p-cresyl sulfate shows the highest association with cardiovascular outcome in chronic kidney disease. Nephrol Dial Transplant 2021; 36:998-1005. [DOI: 10.1093/ndt/gfab004] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Indexed: 12/13/2022] Open
Abstract
Abstract
Background
Several protein-bound uraemic toxins (PBUTs) have been associated with cardiovascular (CV) and all-cause mortality in chronic kidney disease (CKD) but the degree to which this is the case per individual PBUT and the pathophysiological mechanism have only partially been unraveled.
Methods
We compared the prognostic value of both total and free concentrations of five PBUTs [p-cresyl sulfate (pCS), p-cresyl glucuronide, indoxyl sulfate, indole acetic acid and hippuric acid] in a cohort of 523 patients with non-dialysis CKD Stages G1–G5. Patients were followed prospectively for the occurrence of a fatal or non-fatal CV event as the primary endpoint and a number of other major complications as secondary endpoints. In addition, association with and the prognostic value of nine markers of endothelial activation/damage was compared.
Results
After a median follow-up of 5.5 years, 149 patients developed the primary endpoint. In multivariate Cox regression models adjusted for age, sex, systolic blood pressure, diabetes mellitus and estimated glomerular filtration rate, and corrected for multiple testing, only free pCS was associated with the primary endpoint {hazard ratio [HR]1.39 [95% confidence interval (CI) 1.14–1.71]; P = 0.0014}. Free pCS also correlated with a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (r = −0.114, P < 0.05), angiopoietin-2 (ANGPT2) (r = 0.194, P < 0.001), matrix metallopeptidase 7 (MMP-7; (r = 0.238, P < 0.001) and syndecan 1 (r = 0.235, P < 0.001). Of these markers of endothelial activation/damage, ANGPT2 [HR 1.46 (95% CI 1.25–1.70); P < 0.0001] and MMP-7 [HR 1.31 (95% CI 1.08–1.59); P = 0.0056] were also predictive of the primary outcome.
Conclusions
Among PBUTs, free pCS shows the highest association with CV outcome in non-dialysed patients with CKD. Two markers of endothelial activation/damage that were significantly correlated with free pCS, ANGPT2 and MMP-7 were also associated with CV outcome. The hypothesis that free pCS exerts its CV toxic effects by an adverse effect on endothelial function deserves further exploration.
Collapse
Affiliation(s)
- Griet Glorieux
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Raymond Vanholder
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Wim Van Biesen
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Anneleen Pletinck
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Eva Schepers
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Nathalie Neirynck
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Marijn Speeckaert
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Dirk De Bacquer
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| | - Francis Verbeke
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
22
|
Lu YC, Wu CC, Tsai IT, Hung WC, Lee TL, Hsuan CF, Yu TH, Wei CT, Chung FM, Lee YJ, Wang CP. Associations among total p-cresylsulfate, indoxyl sulfate and hippuric acid levels with hemodialysis quality indicators in maintenance hemodialysis patients. Clin Chim Acta 2021; 516:83-91. [PMID: 33508250 DOI: 10.1016/j.cca.2021.01.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND Total p-cresylsulfate (PCS), indoxyl sulfate (IS) and hippuric acid (HA) are harmful uremic toxins known to be elevated in patients with uremia. Serum total PCS, IS and HA levels have been associated with coronary atherosclerosis, left ventricular hypertrophy, metabolic acidosis, neurological symptoms, and accelerated renal damage associated with chronic kidney disease; however, no study has examined the effect of total PCS, IS and HA on hemodialysis (HD) quality indicators. The aim of this study was to examine associations among total PCS, IS and HA with HD quality indicators in patients undergoing HD treatment. METHODS This study included 264 consecutive patients at a single HD center who assessed using previously demonstrated HD quality indicators including anemia, bone-mineral metabolism, dialysis dose, cardiovascular risk, and middle molecule removal area. Serum HA was measured using a capillary electrophoresis method. Serum total PCS and IS concentrations were measured using an Ultra Performance LC System. RESULTS Multiple regression analysis showed that sex, potassium, systolic blood pressure (SBP), average BP, β2-microglobulin, and creatinine were independently positively associated with IS level, and that age, total cholesterol, and estimated glomerular filtration rate (eGFR) was independently negatively associated with IS level. In addition, β2-microglobulin was independently positively associated with total PCS. Moreover, potassium, diastolic blood pressure, average BP, β2-microglobulin, dialysis vintage, and albumin were independently positively associated with HA level, and age, transferrin saturation, fasting glucose, and eGFR were independently negatively associated with HA level. When the patients were stratified by age and sex, serum IS and HA levels were still independently associated with some hemodialysis quality indicators. In addition, canonical correlation analysis also confirmed the relationship between uremic toxins (IS and HA) and HD quality indicators (potassium, β2-microglobulin, average BP, creatinine, and eGFR). CONCLUSION This study demonstrated that uremic toxins (IS and HA) and HD quality indicators (potassium, β2-microglobulin, average BP, creatinine, and eGFR) constructs were correlated with each other, and that there were sex and age differences in these associations among maintenance HD patients.
Collapse
Affiliation(s)
- Yung-Chuan Lu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, E-Da Hospital, Kaohsiung 82445, Taiwan; School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Cheng-Ching Wu
- Division of Cardiology, Department of Internal Medicine, E-Da Hospital, Kaohsiung 82445, Taiwan; The School of Chinese Medicine for Post Baccalaureate, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan; Division of Cardiology, Department of Internal Medicine, E-Da Cancer Hospital, Kaohsiung 82445, Taiwan
| | - I-Ting Tsai
- Department of Emergency, E-Da Hospital, Kaohsiung 82445, Taiwan; School of Medicine, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Wei-Chin Hung
- Division of Cardiology, Department of Internal Medicine, E-Da Hospital, Kaohsiung 82445, Taiwan; School of Medicine, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Thung-Lip Lee
- Division of Cardiology, Department of Internal Medicine, E-Da Hospital, Kaohsiung 82445, Taiwan; School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Chin-Feng Hsuan
- Division of Cardiology, Department of Internal Medicine, E-Da Hospital, Kaohsiung 82445, Taiwan; School of Medicine, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Teng-Hung Yu
- Division of Cardiology, Department of Internal Medicine, E-Da Hospital, Kaohsiung 82445, Taiwan; The School of Chinese Medicine for Post Baccalaureate, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Ching-Ting Wei
- Division of General Surgery, Department of Surgery, E-Da Hospital, Kaohsiung 82445, Taiwan; School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan; Department of Biomedical Engineering, I-Shou University, Kaohsiung 82445, Taiwan; Department of Electrical Engineering, I-Shou University, Kaohsiung 82445, Taiwan
| | - Fu-Mei Chung
- Division of Cardiology, Department of Internal Medicine, E-Da Hospital, Kaohsiung 82445, Taiwan
| | | | - Chao-Ping Wang
- Division of Cardiology, Department of Internal Medicine, E-Da Hospital, Kaohsiung 82445, Taiwan; School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan.
| |
Collapse
|
23
|
Vanholder R, Argilés A, Jankowski J. A history of uraemic toxicity and of the European Uraemic Toxin Work Group (EUTox). Clin Kidney J 2021; 14:1514-1523. [PMID: 34413975 PMCID: PMC8371716 DOI: 10.1093/ckj/sfab011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Indexed: 12/13/2022] Open
Abstract
The uraemic syndrome is a complex clinical picture developing in the advanced stages of chronic kidney disease, resulting in a myriad of complications and a high early mortality. This picture is to a significant extent defined by retention of metabolites and peptides that with a preserved kidney function are excreted or degraded by the kidneys. In as far as those solutes have a negative biological/biochemical impact, they are called uraemic toxins. Here, we describe the historical evolution of the scientific knowledge about uraemic toxins and the role played in this process by the European Uraemic Toxin Work Group (EUTox) during the last two decades. The earliest knowledge about a uraemic toxin goes back to the early 17th century when the existence of what would later be named as urea was recognized. It took about two further centuries to better define the role of urea and its link to kidney failure, and one more century to identify the relevance of post-translational modifications caused by urea such as carbamoylation. The knowledge progressively extended, especially from 1980 on, by the identification of more and more toxins and their adverse biological/biochemical impact. Progress of knowledge was paralleled and impacted by evolution of dialysis strategies. The last two decades, when insights grew exponentially, coincide with the foundation and activity of EUTox. In the final section, we summarize the role and accomplishments of EUTox and the part it is likely to play in future action, which should be organized around focus points like biomarker and potential target identification, intestinal generation, toxicity mechanisms and their correction, kidney and extracorporeal removal, patient-oriented outcomes and toxin characteristics in acute kidney injury and transplantation.
Collapse
Affiliation(s)
- Raymond Vanholder
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Angel Argilés
- RD-Néphrologie, Montpellier, France.,Néphrologie Dialyse St Guilhem, Sète, France
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research, University Hospital, RWTH Aachen, Aachen, Germany.,School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | | |
Collapse
|
24
|
Ma YR, Xin MY, Li K, Wang H, Rao Z, Liu TX, Wu XA. An LC-MS/MS analytical method for the determination of uremic toxins in patients with end-stage renal disease. J Pharm Biomed Anal 2020; 191:113551. [PMID: 32889350 DOI: 10.1016/j.jpba.2020.113551] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/09/2020] [Accepted: 08/10/2020] [Indexed: 12/18/2022]
Abstract
End-stage renal disease (ESRD) is the last stage of chronic kidney disease, characterized by the progressive accumulation of uremic toxins (UTs). Hemodialysis is the standard approach to remove UTs from the body. Creatinine and urea levels are important indices of hemodialysis effectiveness, but the utility of those markers to estimate the removal of UTs, especially protein-binding UTs is limited. We developed an LC-MS/MS method for the quantification of UTs and to provide markers for evaluating hemodialysis effectiveness. These substances were extracted from serum samples after acetonitrile precipitation of protein and then separated on a HILIC column. The flow rate was 0.6 mL/min with a run time of 8.0 min for the negative ion mode and positive ion mode each. In this study 26 UTs were determined in normal subjects and in patients with ESRD before and after hemodialysis; serum levels were significantly higher in patients with ESRD than in subjects with normal renal function. A significant decrease in a variety of serum UTs were observed in patients after dialysis treatment, but no change in the levels of orotic acid, CMPF, kynurenic acid, p-cresol sulfate, phenyl-β-d-glucuronide, 4-ethylphenyl sulfate and 3-indolyl-β-d-glucopyranoside was found. These results show that some UTs could not be completely removed by hemodialysis. In addition, some biomarkers of different types of UTs are proposed for evaluating hemodialysis effectiveness.
Collapse
Affiliation(s)
- Yan-Rong Ma
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China; Department of Pharmacy, the First Hospital of Lanzhou University, Lanzhou 730000 China
| | - Ming-Yan Xin
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Kan Li
- Department of Nephrology, the First Hospital of Lanzhou University, Lanzhou 730000 China
| | - Huan Wang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Zhi Rao
- Department of Pharmacy, the First Hospital of Lanzhou University, Lanzhou 730000 China
| | - Tian-Xi Liu
- Department of Nephrology, the First Hospital of Lanzhou University, Lanzhou 730000 China.
| | - Xin-An Wu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China; Department of Pharmacy, the First Hospital of Lanzhou University, Lanzhou 730000 China.
| |
Collapse
|
25
|
De Vos C, Lemarcq L, Dhondt A, Glorieux G, Van Biesen W, Eloot S. The impact of intradialytic cycling on the removal of protein-bound uraemic toxins: A randomised cross-over study. Int J Artif Organs 2020; 44:156-164. [PMID: 32820982 DOI: 10.1177/0391398820949880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The evidence on impact of intradialytic exercise on the removal of urea, is conflictive. Impact of exercise on kinetics of serum levels of protein-bound uraemic toxins, known to exert toxicity and to have kinetics dissimilar of those of urea, has so far not been explored. Furthermore, if any effect, the most optimal intensity, time point and/or required duration of intradialytic exercise to maximise removal remain obscure. We therefore studied the impact of different intradialytic cycling schedules on the removal of protein-bound uraemic toxins during haemodialysis (HD).This randomised cross-over study included seven stable patients who were dialysed with an FX800 dialyser during three consecutive midweek HD sessions of 240 min: (A) without cycling; (B) cycling for 60 min between 60th and 120th minutes of dialysis; and (C) cycling for 60 min between 150th and 210th minutes, with the same cycling load as in session B. Blood and dialysate flows were respectively 300 and 500 mL/min. Blood was sampled from the blood inlet at different time points, and dialysate was partially collected (300 mL/h). Small water soluble solutes and protein-bound toxins were quantified and intradialytic reduction ratios (RR) and overall removal were calculated per solute.Total solute removal and reduction ratios were not different between the three test sessions, except for the reduction ratios RR60-120 and RR150-210 for potassium.In conclusion, we add evidence to the existing literature that, regardless of the timing within the dialysis session, intradialytic exercise has no impact on small solute clearance, and demonstrated also a lack of impact for protein-bound solutes.
Collapse
|
26
|
Lim JH, Park Y, Yook JM, Choi SY, Jung HY, Choi JY, Park SH, Kim CD, Kim YL, Cho JH. Randomized controlled trial of medium cut-off versus high-flux dialyzers on quality of life outcomes in maintenance hemodialysis patients. Sci Rep 2020; 10:7780. [PMID: 32385307 PMCID: PMC7210312 DOI: 10.1038/s41598-020-64622-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 04/20/2020] [Indexed: 11/12/2022] Open
Abstract
Medium cut-off (MCO) dialyzers help remove larger middle molecules associated with symptoms related to the accumulation of uremic retention solutes. We investigated the effect of an MCO dialyzer on the improvement of quality of life (QOL) in maintenance hemodialysis (HD) patients. Forty-nine HD patients with high-flux dialysis were randomly assigned to either an MCO (Theranova 400, Baxter) or a high-flux (FX CorDiax 80 or 60, Fresenius Medical Care) dialyzer and completed the study. QOL was assessed at baseline and after 12 weeks of treatment using the Kidney Disease Quality of Life Short Form-36, and pruritus was assessed using a questionnaire and visual analog scale. The reduction ratios of middle molecules were also evaluated. Laboratory markers, including serum albumin, did not differ between the two groups after 12 weeks. Removals of kappa and lambda free light chains were greater for MCO dialyzer than high-flux dialyzer. The MCO group had higher scores than the high-flux group in the domains of physical functioning and physical role (75.2 ± 20.8 vs. 59.8 ± 30.1, P = 0.042; 61.5 ± 37.6 vs. 39.0 ± 39.6, P = 0.047, respectively), and the MCO group had lower mean scores for morning pruritus distribution and the frequency of scratching during sleep (1.29 ± 0.46 vs. 1.64 ± 0.64, P = 0.034; 0.25 ± 0.53 vs. 1.00 ± 1.47, P = 0.023, respectively). MCO dialyzers may improve patient-reported outcomes, particularly the physical components of QOL and uremic pruritus, in patients with high-flux dialyzers.
Collapse
Affiliation(s)
- Jeong-Hoon Lim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Yeongwoo Park
- Department of Statistics, Kyungpook National University, Daegu, South Korea
| | - Ju-Min Yook
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Soon-Youn Choi
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Hee-Yeon Jung
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Ji-Young Choi
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Sun-Hee Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Chan-Duck Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Yong-Lim Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Jang-Hee Cho
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea.
| |
Collapse
|
27
|
Huang C, Ren J, Ji F, Muyldermans S, Jia L. Nanobody-Based high-performance immunosorbent for selective beta 2-microglobulin purification from blood. Acta Biomater 2020; 107:232-241. [PMID: 32092428 DOI: 10.1016/j.actbio.2020.02.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/14/2020] [Accepted: 02/18/2020] [Indexed: 12/18/2022]
Abstract
Removing β2-microglobulin (β2M) from blood circulation is considered to be the most effective method to delay the occurrence of dialysis-related amyloidosis (DRA). The ideal extracorporeal β2M removal system should be cost-effective, highly specific and having a high capacity. However, the traditional technologies based on size exclusion do not have an adequate specificity, and alternative immunosorbents have limited applications due to low capacity and their high cost. Nanobodies (Nbs), the smallest functional recombinant antibody fragments, offer several advantages to overcome these obstacles. In this study, an anti-β2M Nb with a C-terminal thiol-tag was successfully prepared from E. coli for site-directed and oriented immobilization and usage as capture ligand in a β2M-selective immunosorbent. The prepared immunosorbent showed a high binding capacity of up to 7 mg β2M per mL resin, which is 17 times higher than that of previous studies using single-chain variable antibody fragments (scFv). Furthermore, an exceptional high specificity has been demonstrated as other human serum proteins were not adsorbed during dynamic adsorption experiments. About 80% of the original binding capacity of the immunosorbent was restored after four consecutive easy regenerations, whereas 90% of the original capacity was retained after 1-month storage of the resin. Moreover, the mathematical model fitted very well the in vitro perfusion. The results with this pioneering immunosorbent confirm its possible clinical application and is expected to reach the required clinical effect of immunoadsorption therapy. STATEMENT OF SIGNIFICANCE: Dialysis-related amyloidosis (DRA), associated with the accumulation of β2-microglobulin (β2M), is a serious complication of end-stage kidney disease. Removing β2M from blood circulation by extracorporeal blood purification is considered to be the most effective method to delay the occurrence of DRA. However, the existing methods are incapable to eliminate sufficient quantities of β2M from circulation, either because of lack of specificity, high cost or for low capacity. In this manuscript, we provide a practical and economic immunosorbent based on anti-β2M nanobody for DRA. The prepared immunosorbent was reusable and storable, and demonstrated high specificity and realized a high binding capacity of up to 7 mg β2M per mL resin, which is 17 times higher than that of the previous studies.
Collapse
|
28
|
Nigam SK, Bush KT, Bhatnagar V, Poloyac SM, Momper JD. The Systems Biology of Drug Metabolizing Enzymes and Transporters: Relevance to Quantitative Systems Pharmacology. Clin Pharmacol Ther 2020; 108:40-53. [PMID: 32119114 PMCID: PMC7292762 DOI: 10.1002/cpt.1818] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/12/2020] [Indexed: 12/19/2022]
Abstract
Quantitative systems pharmacology (QSP) has emerged as a transformative science in drug discovery and development. It is now time to fully rethink the biological functions of drug metabolizing enzymes (DMEs) and transporters within the framework of QSP models. The large set of DME and transporter genes are generally considered from the perspective of the absorption, distribution, metabolism, and excretion (ADME) of drugs. However, there is a growing amount of data on the endogenous physiology of DMEs and transporters. Recent studies—including systems biology analyses of “omics” data as well as metabolomics studies—indicate that these enzymes and transporters, which are often among the most highly expressed genes in tissues like liver, kidney, and intestine, have coordinated roles in fundamental biological processes. Multispecific DMEs and transporters work together with oligospecific and monospecific ADME proteins in a large multiorgan remote sensing and signaling network. We use the Remote Sensing and Signaling Theory (RSST) to examine the roles of DMEs and transporters in intratissue, interorgan, and interorganismal communication via metabolites and signaling molecules. This RSST‐based view is applicable to bile acids, uric acid, eicosanoids, fatty acids, uremic toxins, and gut microbiome products, among other small organic molecules of physiological interest. Rooting this broader perspective of DMEs and transporters within QSP may facilitate an improved understanding of fundamental biology, physiologically based pharmacokinetics, and the prediction of drug toxicities based upon the interplay of these ADME proteins with key pathways in metabolism and signaling. The RSST‐based view should also enable more tailored pharmacotherapy in the setting of kidney disease, liver disease, metabolic syndrome, and diabetes. We further discuss the pharmaceutical and regulatory implications of this revised view through the lens of systems physiology.
Collapse
Affiliation(s)
- Sanjay K Nigam
- Departments of Pediatrics and Medicine, School of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Kevin T Bush
- Departments of Pediatrics and Medicine, School of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Vibha Bhatnagar
- Department of Family Medicine and Public Health, School of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Samuel M Poloyac
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jeremiah D Momper
- Division of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
29
|
Serum Levels and Removal by Haemodialysis and Haemodiafiltration of Tryptophan-Derived Uremic Toxins in ESKD Patients. Int J Mol Sci 2020; 21:ijms21041522. [PMID: 32102247 PMCID: PMC7073230 DOI: 10.3390/ijms21041522] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/19/2020] [Accepted: 02/21/2020] [Indexed: 12/30/2022] Open
Abstract
Tryptophan is an essential dietary amino acid that originates uremic toxins that contribute to end-stage kidney disease (ESKD) patient outcomes. We evaluated serum levels and removal during haemodialysis and haemodiafiltration of tryptophan and tryptophan-derived uremic toxins, indoxyl sulfate (IS) and indole acetic acid (IAA), in ESKD patients in different dialysis treatment settings. This prospective multicentre study in four European dialysis centres enrolled 78 patients with ESKD. Blood and spent dialysate samples obtained during dialysis were analysed with high-performance liquid chromatography to assess uremic solutes, their reduction ratio (RR) and total removed solute (TRS). Mean free serum tryptophan and IS concentrations increased, and concentration of IAA decreased over pre-dialysis levels (67%, 49%, −0.8%, respectively) during the first hour of dialysis. While mean serum total urea, IS and IAA concentrations decreased during dialysis (−72%, −39%, −43%, respectively), serum tryptophan levels increased, resulting in negative RR (−8%) towards the end of the dialysis session (p < 0.001), despite remarkable Trp losses in dialysate. RR and TRS values based on serum (total, free) and dialysate solute concentrations were lower for conventional low-flux dialysis (p < 0.001). High-efficiency haemodiafiltration resulted in 80% higher Trp losses than conventional low-flux dialysis, despite similar neutral Trp RR values. In conclusion, serum Trp concentrations and RR behave differently from uremic solutes IS, IAA and urea and Trp RR did not reflect dialysis Trp losses. Conventional low-flux dialysis may not adequately clear Trp-related uremic toxins while high efficiency haemodiafiltration increased Trp losses.
Collapse
|
30
|
Zhang R, Saredy J, Shao Y, Yao T, Liu L, Saaoud F, Yang WY, Sun Y, Johnson C, Drummer C, Fu H, Lu Y, Xu K, Liu M, Wang J, Cutler E, Yu D, Jiang X, Li Y, Li R, Wang L, Choi ET, Wang H, Yang X. End-stage renal disease is different from chronic kidney disease in upregulating ROS-modulated proinflammatory secretome in PBMCs - A novel multiple-hit model for disease progression. Redox Biol 2020; 34:101460. [PMID: 32179051 PMCID: PMC7327976 DOI: 10.1016/j.redox.2020.101460] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/28/2020] [Accepted: 02/07/2020] [Indexed: 12/17/2022] Open
Abstract
Background The molecular mechanisms underlying chronic kidney disease (CKD) transition to end-stage renal disease (ESRD) and CKD acceleration of cardiovascular and other tissue inflammations remain poorly determined. Methods We conducted a comprehensive data analyses on 7 microarray datasets in peripheral blood mononuclear cells (PBMCs) from patients with CKD and ESRD from NCBI-GEO databases, where we examined the expressions of 2641 secretome genes (SG). Results 1) 86.7% middle class (molecular weight >500 Daltons) uremic toxins (UTs) were encoded by SGs; 2) Upregulation of SGs in PBMCs in patients with ESRD (121 SGs) were significantly higher than that of CKD (44 SGs); 3) Transcriptomic analyses of PBMC secretome had advantages to identify more comprehensive secretome than conventional secretomic analyses; 4) ESRD-induced SGs had strong proinflammatory pathways; 5) Proinflammatory cytokines-based UTs such as IL-1β and IL-18 promoted ESRD modulation of SGs; 6) ESRD-upregulated co-stimulation receptors CD48 and CD58 increased secretomic upregulation in the PBMCs, which were magnified enormously in tissues; 7) M1-, and M2-macrophage polarization signals contributed to ESRD- and CKD-upregulated SGs; 8) ESRD- and CKD-upregulated SGs contained senescence-promoting regulators by upregulating proinflammatory IGFBP7 and downregulating anti-inflammatory TGF-β1 and telomere stabilizer SERPINE1/PAI-1; 9) ROS pathways played bigger roles in mediating ESRD-upregulated SGs (11.6%) than that in CKD-upregulated SGs (6.8%), and half of ESRD-upregulated SGs were ROS-independent. Conclusions Our analysis suggests novel secretomic upregulation in PBMCs of patients with CKD and ESRD, act synergistically with uremic toxins, to promote inflammation and potential disease progression. Our findings have provided novel insights on PBMC secretome upregulation to promote disease progression and may lead to the identification of new therapeutic targets for novel regimens for CKD, ESRD and their accelerated cardiovascular disease, other inflammations and cancers. (Total words: 279).
Collapse
Affiliation(s)
- Ruijing Zhang
- Center for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA; Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030013, China; Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030012, China
| | - Jason Saredy
- Centers for Metabolic Disease Research, Cardiovascular Research, & Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Ying Shao
- Center for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Tian Yao
- Shanxi Medical University, Taiyuan, Shanxi Province, 030001, China
| | - Lu Liu
- Centers for Metabolic Disease Research, Cardiovascular Research, & Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Fatma Saaoud
- Center for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | | | - Yu Sun
- Center for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Candice Johnson
- Center for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Charles Drummer
- Center for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Hangfei Fu
- Center for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Yifan Lu
- Center for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Keman Xu
- Center for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Ming Liu
- Center for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA; Shanxi Medical University, Taiyuan, Shanxi Province, 030001, China
| | - Jirong Wang
- Center for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Elizabeth Cutler
- Center for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA; School of Science and Engineering, Tulane University, New Orleans, LA, 70118, USA
| | - Daohai Yu
- Department of Clinical Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Xiaohua Jiang
- Center for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Yafeng Li
- Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030012, China
| | - Rongshan Li
- Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030012, China
| | - Lihua Wang
- Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030013, China
| | - Eric T Choi
- Division of Vascular and Endovascular Surgery, Department of Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA; Centers for Metabolic Disease Research, Cardiovascular Research, & Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA; Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Hong Wang
- Centers for Metabolic Disease Research, Cardiovascular Research, & Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA; Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Xiaofeng Yang
- Center for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA; Centers for Metabolic Disease Research, Cardiovascular Research, & Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA; Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
31
|
Shen Y, Wang Y, Shi Y, Bi X, Xu J, Zhu Q, Ding F. Improving the clearance of protein-bound uremic toxins using cationic liposomes as an adsorbent in dialysate. Colloids Surf B Biointerfaces 2019; 186:110725. [PMID: 31862563 DOI: 10.1016/j.colsurfb.2019.110725] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/26/2019] [Accepted: 12/12/2019] [Indexed: 12/19/2022]
Abstract
Anionic and protein-bound uremic toxins, represented by indoxyl sulfate (IS), may be associated with cardiovascular outcomes and the progression of chronic kidney disease in cases of injured kidney function and are not easily cleared by traditional dialysis therapy. We fabricated cationic liposomes that were modified with polyethyleneimine (PEI), octadecylamine (Oct), and hexadecyl trimethyl ammonium bromide (CTAB), and evaluated the effects on the clearance of the representative protein-bound uremic toxins (PBUTs). The binding rate was obtained by ultrafiltration and in vitro dialysis was performed in a Rapid Equilibrium Dialysis (RED) device to assay the clearing efficiency of the dialysate supported by three types of cationic liposomes. The cationic liposomes showed a higher binding rate with IS (1.24-1.38 fold higher) and p-cresol (1.07-1.09 fold higher) than in the unmodified plain liposomes. The dialysate supported by cationic liposomes also exhibited better clearing efficiency for IS (PEI-20: 57.65 ± 1.74 %; Oct-5: 62.80 ± 0.69 %; CTAB-10: 66.54 ± 0.91 %; p < 0.05) and p-cresol (PEI-20: 67.05 ± 3.09 %; Oct-5: 79.26 ± 0.43 %; CTAB-5: 68.45 ± 1.72 %; p < 0.05) than for phosphate buffer saline (IS: 29.70 ± 2.38 %; p-cresol: 33.59 ± 3.44 %) or dialysate supported by bovine serum albumin (IS: 50.00 ± 4.01 %; p-cresol: 53.06 ± 0.97 %). In conclusion, cationic liposomes are efficient in the clearance of anionic PBUTs, and these modified liposomes suggest a potential application in blood purification.
Collapse
Affiliation(s)
- Yue Shen
- Department of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Yifeng Wang
- Department of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Yuanyuan Shi
- Department of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Xiao Bi
- Department of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Jingyi Xu
- Department of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Qiuyu Zhu
- Department of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Feng Ding
- Department of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China.
| |
Collapse
|
32
|
Bennis Y, Cluet Y, Titeca-Beauport D, El Esper N, Ureña P, Bodeau S, Combe C, Dussol B, Fouque D, Choukroun G, Liabeuf S. The Effect of Sevelamer on Serum Levels of Gut-Derived Uremic Toxins: Results from In Vitro Experiments and A Multicenter, Double-Blind, Placebo-Controlled, Randomized Clinical Trial. Toxins (Basel) 2019; 11:toxins11050279. [PMID: 31109001 PMCID: PMC6563242 DOI: 10.3390/toxins11050279] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 02/07/2023] Open
Abstract
High serum levels of gut-derived uremic toxins, especially p-cresyl sulfate (pCS), indoxyl sulfate (IS) and indole acetic acid (IAA), have been linked to adverse outcomes in patients with chronic kidney disease (CKD). Sevelamer carbonate could represent an interesting option to limit the elevation of gut-derived uremic toxins. The aim of the present study was to evaluate the adsorptive effect of sevelamer carbonate on different gut-derived protein-bound uremic toxins or their precursors in vitro, and its impact on the serum levels of pCS, IS and IAA in patients with CKD stage 3b/4. For the in vitro experiments, IAA, p-cresol (precursor of pCS) and indole (precursor of IS), each at a final concentration of 1 or 10 µg/mL, were incubated in centrifugal 30 kDa filter devices with 3 or 15 mg/mL sevelamer carbonate in phosphate-buffered saline at a pH adjusted to 6 or 8. Then, samples were centrifuged and free uremic toxins in the filtrates were analyzed. As a control experiment, the adsorption of phosphate was also evaluated. Additionally, patients with stage 3b/4 CKD (defined as an eGFR between 15 and 45 mL/min per 1.73 m2) were included in a multicenter, double-blind, placebo-controlled, randomized clinical trial. The participants received either placebo or sevelamer carbonate (4.8 g) three times a day for 12 weeks. The concentrations of the toxins and their precursors were measured using a validated high-performance liquid chromatography method with a diode array detector. In vitro, regardless of the pH and concentration tested, sevelamer carbonate did not show adsorption of indole and p-cresol. Conversely, with 10 µg/mL IAA, use of a high concentration of sevelamer carbonate (15 mg/mL) resulted in a significant toxin adsorption both at pH 8 (mean reduction: 26.3 ± 3.4%) and pH 6 (mean reduction: 38.7 ± 1.7%). In patients with CKD stage 3b/4, a 12-week course of treatment with sevelamer carbonate was not associated with significant decreases in serum pCS, IS and IAA levels (median difference to baseline levels: −0.12, 0.26 and −0.06 µg/mL in the sevelamer group vs. 1.97, 0.38 and 0.05 µg/mL in the placebo group, respectively). Finally, in vitro, sevelamer carbonate was capable of chelating a gut-derived uremic toxin IAA but not p-cresol and indole, the precursors of pCS and IS in the gut. In a well-designed clinical study of patients with stage 3b/4 CKD, a 12-week course of treatment with sevelamer carbonate was not associated with significant changes in the serum concentrations of pCS, IS and IAA.
Collapse
Affiliation(s)
- Youssef Bennis
- Pharmacology Department, Amiens University Hospital, 80000 Amiens, France.
- MP3CV Laboratory, EA7517, University of Picardie Jules Verne, 80000 Amiens, France.
| | - Yan Cluet
- Pharmacology Department, Amiens University Hospital, 80000 Amiens, France.
- MP3CV Laboratory, EA7517, University of Picardie Jules Verne, 80000 Amiens, France.
| | - Dimitri Titeca-Beauport
- MP3CV Laboratory, EA7517, University of Picardie Jules Verne, 80000 Amiens, France.
- Nephrology Department, Amiens University Hospital, 80000 Amiens, France.
| | - Najeh El Esper
- Nephrology Department, Amiens University Hospital, 80000 Amiens, France.
| | - Pablo Ureña
- Department of Nephrology and Dialysis, AURA Nord Saint Ouen, 93400 Saint Ouen, France.
| | - Sandra Bodeau
- Pharmacology Department, Amiens University Hospital, 80000 Amiens, France.
- MP3CV Laboratory, EA7517, University of Picardie Jules Verne, 80000 Amiens, France.
| | - Christian Combe
- Nephrology Department, Bordeaux University Hospital, 33000 Bordeaux, France.
| | - Bertrand Dussol
- Clinical Inverstigation Center, Aix Marseille University, 13354 Marseille, France.
| | - Denis Fouque
- Dept Nephrology, Université de Lyon, Hospital Lyon Sud, F-69495 Pierre-Benite, France.
| | - Gabriel Choukroun
- MP3CV Laboratory, EA7517, University of Picardie Jules Verne, 80000 Amiens, France.
- Nephrology Department, Amiens University Hospital, 80000 Amiens, France.
| | - Sophie Liabeuf
- Pharmacology Department, Amiens University Hospital, 80000 Amiens, France.
- MP3CV Laboratory, EA7517, University of Picardie Jules Verne, 80000 Amiens, France.
| |
Collapse
|
33
|
Adesso S, Ruocco M, Rapa SF, Piaz FD, Raffaele Di Iorio B, Popolo A, Autore G, Nishijima F, Pinto A, Marzocco S. Effect of Indoxyl Sulfate on the Repair and Intactness of Intestinal Epithelial Cells: Role of Reactive Oxygen Species' Release. Int J Mol Sci 2019; 20:ijms20092280. [PMID: 31072046 PMCID: PMC6539031 DOI: 10.3390/ijms20092280] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/05/2019] [Accepted: 05/06/2019] [Indexed: 12/17/2022] Open
Abstract
Chronic kidney disease (CKD) is characterized by an oxidative stress status, driving some CKD-associated complications, even at the gastrointestinal level. Indoxyl Sulfate (IS) is a protein-bound uremic toxin, poorly eliminated by dialysis. This toxin is able to affect the intestinal system, but its molecular mechanism/s in intestinal epithelial cells (IECs) remain poorly understood. This study's aim was to evaluate the effect of IS (31.2-250 µM) on oxidative stress in IEC-6 cells and on the intactness of IECs monolayers. Our results indicated that IS enhanced oxidative cell damage by inducing reactive oxygen species (ROS) release, reducing the antioxidant response and affecting Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) nuclear translocation as well its related antioxidant enzymes. In the wound healing assay model, IS reduced IEC-6 migration, slightly impaired actin cytoskeleton rearrangement; this effect was associated with connexin 43 alteration. Moreover, we reported the effect of CKD patients' sera in IEC-6 cells. Our results indicated that patient sera induced ROS release in IEC-6 cells directly related to IS sera content and this effect was reduced by AST-120 serum treatment. Results highlighted the effect of IS in inducing oxidative stress in IECs and in impairing the intactness of the IECs cell monolayer, thus significantly contributing to CKD-associated intestinal alterations.
Collapse
Affiliation(s)
- Simona Adesso
- Department of Pharmacy, University of Salerno, I-84084, Fisciano, Salerno, Italy.
| | - Marco Ruocco
- Department of Pharmacy, University of Salerno, I-84084, Fisciano, Salerno, Italy.
| | - Shara Francesca Rapa
- Department of Pharmacy, University of Salerno, I-84084, Fisciano, Salerno, Italy.
| | - Fabrizio Dal Piaz
- Department of Medicine, University of Salerno, I-84084 Fisciano, Salerno, Italy.
| | | | - Ada Popolo
- Department of Pharmacy, University of Salerno, I-84084, Fisciano, Salerno, Italy.
| | - Giuseppina Autore
- Department of Pharmacy, University of Salerno, I-84084, Fisciano, Salerno, Italy.
| | - Fuyu Nishijima
- Pharmaceuticals Division, Kureha Corporation, 169-8503 Tokyo, Japan.
| | - Aldo Pinto
- Department of Pharmacy, University of Salerno, I-84084, Fisciano, Salerno, Italy.
| | - Stefania Marzocco
- Department of Pharmacy, University of Salerno, I-84084, Fisciano, Salerno, Italy.
| |
Collapse
|
34
|
Hulko M, Dietrich V, Koch I, Gekeler A, Gebert M, Beck W, Krause B. Pyrogen retention: Comparison of the novel medium cut-off (MCO) membrane with other dialyser membranes. Sci Rep 2019; 9:6791. [PMID: 31043670 PMCID: PMC6494805 DOI: 10.1038/s41598-019-43161-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 04/03/2019] [Indexed: 11/09/2022] Open
Abstract
Haemodialysis effectively removes small solutes and smaller-sized middle molecules from the blood; however, the clearance of larger middle molecules, which have been associated with negative effects, is poor. The novel medium cut-off (MCO) dialysis membrane has larger pore sizes and a more open structure than other high-flux membranes, providing improved removal of larger middle molecules while retaining albumin. However, larger pore sizes may potentially increase permeability to pyrogens, including endotoxins and other bacterial contaminants, that could be present in the dialysis fluid. In this study, we tested the capacity of low-flux, high-flux, MCO and high cut-off dialyser membranes with different pore sizes to prevent pyrogens crossing from dialysate to the blood side in a closed-loop test system, differentiating among lipopolysaccharides, peptidoglycans and bacterial DNA using a toll-like receptor assay. Even though the bacterial contamination levels in our test system exceeded the acceptable pyrogen dose for standard dialysis fluid, levels of lipopolysaccharides, peptidoglycans and bacterial DNA in the blood-side samples were too low to identify potential differences in pyrogen permeability among the membranes. Our results suggest that MCO membranes are suitable for haemodialysis using ISO standard dialysis fluid quality, and retain endotoxins at a similar level as other membranes.
Collapse
Affiliation(s)
- Michael Hulko
- Baxter International Inc., Research & Development, Holger-Crafoord-Str. 26, 72379, Hechingen, Germany.
| | - Verena Dietrich
- Baxter International Inc., Research & Development, Holger-Crafoord-Str. 26, 72379, Hechingen, Germany
| | - Ilona Koch
- Baxter International Inc., Research & Development, Holger-Crafoord-Str. 26, 72379, Hechingen, Germany
| | - Alexander Gekeler
- Baxter International Inc., Research & Development, Holger-Crafoord-Str. 26, 72379, Hechingen, Germany
| | - Michael Gebert
- Baxter International Inc., Research & Development, Holger-Crafoord-Str. 26, 72379, Hechingen, Germany
| | - Werner Beck
- Baxter International Inc., Research & Development, Holger-Crafoord-Str. 26, 72379, Hechingen, Germany
| | - Bernd Krause
- Baxter International Inc., Research & Development, Holger-Crafoord-Str. 26, 72379, Hechingen, Germany
| |
Collapse
|
35
|
Abstract
Dialyzer clearance of urea multiplied by dialysis time and normalized for urea distribution volume (Kt/Vurea or simply Kt/V) has been used as an index of dialysis adequacy since more than 30 years. This article reviews the flaws of Kt/V, starting with a lack of proof of concept in three randomized controlled hard outcome trials (RCTs), and continuing with a long list of conditions where the concept of Kt/V was shown to be flawed. This information leaves little room for any conclusion other than that Kt/V, as an indicator of dialysis adequacy, is obsolete. The dialysis patient might benefit more if, instead, the nephrology community concentrates in the future on pursuing the optimal dialysis dose that conforms with adequate quality of life and on factors that are likely to affect outcomes more than Kt/V. These include residual renal function, volume status, dialysis length, ultrafiltration rate, the number of intra-dialytic hypotensive episodes, interdialytic blood pressure, serum potassium and phosphate, serum albumin, and C reactive protein.
Collapse
Affiliation(s)
- Raymond Vanholder
- Nephrology Section, Department of Internal Medicine, University Hospital Ghent, Ghent, Belgium
| | - Wim Van Biesen
- Nephrology Section, Department of Internal Medicine, University Hospital Ghent, Ghent, Belgium
| | - Norbert Lameire
- Nephrology Section, Department of Internal Medicine, University Hospital Ghent, Ghent, Belgium
| |
Collapse
|
36
|
Tohyama K, Chisaki I, Takai Y, Handa Y, Miyamoto M, Amano N. Relationship of MATE1 Inhibition and Cytotoxicity in Nephrotoxicity: Application for Safety Evaluation in Early Drug Discovery. Toxicol Sci 2019; 170:223-233. [DOI: 10.1093/toxsci/kfz093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Kimio Tohyama
- Drug Metabolism and Pharmacokinetics Research Laboratories
| | - Ikumi Chisaki
- Drug Metabolism and Pharmacokinetics Research Laboratories
| | | | - Yasuhiro Handa
- Biomolecular Research Laboratories, Research, Takeda Pharmaceutical Company, Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | | | - Nobuyuki Amano
- Drug Metabolism and Pharmacokinetics Research Laboratories
| |
Collapse
|
37
|
Uremic Toxin Concentrations are Related to Residual Kidney Function in the Pediatric Hemodialysis Population. Toxins (Basel) 2019; 11:toxins11040235. [PMID: 31022857 PMCID: PMC6521157 DOI: 10.3390/toxins11040235] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/19/2019] [Accepted: 04/19/2019] [Indexed: 12/15/2022] Open
Abstract
Protein-bound uremic toxins (PBUTs) play a role in the multisystem disease that children on hemodialysis (HD) are facing, but little is known about their levels and protein binding (%PB). In this study, we evaluated the levels and %PB of six PBUTs cross-sectionally in a large pediatric HD cohort (n = 170) by comparing these with healthy and non-dialysis chronic kidney disease (CKD) stage 4-5 (n = 24) children. In parallel β2-microglobulin (β2M) and uric acid (UA) were evaluated. We then explored the impact of age and residual kidney function on uremic toxin levels and %PB using analysis of covariance and Spearman correlation coefficients (rs). We found higher levels of β2M, p-cresyl glucuronide (pCG), hippuric acid (HA), indole acetic acid (IAA), and indoxyl sulfate (IxS) in the HD compared to the CKD4-5 group. In the HD group, a positive correlation between age and pCG, HA, IxS, and pCS levels was shown. Residual urine volume was negatively correlated with levels of β2M, pCG, HA, IAA, IxS, and CMPF (rs -0.2 to -0.5). In addition, we found overall lower %PB of PBUTs in HD versus the CKD4-5 group, and showed an age-dependent increase in %PB of IAA, IxS, and pCS. Furhtermore, residual kidney function was overall positively correlated with %PB of PBUTs. In conclusion, residual kidney function and age contribute to PBUT levels and %PB in the pediatric HD population.
Collapse
|
38
|
Li J, Wang Y, Xu X, Cao W, Shen Z, Wang N, Leng J, Zou N, Shang E, Zhu Z, Guo J, Duan J. Improved dialysis removal of protein-bound uremic toxins by salvianolic acids. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 57:166-173. [PMID: 30772752 DOI: 10.1016/j.phymed.2018.12.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/20/2018] [Accepted: 12/15/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Indoxyl sulfate (IS) and p-cresyl sulfate (pCS) are two key protein-bound uremic toxins that accumulate in patients with end-stage renal disease. IS and pCS cannot be efficiently removed by conventional hemodialysis because they are highly bound to proteins. One promising means to optimize the removal of protein-bound uremic toxins involves using binding competitors to liberate uremic toxins from protein-binding partners. PURPOSE In this study, we try to identify potential binding competitors that can enhance the dialysis removal of IS and pCS in natural compounds of phytomedicine. METHODS We employed microdialysis to evaluate whether Danhong injection (DHI) and its salvianolic acids can increase the free fractions of IS and pCS and thus improve their dialysis efficiency in vitro. Furthermore, we confirmed the positive effects of DHI and salvianolic acids in vivo on chronic kidney disease model rats in which IS and pCS had heavily accumulated. RESULTS DHI significantly increased the dialysis efficiency of IS and pCS by 99.13% and 142.00% in vitro (10-fold dilution), respectively, and by 135.61% and 272.13% in vivo (4.16 ml/kg). Salvianolic acids including lithospermic acid (LA), salvianolic acid A (SaA), tanshinol (DSS), caffeic acid (CA), salvianolic acid B (SaB), protocatechuic aldehyde (PA) and rosmarinic acid (RA) significantly enhanced the dialysis removal of IS and pCS in a concentration-dependent manner. LA, the best competitor of the tested salvianolic acids, increased dialysis efficiency levels of IS and pCS by 197.23% and 198.31% in vitro (400 μM), respectively, and by 119.55% and 127.56% in vivo (24.69 mg/kg). CONCLUSION The removal of protein-bound uremic toxins IS and pCS using DHI or salvianolic acids as protein-bound competitors is superior to previously reported strategies and drugs and may contribute to clinical hemodialysis therapeutic practice.
Collapse
Affiliation(s)
- Jianping Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yingyi Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xuejun Xu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wenjuan Cao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhiqiang Shen
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Nan Wang
- Pharmaceutical Department, Nanjing General Hospital of People's Liberation Army, Nanjing 210002, China
| | - Jing Leng
- Pharmaceutical Department, Nanjing General Hospital of People's Liberation Army, Nanjing 210002, China
| | - Ning Zou
- Blood Purification Center, Qingdao Municipal Hospital, Qingdao 266000, China
| | - Erxin Shang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhenhua Zhu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jianming Guo
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Jinao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
39
|
Protein-Bound Solute Clearance During Hemodialysis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1153:69-77. [DOI: 10.1007/5584_2019_336] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
40
|
Macías N, Vega A, Abad S, Aragoncillo I, García-Prieto AM, Santos A, Torres E, Luño J. Middle molecule elimination in expanded haemodialysis: only convective transport? Clin Kidney J 2018; 12:447-455. [PMID: 31198548 PMCID: PMC6543970 DOI: 10.1093/ckj/sfy097] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Indexed: 12/25/2022] Open
Abstract
Background New high-retention onset dialysers have shown improved efficacy in the elimination of uraemic toxins, and their depurative capacity has been compared with high convective volumes of online haemodiafiltration. Haemodialysis (HD) using high-flux membranes leads to convective transport by internal filtration [direct filtration (DF)/backfiltration (BF)] and allows the removal of middle molecules (MMs). The aim of this study was to assess solute transport mechanisms in expanded HD (HDx). Methods In 14 4-h HDx sessions with Theranova-500 dialysers under similar dialysis conditions (blood flow 400 mL/min, dialysate flow 700 mL/min, dialysate temperature 35.5°C), pressures at the inlet and outlet of both dialyser compartments (P bi, P bo, P di and P do) were collected hourly to estimate DF/BF volumes by semi-empirical methods. Uraemic toxins with various molecular weights were measured pre-dialysis, at 1 h (pre-filter and post-filter) and post-dialysis to calculate molecules' reduction over time and dialyser in vivo clearances. Results Ultrafiltration was 1.47 ± 0.9 L and Kt/V 1.74 ± 0.3. Hydrodynamic data (P bi: 259 ± 39, P bo: 155 ± 27, P di: 271 ± 30, P do: 145 ± 29 mmHg and oncotic pressure 22.0 ± 3.5 mmHg) allowed the estimation of DF/BF rates. DF flow ranged from 29.5 ± 4.2 to 31.3 ± 3.9 mL/min and BF flow ranged from 25.1 ± 2.3 to 23.4 ± 2.6 mL/min. The highest calculated DF volume was 7506.8 ± 935.3 mL/session. Diffusive clearances (K d) of all solutes were higher than their convective transport (all P < 0.001) except for prolactin (23 kDa) clearances, which showed no differences. Total clearances of all solutes were correlated with their K d (ρ = 0.899-0.987, all P < 0.001) and Kt/V correlated with all reduction rates (ρ = 0.661-0.941, P = 0.010 to <0.001). DF flow was only associated with urea (ρ = -0.793, P = 0.001), creatinine (ρ = -0.675, P = 0.008) and myoglobin clearance (ρ = 0.653, P = 0.011). Conclusion Results suggest that diffusive transport is a main mechanism of MM elimination in HDx. HDx offers an efficient depuration of MM without the need for high convective volumes.
Collapse
Affiliation(s)
- Nicolás Macías
- Department of Nephrology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Almudena Vega
- Department of Nephrology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Soraya Abad
- Department of Nephrology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Inés Aragoncillo
- Department of Nephrology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | | - Alba Santos
- Department of Nephrology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Esther Torres
- Department of Nephrology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Jose Luño
- Department of Nephrology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| |
Collapse
|
41
|
Yamagami F, Tajiri K, Doki K, Hattori M, Honda J, Aita S, Harunari T, Yamasaki H, Murakoshi N, Sekiguchi Y, Homma M, Takahashi N, Aonuma K, Nogami A, Ieda M. Indoxyl Sulphate is Associated with Atrial Fibrillation Recurrence after Catheter Ablation. Sci Rep 2018; 8:17276. [PMID: 30467393 PMCID: PMC6250674 DOI: 10.1038/s41598-018-35226-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/01/2018] [Indexed: 12/27/2022] Open
Abstract
Renal dysfunction results in the accumulation of various uremic toxins, including indoxyl sulphate (IS), and is a major risk factor for atrial fibrillation (AF). Experimental studies have demonstrated that IS exacerbates atrial remodelling via oxidative stress, inflammation, and fibrosis. However, its clinical impact on AF-promoting cardiac remodelling has not been described. Therefore, the purpose of this study was to clarify the relationship between basal IS levels and the 1-year outcomes after catheter ablation for the treatment of AF. Our prospective observational study included data from 125 patients with AF who underwent catheter ablation. Over a 1-year follow-up period, AF recurrence was identified in 21 patients. The 1-year AF-free survival was significantly lower in patients with high serum IS levels (≥0.65 μg/mL) than in those with low IS levels (60.1 ± 10.4% versus 85.2 ± 3.9%, P = 0.007). Univariable analysis identified that an IS concentration ≥ 0.65 μg/mL was associated with AF recurrence (hazard ratio [HR] = 3.10 [1.26-7.32], P = 0.015), and this association was maintained in multivariate analysis (HR = 3.67 [1.13-11.7], P = 0.031). Thus, in patients undergoing AF ablation, serum IS levels at baseline independently predict the recurrence of arrhythmia.
Collapse
Affiliation(s)
- Fumi Yamagami
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kazuko Tajiri
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.
| | - Kosuke Doki
- Department of Pharmaceutical Sciences, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Masayuki Hattori
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Junya Honda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Satoshi Aita
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Tomohiko Harunari
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiro Yamasaki
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Nobuyuki Murakoshi
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yukio Sekiguchi
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Masato Homma
- Department of Pharmaceutical Sciences, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Naohiko Takahashi
- Department of Cardiology and Clinical Examination, Oita University Faculty of Medicine, Oita, Japan
| | - Kazutaka Aonuma
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Akihiko Nogami
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Masaki Ieda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
42
|
Legallais C, Kim D, Mihaila SM, Mihajlovic M, Figliuzzi M, Bonandrini B, Salerno S, Yousef Yengej FA, Rookmaaker MB, Sanchez Romero N, Sainz-Arnal P, Pereira U, Pasqua M, Gerritsen KGF, Verhaar MC, Remuzzi A, Baptista PM, De Bartolo L, Masereeuw R, Stamatialis D. Bioengineering Organs for Blood Detoxification. Adv Healthc Mater 2018; 7:e1800430. [PMID: 30230709 DOI: 10.1002/adhm.201800430] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 08/23/2018] [Indexed: 12/11/2022]
Abstract
For patients with severe kidney or liver failure the best solution is currently organ transplantation. However, not all patients are eligible for transplantation and due to limited organ availability, most patients are currently treated with therapies using artificial kidney and artificial liver devices. These therapies, despite their relative success in preserving the patients' life, have important limitations since they can only replace part of the natural kidney or liver functions. As blood detoxification (and other functions) in these highly perfused organs is achieved by specialized cells, it seems relevant to review the approaches leading to bioengineered organs fulfilling most of the native organ functions. There, the culture of cells of specific phenotypes on adapted scaffolds that can be perfused takes place. In this review paper, first the functions of kidney and liver organs are briefly described. Then artificial kidney/liver devices, bioartificial kidney devices, and bioartificial liver devices are focused on, as well as biohybrid constructs obtained by decellularization and recellularization of animal organs. For all organs, a thorough overview of the literature is given and the perspectives for their application in the clinic are discussed.
Collapse
Affiliation(s)
- Cécile Legallais
- UMR CNRS 7338 Biomechanics & Bioengineering; Université de technologie de Compiègne; Sorbonne Universités; 60203 Compiègne France
| | - Dooli Kim
- (Bio)artificial organs; Department of Biomaterials Science and Technology; Faculty of Science and Technology; TechMed Institute; University of Twente; P.O. Box 217 7500 AE Enschede The Netherlands
| | - Sylvia M. Mihaila
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Universiteitsweg 99 3584 CG Utrecht The Netherlands
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Milos Mihajlovic
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Marina Figliuzzi
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri; via Stezzano 87 24126 Bergamo Italy
| | - Barbara Bonandrini
- Department of Chemistry; Materials and Chemical Engineering “Giulio Natta”; Politecnico di Milano; Piazza Leonardo da Vinci 32 20133 Milan Italy
| | - Simona Salerno
- Institute on Membrane Technology; National Research Council of Italy; ITM-CNR; Via Pietro BUCCI, Cubo 17C - 87036 Rende Italy
| | - Fjodor A. Yousef Yengej
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Maarten B. Rookmaaker
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | | | - Pilar Sainz-Arnal
- Instituto de Investigación Sanitaria de Aragón (IIS Aragon); 50009 Zaragoza Spain
- Instituto Aragonés de Ciencias de la Salud (IACS); 50009 Zaragoza Spain
| | - Ulysse Pereira
- UMR CNRS 7338 Biomechanics & Bioengineering; Université de technologie de Compiègne; Sorbonne Universités; 60203 Compiègne France
| | - Mattia Pasqua
- UMR CNRS 7338 Biomechanics & Bioengineering; Université de technologie de Compiègne; Sorbonne Universités; 60203 Compiègne France
| | - Karin G. F. Gerritsen
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Marianne C. Verhaar
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Andrea Remuzzi
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri; via Stezzano 87 24126 Bergamo Italy
- Department of Management; Information and Production Engineering; University of Bergamo; viale Marconi 5 24044 Dalmine Italy
| | - Pedro M. Baptista
- Instituto de Investigación Sanitaria de Aragón (IIS Aragon); 50009 Zaragoza Spain
- Department of Management; Information and Production Engineering; University of Bergamo; viale Marconi 5 24044 Dalmine Italy
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas (CIBERehd); 28029 Barcelona Spain
- Fundación ARAID; 50009 Zaragoza Spain
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz; 28040 Madrid Spain. Department of Biomedical and Aerospace Engineering; Universidad Carlos III de Madrid; 28911 Madrid Spain
| | - Loredana De Bartolo
- Institute on Membrane Technology; National Research Council of Italy; ITM-CNR; Via Pietro BUCCI, Cubo 17C - 87036 Rende Italy
| | - Rosalinde Masereeuw
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Dimitrios Stamatialis
- (Bio)artificial organs; Department of Biomaterials Science and Technology; Faculty of Science and Technology; TechMed Institute; University of Twente; P.O. Box 217 7500 AE Enschede The Netherlands
| |
Collapse
|
43
|
Masakane I, Sakurai K. Current approaches to middle molecule removal: room for innovation. Nephrol Dial Transplant 2018; 33:iii12-iii21. [PMID: 30281129 PMCID: PMC6168896 DOI: 10.1093/ndt/gfy224] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/13/2018] [Indexed: 01/31/2023] Open
Abstract
Aggressive removal of middle molecules or larger low-molecular-weight proteins (LMWPs) has been a growing concern following studies on their harmful effects on the mortality and morbidity of chronic dialysis patients. To remove larger LMWPs and some protein-bound uremic toxins (PBUTs), high- and medium-cutoff (HCOs and MCOs, respectively) membranes, convective therapy and protein adsorptive membranes are available. When we use HCO or MCO membranes for convective therapy, we have to take care to avoid massive albumin leakage during a dialysis session. Convection volume is an important element to increase middle molecule removal; however, a larger convection volume has a risk of larger leakage of albumin. Predilution hemodiafiltration is a useful measurement to increase larger LMWPs without massive albumin leakage. β2-microglobulin (B2M), α1-microglobulin (A1M) and albumin leakage during a dialysis session are useful parameters for assessing middle-molecule removal. Reduction ratios of B2M >80% and of A1M >35% are favorable to improve severe dialysis-related symptoms. The efficacy of middle molecule removal should be evaluated in comparison with clinical outcomes, mortality, morbidity and the improvement of dialysis-related symptoms. Recently some dialysis-related symptoms such as sleep disturbance, skin itchiness and dialysis hypotension have been recognized as good surrogate makers for mortality. Further studies to evaluate the relationship between middle molecule or PBUTs removal and the improvement of patient symptoms should be performed in well-designed randomized controlled trials.
Collapse
Affiliation(s)
- Ikuto Masakane
- Department of Nephrology, Yabuki Hospital, Yamagata, Japan
| | - Kenji Sakurai
- Hashimoto Clinic, Dialysis Center, Sagamihara, Japan
| |
Collapse
|
44
|
García-Prieto A, Vega A, Linares T, Abad S, Macías N, Aragoncillo I, Torres E, Hernández A, Barbieri D, Luño J. Evaluation of the efficacy of a medium cut-off dialyser and comparison with other high-flux dialysers in conventional haemodialysis and online haemodiafiltration. Clin Kidney J 2018; 11:742-746. [PMID: 30288272 PMCID: PMC6165747 DOI: 10.1093/ckj/sfy004] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/19/2017] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Online haemodiafiltration (OL-HDF) has been shown to reduce all-cause mortality versus conventional haemodialysis (HD); however, it is not always available. In these situations, a novel class of membranes with a higher pore size, medium cut-off (MCO) dialysers, could be promising. The aim of this study is to evaluate the efficacy of an MCO dialyser in the removal of small and medium-size molecules and compare it with standard high-flux (HF) dialysers in HD and OL-HDF. METHODS In this crossover study, 18 prevalent HD patients were studied in three single mid-week dialysis treatments during three consecutive weeks as follows: first week with OL-HDF with a standard HF dialyser, second week with conventional HD with a standard HF dialyser and third week with conventional HD with an MCO dialyser. Reduction ratios (RRs) of different-sized molecules and albumin losses were collected for the different dialysers. RESULTS MCO HD provided a greater reduction of middle and larger middle molecules compared with standard HF HD [rate reduction (RR) β2-microglobulin 74.7% versus 69.7%, P=0.01; RR myoglobin 62.5% versus 34.3%, P=0.001; RR prolactin 60% versus 32.8%, P=0.001; RR α1-glycoprotein 2.8% versus -0.1%, P=0.01]. We found no difference in the clearance of small and larger middle molecules comparing MCO HD with OL-HDF. Albumin losses were 0.03 g/session with MCO HD and 3.1 g/session with OL-HDF (P=0.001). CONCLUSION MCO HD is superior to standard HF HD in the removal of middle and larger middle molecules and it is not inferior to OL-HDF in the clearance of small and larger middle molecules. Thus it could be an alternative in patients in which it is not possible to perform OL-HDF.
Collapse
Affiliation(s)
- Ana García-Prieto
- Nephrology Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Almudena Vega
- Nephrology Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Tania Linares
- Nephrology Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Soraya Abad
- Nephrology Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Nicolás Macías
- Nephrology Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Inés Aragoncillo
- Nephrology Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Esther Torres
- Nephrology Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Andrés Hernández
- Nephrology Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Diego Barbieri
- Nephrology Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - José Luño
- Nephrology Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| |
Collapse
|
45
|
Mair RD, Sirich TL, Plummer NS, Meyer TW. Characteristics of Colon-Derived Uremic Solutes. Clin J Am Soc Nephrol 2018; 13:1398-1404. [PMID: 30087103 PMCID: PMC6140561 DOI: 10.2215/cjn.03150318] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/13/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND OBJECTIVES Colon microbial metabolism produces solutes that are normally excreted in the urine and accumulate in the plasma when the kidneys fail. This study sought to further identify and characterize human colon-derived uremic solutes. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Colon-derived solutes normally excreted in the urine were identified by comparing urine from controls (n=17) and patients with total colectomies (n=12), using an established metabolomic platform. Colon-derived solutes that accumulate in kidney failure were then identified by comparing the plasma of the control patients with that of patients on dialysis (n=14). RESULTS Ninety-one urinary solutes were classified as colon-derived on the basis of the finding of a urine excretion rate at least four-fold higher in control patients than in patients with total colectomies. Forty-six were solutes with known chemical structure, 35 of which had not previously been identified as colon-derived. Sixty of the colon-derived solutes accumulated in the plasma of patients with ESKD to a degree greater than urea and were therefore classified as uremic. The estimated urinary clearance for 27 out of the 32 colon-derived solutes for which clearance could be calculated exceeded that of creatinine, consistent with tubular secretion. Sulfatase treatment revealed that 42 out of the 91 colon-derived solutes detected were likely conjugates. CONCLUSIONS Metabolomic analysis identified numerous colon-derived solutes that are normally excreted in human urine. Clearance by tubular secretion limits plasma levels of many colon-derived solutes.
Collapse
Affiliation(s)
- Robert D Mair
- Department of Medicine, Veterans Affairs Palo Alto Health Care System and Stanford University, Palo Alto, California
| | | | | | | |
Collapse
|
46
|
Rasmussen LD, Bøttcher M, Ivarsen P, Jørgensen HS, Nyegaard M, Buttenschøn H, Gustafsen C, Glerup S, Bøtker HE, Svensson M, Winther S. Association between circulating proprotein convertase subtilisin/kexin type 9 levels and prognosis in patients with severe chronic kidney disease. Nephrol Dial Transplant 2018; 35:632-639. [PMID: 30137516 DOI: 10.1093/ndt/gfy257] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/05/2018] [Indexed: 01/18/2023] Open
Abstract
Abstract
Background
Chronic kidney disease is a risk factor for premature development of coronary atherosclerosis and mortality. A high level of proprotein convertase subtilisin/kexin type 9 (PCSK9) is a recently recognized cardiovascular risk factor and has become the target of effective inhibitory treatment. In 167 kidney transplantation candidates, we aimed to: (i) compare levels of PCSK9 with those of healthy controls, (ii) examine the association between levels of PCSK9 and low-density lipoprotein cholesterol (LDL-c) and the degree of coronary artery disease (CAD) and (iii) evaluate if levels of PCSK9 predict major adverse cardiac events (MACE) and mortality.
Methods
Kidney transplant candidates (n = 167) underwent coronary computed tomography angiography (CCTA) and invasive coronary angiography (ICA) before transplantation. MACE and mortality data were extracted from the Western Denmark Heart Registry, a review of patient records and patient interviews. A group of 79 healthy subjects were used as controls.
Results
Mean PCSK9 levels did not differ between healthy controls and kidney transplant candidates. In patients not receiving lipid-lowering therapy, PCSK9 correlated positively with LDL-c (rho = 0.24, P < 0.05). Mean PCSK9 was similar in patients with and without obstructive CAD at both CCTA and ICA. In a multiple regression analysis, PCSK9 was associated with neither LDL-c (β=−6.45, P = 0.44) nor coronary artery calcium score (β=2.17, P = 0.84). During a follow-up of 3.7 years, PCSK9 levels were not associated with either MACE or mortality.
Conclusions
The ability of PCSK9 levels to predict cardiovascular disease and prognosis does not seem to apply to a cohort of kidney transplant candidates.
Collapse
Affiliation(s)
| | - Morten Bøttcher
- Department of Cardiology, Regional Hospital Unit West Jutland, Herning, Denmark
| | - Per Ivarsen
- Department of Nephrology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Mette Nyegaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Henriette Buttenschøn
- Department of Clinical Medicine, Aarhus University – Translational Neuropsychiatry Unit, Risskov, Denmark
| | | | - Simon Glerup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - My Svensson
- Department of Nephrology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, Søsterhjemmet, Oslo, Norway
| | - Simon Winther
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
47
|
Castillo-Rodriguez E, Fernandez-Prado R, Esteras R, Perez-Gomez MV, Gracia-Iguacel C, Fernandez-Fernandez B, Kanbay M, Tejedor A, Lazaro A, Ruiz-Ortega M, Gonzalez-Parra E, Sanz AB, Ortiz A, Sanchez-Niño MD. Impact of Altered Intestinal Microbiota on Chronic Kidney Disease Progression. Toxins (Basel) 2018; 10:toxins10070300. [PMID: 30029499 PMCID: PMC6070989 DOI: 10.3390/toxins10070300] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 12/12/2022] Open
Abstract
In chronic kidney disease (CKD), accumulation of uremic toxins is associated with an increased risk of CKD progression. Some uremic toxins result from nutrient processing by gut microbiota, yielding precursors of uremic toxins or uremic toxins themselves, such as trimethylamine N-Oxide (TMAO), p-cresyl sulphate, indoxyl sulphate and indole-3 acetic acid. Increased intake of some nutrients may modify the gut microbiota, increasing the number of bacteria that process them to yield uremic toxins. Circulating levels of nutrient-derived uremic toxins are associated to increased risk of CKD progression. This offers the opportunity for therapeutic intervention by either modifying the diet, modifying the microbiota, decreasing uremic toxin production by microbiota, increasing toxin excretion or targeting specific uremic toxins. We now review the link between nutrients, microbiota and uremic toxin with CKD progression. Specific focus will be placed on the generation specific uremic toxins with nephrotoxic potential, the decreased availability of bacteria-derived metabolites with nephroprotective potential, such as vitamin K and butyrate and the cellular and molecular mechanisms linking these toxins and protective factors to kidney diseases. This information provides a conceptual framework that allows the development of novel therapeutic approaches.
Collapse
Affiliation(s)
| | - Raul Fernandez-Prado
- Nephrology Department, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, 28040 Madrid, Spain.
| | - Raquel Esteras
- Nephrology Department, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, 28040 Madrid, Spain.
| | - Maria Vanessa Perez-Gomez
- Nephrology Department, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, 28040 Madrid, Spain.
| | - Carolina Gracia-Iguacel
- Nephrology Department, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, 28040 Madrid, Spain.
| | | | - Mehmet Kanbay
- Department of Internal Medicine, Koc University School of Medicine, Istanbul 34450, Turkey.
| | - Alberto Tejedor
- Nefrología, IIS-Gregorio Marañón, Universidad Complutense de Madrid, 28007 Madrid, Spain.
| | - Alberto Lazaro
- Nefrología, IIS-Gregorio Marañón, Universidad Complutense de Madrid, 28007 Madrid, Spain.
| | - Marta Ruiz-Ortega
- Nephrology Department, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, 28040 Madrid, Spain.
| | - Emilio Gonzalez-Parra
- Nephrology Department, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, 28040 Madrid, Spain.
| | - Ana B Sanz
- Nephrology Department, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, 28040 Madrid, Spain.
| | - Alberto Ortiz
- Nephrology Department, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, 28040 Madrid, Spain.
| | | |
Collapse
|
48
|
Chevtchik NV, Mihajlovic M, Fedecostante M, Bolhuis-Versteeg L, Sastre Toraño J, Masereeuw R, Stamatialis D. A bioartificial kidney device with polarized secretion of immune modulators. J Tissue Eng Regen Med 2018; 12:1670-1678. [DOI: 10.1002/term.2694] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 04/04/2018] [Accepted: 05/03/2018] [Indexed: 12/24/2022]
Affiliation(s)
- N. V. Chevtchik
- Bioartificial Organs, Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine; University of Twente; Enschede the Netherlands
| | - M. Mihajlovic
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Utrecht the Netherlands
| | - M. Fedecostante
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Utrecht the Netherlands
| | - L. Bolhuis-Versteeg
- Bioartificial Organs, Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine; University of Twente; Enschede the Netherlands
| | - J. Sastre Toraño
- Division of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Utrecht the Netherlands
| | - R. Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Utrecht the Netherlands
| | - D. Stamatialis
- Bioartificial Organs, Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine; University of Twente; Enschede the Netherlands
| |
Collapse
|
49
|
Uremic Toxin Clearance and Cardiovascular Toxicities. Toxins (Basel) 2018; 10:toxins10060226. [PMID: 29865226 PMCID: PMC6024759 DOI: 10.3390/toxins10060226] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 05/25/2018] [Accepted: 05/31/2018] [Indexed: 12/31/2022] Open
Abstract
Uremic solutes contribute to cardiovascular disease in renal insufficiency. In this review we describe the clearance of selected uremic solutes, which have been associated with cardiovascular disease. These solutes—indoxyl sulfate (IS), p-cresol sulfate (PCS), phenylacetylglutamine (PAG), trimethylamine-n-oxide (TMAO), and kynurenine—exemplify different mechanisms of clearance. IS and PCS are protein-bound solutes efficiently cleared by the native kidney through tubular secretion. PAG and TMAO are not protein-bound but are also cleared by the native kidney through tubular secretion, while kynurenine is not normally cleared by the kidney. Increases in the plasma levels of the normally secreted solutes IS, PCS, TMAO, and PAG in chronic kidney disease (CKD) are attributable to a reduction in their renal clearances. Levels of each of these potential toxins are even higher in patients on dialysis than in those with advanced chronic kidney disease, which can be accounted for in part by a low ratio of dialytic to native kidney clearance. The rise in plasma kynurenine in CKD and dialysis patients, by contrast, remains to be explained. Our ability to detect lower levels of the potential uremic cardiovascular toxins with renal replacement therapy may be limited by the intermittency of treatment, by increases in solute production, and by the presence of non-renal clearance. Reduction in the levels of uremic cardiovascular toxins may in the future be achieved more effectively by inhibiting their production.
Collapse
|
50
|
Abstract
Uremic solutes contribute to cardiovascular disease in renal insufficiency. In this review we describe the clearance of selected uremic solutes, which have been associated with cardiovascular disease. These solutes-indoxyl sulfate (IS), p-cresol sulfate (PCS), phenylacetylglutamine (PAG), trimethylamine-n-oxide (TMAO), and kynurenine-exemplify different mechanisms of clearance. IS and PCS are protein-bound solutes efficiently cleared by the native kidney through tubular secretion. PAG and TMAO are not protein-bound but are also cleared by the native kidney through tubular secretion, while kynurenine is not normally cleared by the kidney. Increases in the plasma levels of the normally secreted solutes IS, PCS, TMAO, and PAG in chronic kidney disease (CKD) are attributable to a reduction in their renal clearances. Levels of each of these potential toxins are even higher in patients on dialysis than in those with advanced chronic kidney disease, which can be accounted for in part by a low ratio of dialytic to native kidney clearance. The rise in plasma kynurenine in CKD and dialysis patients, by contrast, remains to be explained. Our ability to detect lower levels of the potential uremic cardiovascular toxins with renal replacement therapy may be limited by the intermittency of treatment, by increases in solute production, and by the presence of non-renal clearance. Reduction in the levels of uremic cardiovascular toxins may in the future be achieved more effectively by inhibiting their production.
Collapse
Affiliation(s)
- Robert D Mair
- The Departments of Medicine, VA Palo Alto Healthcare System, 111R, 3801 Miranda Ave., Palo Alto, CA 94304, USA.
- Division of Nephrology, Stanford University, 777 Welch Road, Suite DE, Palo Alto, CA 94304, USA.
| | - Tammy L Sirich
- The Departments of Medicine, VA Palo Alto Healthcare System, 111R, 3801 Miranda Ave., Palo Alto, CA 94304, USA.
- Division of Nephrology, Stanford University, 777 Welch Road, Suite DE, Palo Alto, CA 94304, USA.
| | - Timothy W Meyer
- The Departments of Medicine, VA Palo Alto Healthcare System, 111R, 3801 Miranda Ave., Palo Alto, CA 94304, USA.
- Division of Nephrology, Stanford University, 777 Welch Road, Suite DE, Palo Alto, CA 94304, USA.
| |
Collapse
|