1
|
Escudero-Saiz VJ, Cuadrado-Payán E, Rodriguez-Garcia M, Casals G, Rodas LM, Fontseré N, Salgado MDC, Bastida C, Rico N, Broseta JJ, Maduell F. The Choice of Anti-Inflammatory Influences the Elimination of Protein-Bound Uremic Toxins. Toxins (Basel) 2024; 16:545. [PMID: 39728803 DOI: 10.3390/toxins16120545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024] Open
Abstract
Pain is a frequent and disturbing symptom among hemodialysis patients. Protein-bound uremic toxins (PBUTs) are related to cardiovascular and overall mortality, and they are difficult to remove with current hemodialysis treatments. The PBUT displacers, such as furosemide, tryptophan, or ibuprofen, may be promising new strategies for improving their clearance. This study aims to compare ibuprofen versus other analgesic drugs in PBUT removal. A prospective study was carried out in 23 patients. Patients underwent four dialysis sessions with routine dialysis parameters, except for analgesic drugs administered (lysine acetylsalicylic acid, acetaminophen, dexketoprofen, and ibuprofen). The reduction ratios (RRs) of a wide range of molecular weight molecules were assessed, including total p-cresyl sulfate and total indoxyl-sulfate. There were no complications related to the administered drug, and pain was controlled independently of the drug. There were no differences in the RR of small-size and medium-sized molecules between all four study treatments. However, indoxyl sulfate and p-cresyl sulfate RRs when ibuprofen was administered were significantly higher than lysine acetylsalicylic acid, acetaminophen, and dexketoprofen treatments. In conclusion, patients with pain may benefit from treatment with ibuprofen instead of lysine acetylsalicylic acid, paracetamol, or dexketoprofen, since in addition to improving pain, it increases the removal of PBUTs.
Collapse
Affiliation(s)
| | - Elena Cuadrado-Payán
- Nephrology and Renal Transplantation, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - María Rodriguez-Garcia
- Biochemistry and Molecular Genetics Department-CDB, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Gregori Casals
- Biochemistry and Molecular Genetics Department-CDB, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Lida María Rodas
- Nephrology and Renal Transplantation, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Néstor Fontseré
- Nephrology and Renal Transplantation, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - María Del Carmen Salgado
- Biochemistry and Molecular Genetics Department-CDB, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Carla Bastida
- Pharmacy Department, University of Barcelona, 08036 Barcelona, Spain
| | - Nayra Rico
- Biochemistry and Molecular Genetics Department-CDB, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - José Jesús Broseta
- Nephrology and Renal Transplantation, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
- Medicine Department, University of Barcelona, 08036 Barcelona, Spain
| | - Francisco Maduell
- Nephrology and Renal Transplantation, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
- Medicine Department, University of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
2
|
Van Wesemael P, Eloot S, Raes A, Shroff R, Snauwaert E. How dialysis frequency and duration impact uremic toxin and fluid removal: a pediatric perspective. Pediatr Nephrol 2024:10.1007/s00467-024-06598-w. [PMID: 39641808 DOI: 10.1007/s00467-024-06598-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024]
Abstract
Three-weekly 4-h hemodialysis/hemodiafiltration (HD/HDF) per week has become the "standard HD/HDF" regimen in children across the globe, although increasingly criticized, since crucial determinants such as residual kidney function and patient preferences are not considered. As a consequence, several children fail to achieve adequate dialysis while on a "standard HD/HDF." In these circumstances, an extended dialysis prescription such as short daily (2-3 h/session, 5-7 days a week) or nocturnal HD/HDF (6-9 h/session, 3-5 days a week), either at home or in a dialysis center, may be considered. The purpose of this educational review is to summarize the impact of dialysis duration and frequency on uremic toxin and fluid removal. Moreover, we aim to summarize the existing literature on HD/HDF strategies with extended dialysis duration and/or increased frequency (> 12 h dialysis time per week) in pediatrics. Dialysis duration and frequency plays a crucial role in uremic toxin removal, in particular for uremic toxins with retarded transport in patients, such as phosphate, β2-microglobulin (β2m), and protein-bound uremic toxins. Also, increasing dialysis duration and/or frequency decreases the gap between plasma refilling and ultrafiltration volume), thereby decreasing the need for a high ultrafiltration rate. Observational studies in children demonstrate a beneficial effect of extended dialysis regimens (i.e., more frequent or longer duration) on blood pressure control, left ventricular hypertrophy, growth, and quality of life. PTH levels tend to decrease in the majority of studies, while hypocalcemia or suppressed PTH levels were also reported. Dietary restrictions were decreased or stopped, along with tapering of phosphate binders and potassium chelators. Extended HD/HDF regimens are beneficial in a particular group of children. Pediatric-specific international guidelines are needed to support pediatric nephrologists in determining for which children extended HD regimens are beneficial, along with increasing efforts to decrease the financial, organizational, and psychosocial barriers that are present in extended HD/HDF.
Collapse
Affiliation(s)
| | - Sunny Eloot
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium
| | - Ann Raes
- Department of Pediatric Nephrology, Ghent University Hospital, Ghent, Belgium
| | - Rukshana Shroff
- Department of Pediatric Nephrology, UCL Great Ormond Street Hospital for Children and Institute of Child Health, London, UK
| | - Evelien Snauwaert
- Department of Pediatric Nephrology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
3
|
Biedunkiewicz J, Zakrzewska A, Małgorzewicz S, Komorniczak M, Jasiulewicz K, Płonka N, Tarasewicz A, Jankowska M, Biedunkiewicz B, Dębska‐Ślizień A, Tylicki L. Comparison of uremic toxin removal between expanded hemodialysis and high volume online hemodiafiltrations in different modes. Acta Biochim Pol 2024; 71:13715. [PMID: 39679192 PMCID: PMC11637868 DOI: 10.3389/abp.2024.13715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/08/2024] [Indexed: 12/17/2024]
Abstract
Various high-efficiency hemodialysis techniques exist, including different online high- volume hemodiafiltration (HDF) modes and expanded hemodialysis (HDx) utilizing dialyzers with medium cut-off (MCO) membranes. This study aimed to evaluate the efficacy of uremic toxin removal among four modalities: (I) HDx, (II) pre-dilution HDF (PRE-HDF), (III) mixed-dilution HDF (MIX-HDF), and (IV) post-dilution HDF (POST-HDF), each applied for 1 week in a randomized order. This research was a single-center, prospective, open-label, exploratory crossover study. The reduction ratio (RR) for small molecular toxins (urea and phosphate), a middle molecular toxin (beta-2-microglobulin, β2M), a large-middle molecular toxin (Chitinase-3-like protein 1, YKL-40), and a protein- bound uremic toxin (indoxyl sulfate, IS) was evaluated during a single mid-week dialysis session. Twelve patients were included, with an average age of 52.5 ± 15.47 years and an average dialysis duration of 42.05 ± 31.04 months. The dialysis parameters, including; post-dialysis weight, session duration, dialysate composition, blood and dialysate flow; rates, dialysate temperature, and anticoagulation dosage, were maintained consistently across all modalities. No significant differences in RR for urea, phosphate, β2M, YKL-40, and IS were observed between the treatments. Although the highest IS clearance, though not statistically significant, was observed with POST-HDF and HDx, the differences were not substantial enough to favor any particular modality as the most effective.
Collapse
Affiliation(s)
- Jan Biedunkiewicz
- Department of Anesthesiology and Intensive Therapy, Medical University of Gdansk, Gdańsk, Poland
| | - Agnieszka Zakrzewska
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdansk, Gdańsk, Poland
| | - Sylwia Małgorzewicz
- Department of Clinical Nutrition, Medical University of Gdansk, Gdańsk, Poland
| | - Michał Komorniczak
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdansk, Gdańsk, Poland
| | - Katarzyna Jasiulewicz
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdansk, Gdańsk, Poland
| | - Natalia Płonka
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdansk, Gdańsk, Poland
| | - Agnieszka Tarasewicz
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdansk, Gdańsk, Poland
| | - Magdalena Jankowska
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdansk, Gdańsk, Poland
| | - Bogdan Biedunkiewicz
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdansk, Gdańsk, Poland
| | | | - Leszek Tylicki
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdansk, Gdańsk, Poland
| |
Collapse
|
4
|
Duarte Silveira MA, Malta-Santos H, Rebouças-Silva J, Teles F, Batista dos Santos Galvão E, Pinto de Souza S, Dantas Dutra FR, Dantas Gomes MM, Teixeira MB, Miranda Rebelo da Conceição LF, Nascimento CS, Vasques Nonaka CK, Cezar RS, Pena Batista PB, Berretta AA, Borges VM, da Hora Passos R. Effects of Standardized Brazilian Green Propolis Extract (EPP-AF®) on Inflammation in Haemodialysis Patients: A Clinical Trial. Int J Nephrol 2022; 2022:1035475. [PMID: 36457860 PMCID: PMC9708369 DOI: 10.1155/2022/1035475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Patients on haemodialysis (HD) present a significant inflammatory status, which has a pronounced negative impact on their outcomes. Propolis is a natural resin with anti-inflammatory and immunomodulatory properties. We assessed the safety and impact of a standardized Brazilian green propolis extract (EPP-AF®) on the inflammatory status in patients under conventional HD. METHODS Patients were assigned to receive 200 mg/day of EPP-AF® for 4 weeks followed by 4 weeks without the drug, and changes in plasma levels of interleukins (ILs), interferon gamma (IFN-γ), tumour necrosis factor-alpha (TNF-α), and high-sensitivityc-reactive protein (HsCRP) were measured. A heatmap was used to illustrate trends in data variation. RESULTS In total, 37 patients were included in the final analysis. Patients presented an exacerbated inflammatory state at baseline. During EPP-AF® use, there was a significant reduction in IFN-γ (p=0.005), IL-13 (p=0.04 2), IL-17 (p=0.039), IL-1ra (p=0.008), IL-8 (p=0.009), and TNF-α (p < 0.001) levels compared to baseline, and significant changes were found in Hs-CRP levels. The heatmap demonstrated a pattern of pronounced proinflammatory status at baseline, especially in patients with primary glomerulopathies, and a clear reduction in this pattern during the use of EPP-AF®. There was a tendency to maintain this reduction after suspension of EPP-AF®. No significant side effects were observed. CONCLUSION Patients under haemodialysis presented a pronounced inflammatory status, and EPP-AF® was demonstrated to be safe and associated with a significant and maintained reduction in proinflammatory cytokines in this population. This trial is registered with Clinicaltrials.gov NCT04072341.
Collapse
Affiliation(s)
- Marcelo Augusto Duarte Silveira
- D'Or Institute for Research and Education (IDOR), Hospital São Rafael, Nephrology Department, Avenida São Rafael 2152, São Marcos, Salvador, BA 41253190, Brazil
- UNIME Medical School, Fazenda Pitangueira, Lauro de Freitas, Bahia 42700000, Brazil
| | - Hayna Malta-Santos
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rua Waldemar Falcão 121, Candeal, Salvador, BA 40296710, Brazil
- School of Medicine, Federal University of Bahia, Rua Augusto Viana s/n, Canela, Salvador, BA 40110909, Brazil
| | - Jéssica Rebouças-Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rua Waldemar Falcão 121, Candeal, Salvador, BA 40296710, Brazil
- School of Medicine, Federal University of Bahia, Rua Augusto Viana s/n, Canela, Salvador, BA 40110909, Brazil
| | - Flávio Teles
- School of Medicine, Federal University of Alagoas, Av. Lourival de Melo Mota S/N, Tabuleiro do Martins 57072900, Maceió, Alagoas, Brazil
| | - Erica Batista dos Santos Galvão
- D'Or Institute for Research and Education (IDOR), Hospital São Rafael, Nephrology Department, Avenida São Rafael 2152, São Marcos, Salvador, BA 41253190, Brazil
| | - Sergio Pinto de Souza
- D'Or Institute for Research and Education (IDOR), Hospital São Rafael, Nephrology Department, Avenida São Rafael 2152, São Marcos, Salvador, BA 41253190, Brazil
- Escola Bahiana de Medicina e Saúde Pública-EBMSP, Av. Dom João VI 275, Brotas, Salvador, BA 40290000, Brazil
| | - Fábio Ricardo Dantas Dutra
- D'Or Institute for Research and Education (IDOR), Hospital São Rafael, Nephrology Department, Avenida São Rafael 2152, São Marcos, Salvador, BA 41253190, Brazil
| | - Marcel Miranda Dantas Gomes
- D'Or Institute for Research and Education (IDOR), Hospital São Rafael, Nephrology Department, Avenida São Rafael 2152, São Marcos, Salvador, BA 41253190, Brazil
| | - Maurício Brito Teixeira
- D'Or Institute for Research and Education (IDOR), Hospital São Rafael, Nephrology Department, Avenida São Rafael 2152, São Marcos, Salvador, BA 41253190, Brazil
- Escola Bahiana de Medicina e Saúde Pública-EBMSP, Av. Dom João VI 275, Brotas, Salvador, BA 40290000, Brazil
- Universidade do Estado da Bahia (UNEB), Rua Silveira Martin 2555, Cabula, Salvador, BA 41150000, Brazil
| | - Luis Filipe Miranda Rebelo da Conceição
- D'Or Institute for Research and Education (IDOR), Hospital São Rafael, Nephrology Department, Avenida São Rafael 2152, São Marcos, Salvador, BA 41253190, Brazil
| | - Carolina Sa Nascimento
- D'Or Institute for Research and Education (IDOR), Hospital São Rafael, Nephrology Department, Avenida São Rafael 2152, São Marcos, Salvador, BA 41253190, Brazil
| | - Carolina Kymie Vasques Nonaka
- D'Or Institute for Research and Education (IDOR), Hospital São Rafael, Nephrology Department, Avenida São Rafael 2152, São Marcos, Salvador, BA 41253190, Brazil
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rua Waldemar Falcão 121, Candeal, Salvador, BA 40296710, Brazil
| | - Rodrigo Silva Cezar
- D'Or Institute for Research and Education (IDOR), Hospital São Rafael, Nephrology Department, Avenida São Rafael 2152, São Marcos, Salvador, BA 41253190, Brazil
| | - Paulo Benigno Pena Batista
- D'Or Institute for Research and Education (IDOR), Hospital São Rafael, Nephrology Department, Avenida São Rafael 2152, São Marcos, Salvador, BA 41253190, Brazil
- UNIME Medical School, Fazenda Pitangueira, Lauro de Freitas, Bahia 42700000, Brazil
| | - Andresa Aparecida Berretta
- Research, Development and Innovation Department, Apis Flora Indl. Coml. Ltd, Rua Triunfo 945, Subse-tor Sul 3, Ribeirão Preto, SP 14020670, Brazil
| | - Valeria M. Borges
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rua Waldemar Falcão 121, Candeal, Salvador, BA 40296710, Brazil
- School of Medicine, Federal University of Bahia, Rua Augusto Viana s/n, Canela, Salvador, BA 40110909, Brazil
| | - Rogerio da Hora Passos
- D'Or Institute for Research and Education (IDOR), Hospital São Rafael, Nephrology Department, Avenida São Rafael 2152, São Marcos, Salvador, BA 41253190, Brazil
| |
Collapse
|
5
|
Yaxley J, Scott T. Dialysis and Extracorporeal Therapies for Enhanced Elimination of Toxic Ingestions and Poisoning. Ther Apher Dial 2022; 26:865-878. [PMID: 35355407 DOI: 10.1111/1744-9987.13843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/16/2022] [Accepted: 03/27/2022] [Indexed: 11/28/2022]
Abstract
Poisoning and toxic ingestions cause significant morbidity and mortality worldwide. Extracorporeal therapies such as dialysis, haemoperfusion and plasma exchange are selectively applied to patients with severe intoxications unresponsive to standard interventions and can be lifesaving. Extracorporeal therapies are a complex but fundamental aspect of the practice of nephrology. Without high-quality evidence to guide implementation, an understanding of toxicokinetics and the physiochemical principles of the enhanced elimination techniques is especially important. This review provides a comphrensive, user-friendly outline of the application of extracorporeal therapy in the poisoned patient.
Collapse
Affiliation(s)
- Julian Yaxley
- Department of Nephrology, Gold Coast University Hospital, Southport, Qld, Australia.,Department of Nephrology, Cairns Hospital, Cairns, Qld, Australia.,Department of Intensive Care Medicine, Gold Coast University Hospital, Southport, Qld, Australia
| | - Tahira Scott
- Department of Nephrology, Cairns Hospital, Cairns, Qld, Australia.,Department of Nephrology, Royal Brisbane and Women's Hospital, Herston, Qld, Australia
| |
Collapse
|
6
|
Hanoy M, Le Roy F. Prescription en hémodiafiltration : quel abord vasculaire, quel débit de pompe à sang, quel concentré acide, quel débit de substitution, quelle modalité ? Nephrol Ther 2022. [DOI: 10.1016/s1769-7255(22)00034-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
7
|
Tiranathanagul K, Khemnark N, Takkavatakarn K, Limjariyakul M, Mahatanan N, Chariyavilaskul P, Wittayalertpanya S, Susantitaphong P, Eiam-Ong S. Comparative efficacy between hemodialysis using super high-flux dialyzer with hemoperfusion and high-volume postdilution online hemodiafiltration in removing protein bound and middle molecule uremic toxins: A cross-over randomized controlled trial. Artif Organs 2022; 46:775-785. [PMID: 35028951 DOI: 10.1111/aor.14161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND Hemodialysis (HD) using super high-flux dialyzer (HD + SHF) comparably removed uremic toxins to high-volume postdilution online hemodiafiltration (olHDF). Integration of hemoperfusion (HP) to HD + SHF (HD + SHF + HP) might provide superior uremic toxin removing capability to high-volume postdilution olHDF. METHOD The present study was conducted in thrice-a-week HD patients to compare the efficacy in removing indoxyl sulfate (IS), beta-2 microglobulin (β2 M), and urea between high-volume postdilution ol-HDF and HD + SHF + HP, comprising HD + SHF as the main treatment plus HD + SHF + HP 1/week in the first 4 weeks and 1/2 weeks in the second 4 weeks. RESULTS Ten prevalent HD patients with blood flow rate (BFR) above 400 ml/min were randomized into two sequences of 8-week treatment periods of HD + SHF + HP and later high-volume postdilution olHDF or vice versa. When compared with high-volume postdilution olHDF (convective volume of 26.02 ± 1.8 L/session), HD + SHF + HP provided comparable values of percentage reduction ratio of IS (52.0 ± 11.7 vs. 56.3 ± 7.5%, p = 0.14) and β2 M (83.7 ± 4.9 vs. 84.0 ± 4.3%, p = 0.37) and slightly lower urea reduction ratio. Despite greater dialysate albumin loss (p = 0.008), there was no significant change in serum albumin level in HD + SHF + HP group. CONCLUSIONS HD + SHF + HP could not provide superior efficacy in removing uremic toxins to high-volume postdilution olHDF. The use of low BFR of 200 ml/min during the first 2 h of HD + SHF + HP session, according to the instruction of manufacturer, might impair the efficacy of the HD + SHF part in removing uremic toxins.
Collapse
Affiliation(s)
- Khajohn Tiranathanagul
- Division of Nephrology, Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society and Chulalongkorn University, Bangkok, Thailand
| | - Nutchaya Khemnark
- Division of Nephrology, Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society and Chulalongkorn University, Bangkok, Thailand
| | - Kullaya Takkavatakarn
- Division of Nephrology, Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society and Chulalongkorn University, Bangkok, Thailand
| | - Maneerut Limjariyakul
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Clinical Pharmacokinetics and Pharmacogenomics Research Unit, Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nunta Mahatanan
- Division of Nephrology, Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society and Chulalongkorn University, Bangkok, Thailand.,Department of Nursing, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Pajaree Chariyavilaskul
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Clinical Pharmacokinetics and Pharmacogenomics Research Unit, Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Paweena Susantitaphong
- Division of Nephrology, Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society and Chulalongkorn University, Bangkok, Thailand.,Research Unit for Metabolic Bone Disease in CKD patients, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Somchai Eiam-Ong
- Division of Nephrology, Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society and Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
8
|
Determinants of Hemodialysis Performance: Modeling Fluid and Solute Transport in Hollow-Fiber Dialyzers. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2021; 7:291-300. [PMID: 34926787 DOI: 10.1007/s40883-019-00135-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Hemodialysis constitutes the lifeline of patients with end stage renal disease, yet the parameters that affect hemodialyzer performance remain incompletely understood. We developed a computational model of mass transfer and solute transport in a hollow-fiber dialyzer to gain greater insight into the determinant factors. The model predicts fluid velocity, pressure, and solute concentration profiles for given geometric characteristics, membrane transport properties, and inlet conditions. We examined the impact of transport and structural parameters on uremic solute clearance by varying parameter values within the constraints of standard clinical practice. The model was validated by comparison with published experimental data. Our results suggest solute clearance can be significantly altered by changes in blood and dialysate flow rates, fiber radius and length, and net ultrafiltration rate. Our model further suggests that the main determinant of the clearance of unreactive solutes is their diffusive permeability. The clearance of protein-bound toxins is also strongly determined by blood hematocrit and plasma protein concentrations. Results from this model may serve to optimize hemodialyzer operating conditions in clinical practice to achieve better clearance of pathogenic uremic solutes.
Collapse
|
9
|
Is there an association between the plasma levels of uremic toxins from gut microbiota and anemia in patients on hemodialysis? Int Urol Nephrol 2021; 54:1271-1277. [PMID: 34561817 DOI: 10.1007/s11255-021-03001-7] [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: 05/28/2021] [Accepted: 09/19/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Anemia is one of the most frequent complications in patients with chronic kidney disease (CKD). Despite being multifactorial, the relative or absolute deficiency of erythropoietin production is the leading cause. Recent studies have shown that uremic toxins produced by the gut microbiota also may play a role in the genesis of anemia in these patients. OBJECTIVE To evaluate the possible association between uremic toxins plasma levels and anemia in patients with CKD on hemodialysis (HD). METHODS This cross-sectional study evaluated one hundred fifty-four patients (53.2% men, 51.2 ± 11.2 years, hemoglobin (Hb) levels of 11.2 ± 1.6 g/dL). Biochemical variables such as urea, creatinine, hemoglobin, hematocrit, were measured according to standard methods and uremic toxins such as indoxyl sulfate (IS), indole-3-acetic acid (IAA), p-cresyl sulfate (p-CS) plasma levels were measured by reverse-phase high-performance liquid chromatography (RP-HPLC). RESULTS The levels of uremic toxins such as IS, IAA, p-CS were increased in all patients. However, no correlation was found between uremic toxins plasma levels and anemia parameters. Only patients with Hb < 11 g/dL presented a negative correlation between hematocrit and IAA plasma levels. CONCLUSION There is no strong evidence that uremic toxins produced by the gut microbiota may be associated with anemia in patients with CKD on HD.
Collapse
|
10
|
Saar-Kovrov V, Zidek W, Orth-Alampour S, Fliser D, Jankowski V, Biessen EAL, Jankowski J. Reduction of protein-bound uraemic toxins in plasma of chronic renal failure patients: A systematic review. J Intern Med 2021; 290:499-526. [PMID: 33792983 DOI: 10.1111/joim.13248] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/07/2020] [Accepted: 12/16/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND Protein-bound uraemic toxins (PBUTs) accumulate in patients with chronic kidney disease and impose detrimental effects on the vascular system. However, a unanimous consensus on the most optimum approach for the reduction of plasma PBUTs is still lacking. METHODS In this systematic review, we aimed to identify the most efficient clinically available plasma PBUT reduction method reported in the literature between 1980 and 2020. The literature was screened for clinical studies describing approaches to reduce the plasma concentration of known uraemic toxins. There were no limits on the number of patients studied or on the duration or design of the studies. RESULTS Out of 1274 identified publications, 101 studies describing therapeutic options aiming at the reduction of PBUTs in CKD patients were included in this review. We stratified the studies by the PBUTs and the duration of the analysis into acute (data from a single procedure) and longitudinal (several treatment interventions) trials. Reduction ratio (RR) was used as the measure of plasma PBUTs lowering efficiency. For indoxyl sulphate and p-cresyl sulphate, the highest RR in the acute studies was demonstrated for fractionated plasma separation, adsorption and dialysis system. In the longitudinal trials, supplementation of haemodialysis patients with AST-120 (Kremezin®) adsorbent showed the highest RR. However, no superior method for the reduction of all types of PBUTs was identified based on the published studies. CONCLUSIONS Our study shows that there is presently no technique universally suitable for optimum reduction of all PBUTs. There is a clear need for further research in this field.
Collapse
Affiliation(s)
- V Saar-Kovrov
- From the, Institute for Molecular Cardiovascular Research IMCAR, University hospital, Aachen, Germany.,Experimental Vascular Pathology Group, Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
| | - W Zidek
- Department of Nephrology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - S Orth-Alampour
- From the, Institute for Molecular Cardiovascular Research IMCAR, University hospital, Aachen, Germany
| | - D Fliser
- From the, Institute for Molecular Cardiovascular Research IMCAR, University hospital, Aachen, Germany.,Department of Internal Medicine IV - Nephrology and Hypertension, Saarland University Medical Center, Homburg, Germany
| | - V Jankowski
- From the, Institute for Molecular Cardiovascular Research IMCAR, University hospital, Aachen, Germany
| | - E A L Biessen
- From the, Institute for Molecular Cardiovascular Research IMCAR, University hospital, Aachen, Germany.,Experimental Vascular Pathology Group, Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
| | - J Jankowski
- From the, Institute for Molecular Cardiovascular Research IMCAR, University hospital, Aachen, Germany.,Department of Nephrology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
11
|
Chronic Kidney Disease-Associated Itch (CKD-aI) in Children-A Narrative Review. Toxins (Basel) 2021; 13:toxins13070450. [PMID: 34209560 PMCID: PMC8309841 DOI: 10.3390/toxins13070450] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 06/26/2021] [Accepted: 06/28/2021] [Indexed: 01/08/2023] Open
Abstract
Chronic kidney disease (CKD) is a condition of widespread epidemiology and serious consequences affecting all organs of the organism and associated with significant mortality. The knowledge on CKD is rapidly evolving, especially concerning adults. Recently, more data is also appearing regarding CKD in children. Chronic itch (CI) is a common symptom appearing due to various underlying dermatological and systemic conditions. CI may also appear in association with CKD and is termed chronic kidney disease-associated itch (CKD-aI). CKD-aI is relatively well-described in the literature concerning adults, yet it also affects children. Unfortunately, the data on paediatric CKD-aI is particularly scarce. This narrative review aims to describe various aspects of CKD-aI with an emphasis on children, based on the available data in this population and the data extrapolated from adults. Its pathogenesis is described in details, focusing on the growing role of uraemic toxins (UTs), as well as immune dysfunction, altered opioid transmission, infectious agents, xerosis, neuropathy and dialysis-associated aspects. Moreover, epidemiological and clinical aspects are reviewed based on the few data on CKD-aI in children, whereas treatment recommendations are proposed as well, based on the literature on CKD-aI in adults and own experience in managing CI in children.
Collapse
|
12
|
ter Beek O, van Gelder M, Lokhorst C, Hazenbrink D, Lentferink B, Gerritsen K, Stamatialis D. In vitro study of dual layer mixed matrix hollow fiber membranes for outside-in filtration of human blood plasma. Acta Biomater 2021; 123:244-253. [PMID: 33450414 DOI: 10.1016/j.actbio.2020.12.063] [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: 08/06/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022]
Abstract
Hemodialysis mainly removes small water-soluble uremic toxins but cannot effectively remove middle molecules and protein-bound uremic toxins. Besides, the therapy is intermittent leading to fluctuating blood values and fluid status which adversely impacts patients' health. Prolonged hemodialysis (with adequate anticoagulation) could improve the removal of toxins and the development of portable and wearable artificial kidneys could offer more flexibility in the dialysis scheme. This would enhance patients' overall health, autonomy, mobility and flexibility, allowing patients to participate in social and economic life. However, the time that patients' blood is exposed to the dialyzer material is longer during prolonged hemodialysis, and blood clots could obstruct the fiber lumen, resulting in a decrease of the effective membrane surface area available for toxin removal. The outside-in filtration (OIF) mode, wherein blood flows through the inter-fiber space instead of through the fiber lumina, has been applied widely in blood oxygenators to prevent fiber clotting, but not in hemodialysis. In this study, we present for the first time the development of a mixed matrix membrane (MMM) for OIF of human blood plasma. This MMM combines diffusion and adsorption and consists of a polymeric membrane matrix with activated carbon (AC) particles on the inside layer, and a polymeric particle-free layer on the outer fiber layer. Our results show that in vitro MMM fibers for OIF demonstrate superior removal of the protein-bound uremic toxins, indoxyl sulfate and hippuric acid, compared to both earlier MMM fibers designed for inside-out filtration mode and commercial high-flux fibers. STATEMENT OF SIGNIFICANCE: Current hemodialysis therapy cannot effectively remove protein-bound toxins. Prolonged hemodialysis could improve toxin removal. However, during prolonged hemodialysis, blood clots could obstruct the fiber lumen, resulting in decreased effective membrane surface area available for toxin removal. We have prepared, for the first time, dual layer mixed matrix hollow fiber membranes (MMM) for outside-in filtration (OIF). The OIF mode wherein blood would flow through the inter-fiber space instead of through the fiber lumina could prevent fiber clotting. Moreover, the MMMs combine diffusion and adsorption to improve (protein-bound) toxin removal. We believe that the new design of our MMM fibers is an important contribution concerning the development of artificial kidney systems and the improvement of the health and well-being of patients with renal failure.
Collapse
|
13
|
Challenges of reducing protein-bound uremic toxin levels in chronic kidney disease and end stage renal disease. Transl Res 2021; 229:115-134. [PMID: 32891787 DOI: 10.1016/j.trsl.2020.09.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/24/2020] [Accepted: 09/02/2020] [Indexed: 12/11/2022]
Abstract
The prevalence of chronic kidney disease (CKD) in the worldwide population is currently estimated between 11% and 13%. Adequate renal clearance is compromised in these patients and the accumulation of a large number of uremic retention solutes results in an irreversible worsening of renal function which can lead to end stage renal disease (ESRD). Approximately three million ESRD patients currently receive renal replacement therapies (RRTs), such as hemodialysis, which only partially restore kidney function, as they are only efficient in removing mainly small, unbound solutes from the circulation while leaving larger and protein-bound uremic toxins (PBUTs) untouched. The accumulation of PBUTs in patients highly increases the risk of cardiovascular events and is associated with higher mortality and morbidity in CKD and ESRD. In this review, we address several strategies currently being explored toward reducing PBUT concentrations, including clinical and medical approaches, therapeutic techniques, and recent developments in RRT technology. These include preservation of renal function, limitation of colon derived PBUTs, oral sorbents, adsorbent RRT technology, and use of albumin displacers. Despite the promising results of the different approaches to promote enhanced removal of a small percentage of the more than 30 identified PBUTs, on their own, none of them provide a treatment with the required efficiency, safety and cost-effectiveness to prevent CKD-related complications and decrease mortality and morbidity in ESRD.
Collapse
|
14
|
Daneshamouz S, Eduok U, Abdelrasoul A, Shoker A. Protein-bound uremic toxins (PBUTs) in chronic kidney disease (CKD) patients: Production pathway, challenges and recent advances in renal PBUTs clearance. NANOIMPACT 2021; 21:100299. [PMID: 35559786 DOI: 10.1016/j.impact.2021.100299] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 06/15/2023]
Abstract
Uremic toxins, a group of uremic retention solutes with high concentration which their accumulation on the body makes several biological problems, have recently gained a large interest. The importance of this issue more targets patients with compromised kidney function since the presence of these toxins in their bodies contributes to serious illness and death. It is reported that around 14% of people are subjected of CKD's problems. Among different classifications of uremic toxins, protein bound uremic toxins are poorly removed from the body as they tightly bind to proteins like serum albumin. A deeper and closer understanding of methods for removing protein bound uremic toxins and their efficiency is of paramount importance. This article discussed the most critical protein bound uremic toxins from different points of view including their chemistry, binding sites, interactions, and their biological impacts. Concerning the toxicity and high concentration, p-cresyl sulfate (PCS), Indoxyl sulfate (IS), 3-Carboxy-4-methyl-5-propyl-2-furanpropionic acid (CMPF), and Indole- 3-acetic acid (IAA) was chosen to study in this article. Results offered that the functional groups of mentioned PBUTs and the way that they interact with the adsorbent play an important role in finding substances for removal of them. Furthermore, the development of nanoparticle (NPs) for promising biomedical purposes has been explored. However, there is still a need for further investigation to find biocompatible substances focusing on the removal of PBUTs. PBUTs are a unique class of uremic toxins whose renal clearance mechanisms and role in uremic pathophysiology are still unclear. This review outlines the biochemical aspects of PBUT/protein binding in a view to explaining their renal formation to elimination mechanisms; some examples are drawn from routes involving albumin-binding with indoxyl sulphate, p-cresyl sulfate, p-cresyl glucuronide and hippuric acid. We have also highlighted the kinetic behaviors during dialytic removal of PBUTs to address future concerns regarding dialytic therapy.
Collapse
Affiliation(s)
- Sana Daneshamouz
- Department of Chemical and Biological Engineering, University of Saskatchewan, 57 Campus Drive, Saskatoon, Saskatchewan S7N 5A9, Canada
| | - Ubong Eduok
- Department of Chemical and Biological Engineering, University of Saskatchewan, 57 Campus Drive, Saskatoon, Saskatchewan S7N 5A9, Canada
| | - Amira Abdelrasoul
- Department of Chemical and Biological Engineering, University of Saskatchewan, 57 Campus Drive, Saskatoon, Saskatchewan S7N 5A9, Canada; Department of Biomedical Engineering, University of Saskatchewan, 57 Campus Drive, Saskatoon, Saskatchewan S7N 5A9, Canada.
| | - Ahmed Shoker
- Nephrology Division, College of Medicine, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, SK S7N 5E5, Canada; Saskatchewan Transplant Program, St. Paul's Hospital, 1702 20th Street West Saskatoon Saskatchewan S7M 0Z9, Canada
| |
Collapse
|
15
|
|
16
|
How do Uremic Toxins Affect the Endothelium? Toxins (Basel) 2020; 12:toxins12060412. [PMID: 32575762 PMCID: PMC7354502 DOI: 10.3390/toxins12060412] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/15/2020] [Accepted: 06/19/2020] [Indexed: 12/11/2022] Open
Abstract
Uremic toxins can induce endothelial dysfunction in patients with chronic kidney disease (CKD). Indeed, the structure of the endothelial monolayer is damaged in CKD, and studies have shown that the uremic toxins contribute to the loss of cell–cell junctions, increasing permeability. Membrane proteins, such as transporters and receptors, can mediate the interaction between uremic toxins and endothelial cells. In these cells, uremic toxins induce oxidative stress and activation of signaling pathways, including the aryl hydrocarbon receptor (AhR), nuclear factor kappa B (NF-κB), and mitogen-activated protein kinase (MAPK) pathways. The activation of these pathways leads to overexpression of proinflammatory (e.g., monocyte chemoattractant protein-1, E-selectin) and prothrombotic (e.g., tissue factor) proteins. Uremic toxins also induce the formation of endothelial microparticles (EMPs), which can lead to the activation and dysfunction of other cells, and modulate the expression of microRNAs that have an important role in the regulation of cellular processes. The resulting endothelial dysfunction contributes to the pathogenesis of cardiovascular diseases, such as atherosclerosis and thrombotic events. Therefore, uremic toxins as well as the pathways they modulated may be potential targets for therapies in order to improve treatment for patients with CKD.
Collapse
|
17
|
Cobo G, Lindholm B, Stenvinkel P. Chronic inflammation in end-stage renal disease and dialysis. Nephrol Dial Transplant 2019; 33:iii35-iii40. [PMID: 30281126 PMCID: PMC6168801 DOI: 10.1093/ndt/gfy175] [Citation(s) in RCA: 256] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Indexed: 12/15/2022] Open
Abstract
Under normal conditions, inflammation is a protective and physiological response to various harmful stimuli. However, in several chronic debilitating disorders, such as chronic kidney disease, inflammation becomes maladaptive, uncontrolled and persistent. Systemic persistent inflammation has, for almost 20 years, been recognized as a major contributor to the uraemic phenotype (such as cardiovascular disease, protein energy wasting, depression, osteoporosis and frailty), and a predictor of cardiovascular and total mortality. Since inflammation is mechanistically related to several ageing processes (inflammageing), it may be a major driver of a progeric phenotype in the uraemic milieu. Inflammation is likely the consequence of a multifactorial aetiology and interacts with a number of factors that emerge when uraemic toxins accumulate. Beside interventions aiming to decrease the production of inflammatory molecules in the uraemic milieu, novel strategies to increase the removal of large middle molecules, such as expanded haemodialysis, may be an opportunity to decrease the inflammatory allostatic load associated with retention of middle molecular weight uraemic toxins.
Collapse
Affiliation(s)
- Gabriela Cobo
- Department of Education and Research, Hospital Eugenio Espejo, Quito, Ecuador
| | - Bengt Lindholm
- Division of Renal Medicine and Baxter Novum, Karolinska Institutet, Stockholm, Sweden
| | - Peter Stenvinkel
- Division of Renal Medicine and Baxter Novum, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
18
|
Blankestijn PJ, Grooteman MP, Nube MJ, Bots ML. Clinical evidence on haemodiafiltration. Nephrol Dial Transplant 2019; 33:iii53-iii58. [PMID: 30281128 PMCID: PMC6168838 DOI: 10.1093/ndt/gfy218] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 06/09/2018] [Indexed: 01/17/2023] Open
Abstract
Haemodiafiltration (HDF) combines diffusive and convective solute removal in a single treatment session. HDF provides a greater removal of higher molecular weight uraemic retention solutes than conventional high-flux haemodialysis (HD). Recently completed randomized clinical trials suggest better patient survival with online HDF. The treatment is mainly used in Europe and Japan. This review gives a brief overview of the presently available evidence of the effects of HDF on clinical end points, it speculates on possible mechanisms of a beneficial effect of HDF as compared with standard HD and ends with some perspectives for the future.
Collapse
Affiliation(s)
- Peter J Blankestijn
- Department of Nephrology & Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Muriel P Grooteman
- Department of Nephrology, Free University Amsterdam, Amsterdam, The Netherlands
| | - Menso J Nube
- Department of Nephrology, Free University Amsterdam, Amsterdam, The Netherlands
| | - Michiel L Bots
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
19
|
Snauwaert E, Van Biesen W, Raes A, Glorieux G, Vande Walle J, Roels S, Vanholder R, Askiti V, Azukaitis K, Bayazit A, Canpolat N, Fischbach M, Saoussen K, Litwin M, Obrycki L, Paglialonga F, Ranchin B, Samaille C, Schaefer F, Schmitt CP, Spasojevic B, Stefanidis CJ, Shroff R, Eloot S. Haemodiafiltration does not lower protein-bound uraemic toxin levels compared with haemodialysis in a paediatric population. Nephrol Dial Transplant 2019; 35:648-656. [DOI: 10.1093/ndt/gfz132] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/20/2019] [Indexed: 12/13/2022] Open
Abstract
Abstract
Background
Haemodiafiltration (HDF) is accepted to effectively lower plasma levels of middle molecules in the long term, while data are conflicting with respect to the additive effect of convection on lowering protein-bound uraemic toxins (PBUTs). Here we compared pre-dialysis β2-microglobulin (β2M) and PBUT levels and the percentage of protein binding (%PB) in children on post-dilution HDF versus conventional high- (hf) or low-flux (lf) haemodialysis (HD) over 12 months of treatment.
Methods
In a prospective multicentre, non-randomized parallel-arm intervention study, pre-dialysis levels of six PBUTs and β2M were measured in children (5–20 years) on post-HDF (n = 37), hf-HD (n = 42) and lf-HD (n = 18) at baseline and after 12 months. Analysis of variance was used to compare levels and %PB in post-HDF versus conventional hf-HD and lf-HD cross-sectionally at 12 months and longitudinal from baseline to 12 months.
Results
For none of the PBUTs, no difference was found in either total and free plasma levels or %PB between post-HDF versus the hf-HD and lf-HD groups. Children treated with post-HDF had lower pre-dialysis β2M levels [median 23.2 (21.5; 26.6) mg/dL] after 12 months versus children on hf-HD [P<0.01; 35.2 (29.3; 41.2) mg/dL] and children on lf-HD [P<0.001; 47.2 (34.3; 53.0) mg/dL]. While β2M levels remained steady in the hf-HD and lf-HD group, a decrease in β2M was demonstrated for children on post-HDF (P<0.01).
Conclusions
While post-HDF successfully decreased β2M, no additive effect on PBUT over 12 months of treatment was found. PBUT removal is complex and hampered by several factors. In children, these factors might be different from adults and should be explored in future research.
Collapse
Affiliation(s)
- Evelien Snauwaert
- Department of Internal Medicine & Pediatrics, Ghent University, Ghent, Belgium
| | - Wim Van Biesen
- Department of Internal Medicine & Pediatrics, Ghent University, Ghent, Belgium
| | - Ann Raes
- Department of Internal Medicine & Pediatrics, Ghent University, Ghent, Belgium
| | - Griet Glorieux
- Department of Internal Medicine & Pediatrics, Ghent University, Ghent, Belgium
| | - Johan Vande Walle
- Department of Internal Medicine & Pediatrics, Ghent University, Ghent, Belgium
| | - Sanne Roels
- Department of Data Analysis, Faculty of Psychology and Pedagogy, Ghent University, Ghent, Belgium
| | - Raymond Vanholder
- Department of Internal Medicine & Pediatrics, Ghent University, Ghent, Belgium
| | | | - Karolis Azukaitis
- Clinic of Pediatrics, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Aysun Bayazit
- Department of Pediatric Nephrology, Cukurova University, Adana, Turkey
| | - Nur Canpolat
- Department of Pediatric Nephrology, Istanbul University Cerrahpasa School of Medicine, Istanbul, Turkey
| | | | | | - Mieczyslaw Litwin
- Department of Nephrology, Kidney Transplantation and Hypertension, Children's Memorial Health Institute, Warsaw, Poland
| | - Lukasz Obrycki
- Department of Nephrology, Kidney Transplantation and Hypertension, Children's Memorial Health Institute, Warsaw, Poland
| | - Fabio Paglialonga
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Bruno Ranchin
- Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Bron, France
| | - Charlotte Samaille
- Nephrologie pédiatrique, Hôpital Jeanne de Flandre, CHU de Lille, Lille, France
| | - Franz Schaefer
- Center for Pediatrics and Adolescent Medicine, Heidelberg, Germany
| | | | - Brankica Spasojevic
- University Children's Hospital, Belgrade, Serbia
- School of Medicine, University of Belgrade, Belgrade, Serbia
| | | | - Rukshana Shroff
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Sunny Eloot
- Department of Internal Medicine & Pediatrics, Ghent University, Ghent, Belgium
| |
Collapse
|
20
|
A Bifunctional Adsorber Particle for the Removal of Hydrophobic Uremic Toxins from Whole Blood of Renal Failure Patients. Toxins (Basel) 2019; 11:toxins11070389. [PMID: 31277311 PMCID: PMC6669679 DOI: 10.3390/toxins11070389] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/29/2019] [Accepted: 07/01/2019] [Indexed: 01/10/2023] Open
Abstract
Hydrophobic uremic toxins accumulate in patients with chronic kidney disease, contributing to a highly increased cardiovascular risk. The clearance of these uremic toxins using current hemodialysis techniques is limited due to their hydrophobicity and their high binding affinity to plasma proteins. Adsorber techniques may be an appropriate alternative to increase hydrophobic uremic toxin removal. We developed an extracorporeal, whole-blood bifunctional adsorber particle consisting of a porous, activated charcoal core with a hydrophilic polyvinylpyrrolidone surface coating. The adsorption capacity was quantified using analytical chromatography after perfusion of the particles with an albumin solution or blood, each containing mixtures of hydrophobic uremic toxins. A time-dependent increase in hydrophobic uremic toxin adsorption was depicted and all toxins showed a high binding affinity to the adsorber particles. Further, the particle showed a sufficient hemocompatibility without significant effects on complement component 5a, thrombin-antithrombin III complex, or thrombocyte concentration in blood in vitro, although leukocyte counts were slightly reduced. In conclusion, the bifunctional adsorber particle with cross-linked polyvinylpyrrolidone coating showed a high adsorption capacity without adverse effects on hemocompatibility in vitro. Thus, it may be an interesting candidate for further in vivo studies with the aim to increase the efficiency of conventional dialysis techniques.
Collapse
|
21
|
Madero M, Cano KB, Campos I, Tao X, Maheshwari V, Brown J, Cornejo B, Handelman G, Thijssen S, Kotanko P. Removal of Protein-Bound Uremic Toxins during Hemodialysis Using a Binding Competitor. Clin J Am Soc Nephrol 2019; 14:394-402. [PMID: 30755453 PMCID: PMC6419294 DOI: 10.2215/cjn.05240418] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 01/03/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND OBJECTIVES Current hemodialysis techniques fail to efficiently remove the protein-bound uremic toxins p-cresyl sulfate and indoxyl sulfate due to their high degree of albumin binding. Ibuprofen, which shares the same primary albumin binding site with p-cresyl sulfate and indoxyl sulfate, can be infused during hemodialysis to displace these toxins, thereby augmenting their removal. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We infused 800 mg ibuprofen into the arterial bloodline between minutes 21 and 40 of a conventional 4-hour high-flux hemodialysis treatment. We measured arterial, venous, and dialysate outlet concentrations of indoxyl sulfate, p-cresyl sulfate, tryptophan, ibuprofen, urea, and creatinine before, during, and after the ibuprofen infusion. We report clearances of p-cresyl sulfate and indoxyl sulfate before and during ibuprofen infusion and dialysate concentrations of protein-bound uremic toxins normalized to each patient's average preinfusion concentrations. RESULTS We studied 18 patients on maintenance hemodialysis: age 36±11 years old, ten women, and mean vintage of 37±37 months. Compared with during the preinfusion period, the median (interquartile range) clearances of indoxyl sulfate and p-cresyl sulfate increased during ibuprofen infusion from 6.0 (6.5) to 20.2 (27.1) ml/min and from 4.4 (6.7) to 14.9 (27.1) ml/min (each P<0.001), respectively. Relative median (interquartile range) protein-bound uremic toxin dialysate outlet levels increased from preinfusion 1.0 (reference) to 2.4 (1.2) for indoxyl sulfate and to 2.4 (1.0) for p-cresyl sulfate (each P<0.001). Although median serum post- and predialyzer levels in the preinfusion period were similar, infusion led to a marked drop in serum postdialyzer levels for both indoxyl sulfate and p-cresyl sulfate (-1.0 and -0.3 mg/dl, respectively; each P<0.001). The removal of the nonprotein-bound solutes creatinine and urea was not increased by the ibuprofen infusion. CONCLUSIONS Infusion of ibuprofen into the arterial bloodline during hemodialysis significantly increases the dialytic removal of indoxyl sulfate and p-cresyl sulfate and thereby, leads to greater reduction in their serum levels.
Collapse
Affiliation(s)
- Magdalena Madero
- Division of Nephrology, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de Mexico, Mexico
| | - Karla B. Cano
- Division of Nephrology, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de Mexico, Mexico
| | - Israel Campos
- Renal Research Institute, Research Division, New York, New York
| | - Xia Tao
- Renal Research Institute, Research Division, New York, New York
| | | | - Jillian Brown
- Biomedical and Nutritional Sciences, University of Massachusetts, Lowell, Massachusetts; and
| | - Beatriz Cornejo
- Division of Nephrology, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de Mexico, Mexico
| | - Garry Handelman
- Biomedical and Nutritional Sciences, University of Massachusetts, Lowell, Massachusetts; and
| | | | - Peter Kotanko
- Renal Research Institute, Research Division, New York, New York
- Department of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
22
|
Pedrini LA, Zawada AM, Winter AC, Pham J, Klein G, Wolf M, Feuersenger A, Ruggiero P, Feliciani A, Barbieri C, Gauly A, Canaud B, Stuard S. Effects of high-volume online mixed-hemodiafiltration on anemia management in dialysis patients. PLoS One 2019; 14:e0212795. [PMID: 30794672 PMCID: PMC6386285 DOI: 10.1371/journal.pone.0212795] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/08/2019] [Indexed: 12/16/2022] Open
Abstract
Background Anemia is a major comorbidity of patients with end-stage renal disease and poses an enormous economic burden to health-care systems. High dose erythropoiesis-stimulating agents (ESAs) have been associated with unfavorable clinical outcomes. We explored whether mixed-dilution hemodiafiltration (Mixed-HDF), based on its innovative substitution modality, may improve anemia outcomes compared to the traditional post-dilution hemodiafiltration (Post-HDF). Methods We included 174 adult prevalent dialysis patients (87 on Mixed-HDF, 87 on Post-HDF) treated in 24 NephroCare dialysis centers between January 2010 and August 2016 into this retrospective cohort study. All patients were dialyzed three times per week and had fistula/graft as vascular access. Patients were matched at baseline and followed over a one-year period. The courses of hemoglobin levels (Hb) and monthly ESA consumption were compared between the two groups with linear mixed models. Results Mean baseline Hb was 11.9±1.3 and 11.8±1.1g/dl in patients on Mixed- and Post-HDF, respectively. While Hb remained stable in patients on Mixed-HDF, it decreased slightly in patients on Post-HDF (at month 12: 11.8±1.2 vs 11.1±1.2g/dl). This tendency was confirmed by our linear mixed model (p = 0.0514 for treatment x time interaction). Baseline median ESA consumption was 6000 [Q1:0;Q3:16000] IU/4 weeks in both groups. Throughout the observation period ESA doses tended to be lower in the Mixed-HDF group (4000 [Q1:0;Q3:16000] vs 8000 [Q1:0;Q3:20000] IU/4 weeks at month 12; p = 0.0791 for treatment x time interaction). Sensitivity analyses, adjusting for differences not covered by matching at baseline, strengthened our results (Hb: p = 0.0124; ESA: p = 0.0687). Conclusions Results of our explorative study suggest that patients on Mixed-HDF may have clinical benefits in terms of anemia management. This may also have a beneficial economic impact. Future studies are needed to confirm our hypothesis-generating results and to provide additional evidence on the potential beneficial effects of Mixed-HDF.
Collapse
Affiliation(s)
- Luciano A. Pedrini
- Department of Nephrology and Dialysis, NephroCare-ASST Bergamo-Est, Bolognini Hospital, Seriate, Italy
| | - Adam M. Zawada
- Fresenius Medical Care Deutschland GmbH, Clinical and Epidemiological Research, Bad Homburg, Germany
- * E-mail:
| | - Anke C. Winter
- Fresenius Medical Care Deutschland GmbH, Clinical and Epidemiological Research, Bad Homburg, Germany
| | - Jenny Pham
- Fresenius Medical Care Deutschland GmbH, Clinical and Epidemiological Research, Bad Homburg, Germany
| | - Gudrun Klein
- Fresenius Medical Care Deutschland GmbH, Clinical and Epidemiological Research, Bad Homburg, Germany
| | - Melanie Wolf
- Fresenius Medical Care Deutschland GmbH, Clinical and Epidemiological Research, Bad Homburg, Germany
| | - Astrid Feuersenger
- Fresenius Medical Care Deutschland GmbH, Clinical and Epidemiological Research, Bad Homburg, Germany
| | - Pio Ruggiero
- Department of Nephrology and Dialysis, NephroCare-ASST Bergamo-Est, Bolognini Hospital, Seriate, Italy
| | - Annalisa Feliciani
- Department of Nephrology and Dialysis, NephroCare-ASST Bergamo-Est, Bolognini Hospital, Seriate, Italy
| | | | - Adelheid Gauly
- Fresenius Medical Care Deutschland GmbH, Clinical and Epidemiological Research, Bad Homburg, Germany
| | - Bernard Canaud
- Fresenius Medical Care Deutschland GmbH, Medical Office EMEA, Bad Homburg, Germany
| | - Stefano Stuard
- Fresenius Medical Care Deutschland GmbH, Clinical &Therapeutical Governance, Bad Homburg, Germany
| |
Collapse
|
23
|
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: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
24
|
In silico comparison of protein-bound uremic toxin removal by hemodialysis, hemodiafiltration, membrane adsorption, and binding competition. Sci Rep 2019; 9:909. [PMID: 30696874 PMCID: PMC6351554 DOI: 10.1038/s41598-018-37195-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 12/04/2018] [Indexed: 11/19/2022] Open
Abstract
Protein-bound uremic toxins (PBUTs) are poorly removed during hemodialysis (HD) due to their low free (dialyzable) plasma concentration. We compared PBUT removal between HD, hemodiafiltration (HDF), membrane adsorption, and PBUT displacement in HD. The latter involves infusing a binding competitor pre-dialyzer, which competes with PBUTs for their albumin binding sites and increases their free fraction. We used a mathematical model of PBUT/displacer kinetics in dialysis comprising a three-compartment patient model, an arterial/venous tube segment model, and a dialyzer model. Compared to HD, improvements in removal of prototypical PBUTs indoxyl sulfate (initial concentration 100 µM, 7% free) and p-cresyl sulfate (150 µM, 5% free) were: 5.5% and 6.4%, respectively, for pre-dilution HDF with 20 L replacement fluid; 8.1% and 9.1% for post-dilution HDF 20 L; 15.6% and 18.3% for pre-dilution HDF 60 L; 19.4% and 22.2% for complete membrane adsorption; 35.0% and 41.9% for displacement with tryptophan (2000 mg in 500 mL saline); 26.7% and 32.4% for displacement with ibuprofen (800 mg in 200 mL saline). Prolonged (one-month) use of tryptophan reduces the IS and pCS time-averaged concentration by 28.1% and 29.9%, respectively, compared to conventional HD. We conclude that competitive binding can be a pragmatic approach for improving PBUT removal.
Collapse
|
25
|
Shi Y, Wang Y, Ma S, Liu T, Tian H, Zhu Q, Wang W, Li Y, Ding F. Increasing the removal of protein-bound uremic toxins by liposome-supported hemodialysis. Artif Organs 2018; 43:490-503. [PMID: 30375673 DOI: 10.1111/aor.13383] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/13/2018] [Accepted: 10/17/2018] [Indexed: 12/14/2022]
Abstract
Protein-bound uremic toxins (PBUTs) accumulate at high plasma levels and cause various deleterious effects in end-stage renal disease patients because their removal by conventional hemodialysis is severely limited by their low free-fraction levels in plasma. Here, we assessed the extent to which solute removal can be increased by adding liposomes to the dialysate. The uptake of liposomes by direct incubation in vitro showed an obvious dose-response relationship for p-cresyl sulfate (PCS) and indoxyl sulfate (IS) but not for hippuric acid (HA). The percent removal of both PCS and IS but not of HA was gradually increased with the increased concentration of liposomes in a rapid equilibrium dialysis setup. In vitro closed circulation showed that adding liposomes to the dialysate markedly increased the dialysances of PBUTs without greatly altering that of urea and creatinine. In vivo experiments in uremic rats demonstrated that adding liposomes to the dialysate resulted in higher reduction ratios (RRs) and more total solute removal (TSR) for several PBUTs compared to the conventional dialysate, which was approximately similar to the addition of bovine serum albumin to the dialysate. These findings highlight that as an adjunct to conventional hemodialysis, addition of liposomes to the dialysate could significantly improve the removal of protein-bound uremic solutes without greatly altering the removal of small, water-soluble solutes.
Collapse
Affiliation(s)
- Yuanyuan Shi
- Division of Nephrology & Unit of Critical Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yifeng Wang
- Division of Nephrology & Unit of Critical Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,The State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science & Technology, Shanghai, China
| | - Shuai Ma
- Division of Nephrology & Unit of Critical Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Tingyan Liu
- Division of Nephrology & Unit of Critical Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Huajun Tian
- Division of Nephrology & Unit of Critical Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Qiuyu Zhu
- Division of Nephrology & Unit of Critical Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wenji Wang
- Division of Nephrology & Unit of Critical Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yulin Li
- The State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science & Technology, Shanghai, China
| | - Feng Ding
- Division of Nephrology & Unit of Critical Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
26
|
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: 7.1] [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
|
27
|
Lekawanvijit S. Cardiotoxicity of Uremic Toxins: A Driver of Cardiorenal Syndrome. Toxins (Basel) 2018; 10:toxins10090352. [PMID: 30200452 PMCID: PMC6162485 DOI: 10.3390/toxins10090352] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 08/19/2018] [Accepted: 08/30/2018] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease (CVD) is highly prevalent in the setting of chronic kidney disease (CKD). Such coexistence of CVD and CKD—the so-called “cardiorenal or renocardiac syndrome”—contributes to exponentially increased risk of cardiovascular (CV) mortality. Uremic cardiomyopathy is a characteristic cardiac pathology commonly found in CKD. CKD patients are also predisposed to heart rhythm disorders especially atrial fibrillation. Traditional CV risk factors as well as known CKD-associated CV risk factors such as anemia are insufficient to explain CV complications in the CKD population. Accumulation of uremic retention solutes is a hallmark of impaired renal excretory function. Many of them have been considered inert solutes until their biological toxicity is unraveled and they become accepted as “uremic toxins”. Direct cardiotoxicity of uremic toxins has been increasingly demonstrated in recent years. This review offers a mechanistic insight into the pathological cardiac remodeling and dysfunction contributed by uremic toxins with a main focus on fibroblastic growth factor-23, an emerging toxin playing a central role in the chronic kidney disease–mineral bone disorder, and the two most investigated non-dialyzable protein-bound uremic toxins, indoxyl sulfate and p-cresyl sulfate. Potential therapeutic strategies that could address these toxins and their relevant mediated pathways since pre-dialysis stages are also discussed.
Collapse
Affiliation(s)
- Suree Lekawanvijit
- Department of Pathology, Faculty of Medicine, Chiang Mai University, 110 Intawaroros Rd, Sribhoom, Chiang Mai 50200, Thailand.
| |
Collapse
|
28
|
Esposito P, La Porta E, Grignano MA, Verzola D, Milanesi S, Ansaldo F, Gregorini M, Libetta C, Garibotto G, Rampino T. Soluble Toll-like Receptor 4: A New Player in Subclinical Inflammation and Malnutrition in Hemodialysis Patients. J Ren Nutr 2018; 28:259-264. [PMID: 29429793 DOI: 10.1053/j.jrn.2017.11.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/24/2017] [Accepted: 11/28/2017] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Toll-like receptor 4 (TLR4) promotes inflammation in hemodialysis patients (HD). A soluble form of extracellular TLR4 (sTLR4) has been recently characterized, which showed the ability to attenuate TLR4 signalling. In this study, we describe the sTLR4 profile in regular HD patients. SUBJECTS In a cross-sectional study we enrolled forty prevalent HD patients (68.2 ± 16.3 years, twenty-five males) with a median dialysis vintage of 41 months. Nineteen patients were undergoing standard bicarbonate HD (BHD) and 21 patients on-line hemodiafiltration (HDF). Ten healthy sex-matched subjects constituted the controls (C). INTERVENTION Before and after the HD session, serum was tested for sTLR4 levels by ELISA. Moreover, clinical and biochemical data were collected, including body mass index, albumin, and C-reactive protein (CRP) levels. Body composition was expressed as a 3-compartment model, providing lean tissue index and fat tissue index (FTI). MAIN OUTCOME MEASURE Describe the profile of sTLR4 in HD patients, evaluating the correlations among sTLR4 levels and the main clinical characteristics, inflammatory and nutritional parameters. RESULTS Patients with subclinical inflammation (i.e., high CRP levels without clinical symptomatology) presented higher sTLR4 levels (0.42 ± 0.25 ng/mL) with respect to both C and not inflamed HD patients (0.23 ± 0.19 ng/mL, P < .05). There was a significant direct correlation between predialysis sTLR4 and body mass index, FTI (r = 0.55), and CRP levels (r = 0.52) and inverse correlation with lean tissue index and albumin (r = -0.4). In multivariate analysis, sTLR4 resulted directly associated with FTI (P = .038). Notably, sTLR4 levels resulted higher in bicarbonate hemodialysis versus hemodiafiltration (0.37 ± 0.18 vs. 0.19 ± 0.21 ng/mL, P < .05). CONCLUSIONS sTLR4 correlates with inflammatory and nutritional parameters, presenting as a new potential player in modulating subclinical inflammation in HD patients.
Collapse
Affiliation(s)
- Pasquale Esposito
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy.
| | - Edoardo La Porta
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Maria Antonietta Grignano
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Daniela Verzola
- Department of Internal Medicine, University of Genoa and IRCCS Azienda Ospedaliera Universitaria San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Samantha Milanesi
- Department of Internal Medicine, University of Genoa and IRCCS Azienda Ospedaliera Universitaria San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Francesca Ansaldo
- Department of Internal Medicine, University of Genoa and IRCCS Azienda Ospedaliera Universitaria San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Marilena Gregorini
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Carmelo Libetta
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Giacomo Garibotto
- Department of Internal Medicine, University of Genoa and IRCCS Azienda Ospedaliera Universitaria San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Teresa Rampino
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| |
Collapse
|
29
|
Yamamoto S. Molecular mechanisms underlying uremic toxin-related systemic disorders in chronic kidney disease: focused on β 2-microglobulin-related amyloidosis and indoxyl sulfate-induced atherosclerosis-Oshima Award Address 2016. Clin Exp Nephrol 2018; 23:151-157. [PMID: 29869756 PMCID: PMC6510801 DOI: 10.1007/s10157-018-1588-9] [Citation(s) in RCA: 21] [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: 02/11/2018] [Accepted: 05/07/2018] [Indexed: 11/27/2022]
Abstract
Uremic toxins are linked to chronic kidney disease (CKD)-related systemic diseases. β2-Microglobulin (β2-m), a water-soluble, middle-sized molecule, is associated with mortality and dialysis-related amyloidosis (DRA). DRA occurs in long-term dialysis patients, with β2-m amyloid deposited mainly in osteoarticular tissues. We investigated a model of β2-m amyloid fibril extension at neutral pH in the presence of trifluoroethanol or sodium dodecyl sulfate. Using this model, some biological molecules, including glycosaminoglycans and lysophospholipids, were found to be chaperones for β2-m amyloid fibril extension. Several protein-bound solutes, such as indoxyl sulfate (IS) and p-cresyl sulfate, are independent risk factors for cardiovascular disease in CKD patients, especially those undergoing dialysis. We investigated kidney injury-induced acceleration of atherosclerosis in association with macrophage phenotypic change to a proinflammatory state as well as increased IS deposition in lesions in an animal model. IS directly induced macrophage inflammation and impaired cholesterol efflux to high-density lipoprotein (HDL) in vitro. In addition, a clinical study showed that HDL isolated from CKD patients induced proinflammatory reactions and impaired cholesterol efflux to macrophages. These findings suggest that protein-bound solutes, including IS, will induce dysfunction of both macrophages and HDL in atherosclerotic lesions. To remove uremic toxins efficiently, we demonstrated the potential efficacy of oral charcoal adsorbent and hexadecyl-immobilized cellulose beads in hemodialysis patients. These findings suggest that uremic toxins induce various CKD-related systemic disorders, and further therapeutic strategies will be needed to reduce uremic toxins enough and improve life expectancy in CKD patients.
Collapse
Affiliation(s)
- Suguru Yamamoto
- Division of Clinical Nephrology and Rheumatology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata, 951-8510, Japan.
| |
Collapse
|
30
|
Bosch-Panadero E, Mas S, Civantos E, Abaigar P, Camarero V, Ruiz-Priego A, Ortiz A, Egido J, González-Parra E. Bisphenol A is an exogenous toxin that promotes mitochondrial injury and death in tubular cells. ENVIRONMENTAL TOXICOLOGY 2018; 33:325-332. [PMID: 29214717 DOI: 10.1002/tox.22519] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/08/2017] [Accepted: 11/17/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND Uremic toxins that accumulate in chronic kidney disease (CKD) contribute to CKD complications, such as CKD progression. Bisphenol A (BPA) is a ubiquitous environmental toxin, structurally related with p-cresol, that accumulates in CKD. Our aim was to characterize the nephrotoxic potential of BPA. Specifically, we addressed BPA toxicity over energy-demanding proximal tubular cells. METHODS Cell death and oxidative stress were evaluated by flow cytometry and confocal microscopy in HK-2 human proximal tubular epithelial cells. Functional assays tested ATP, intracellular Ca2+ , mitochondrial function (tetramethylrhodamine methyl [TMRM]), oxygen consumption, Nrf2-binding, MitoSOX, and NADPH oxidase activity. Gene expression was assessed by qRT-PCR. RESULTS Following acute exposure (24 hours), proximal tubular cell viability was decreased by BPA concentrations ≥50 μM while a seven-day exposure resulted in a progressive loss of cell viability at a nanomolar range. Within 24 hours, BPA promoted mitochondrial dysfunction leading to energy depletion and increased mitochondrial and cytoplasmic oxidative stress and apoptosis in a concentration-dependent manner. An antioxidant response was observed manifested by nuclear Nrf2 translocation and increased expression of the Nrf2 target genes Heme oxygenase 1 (HO-1) and NAD(P)H dehydrogenase [quinone] 1 (NQO-1). CONCLUSIONS This study demonstrates for the first time that BPA causes mitochondrial injury, oxidative stress and apoptotic death in tubular cells. These results characterize BPA as an exogenous toxin that, similar to uremic toxins, may contribute to CKD progression.
Collapse
Affiliation(s)
- Enrique Bosch-Panadero
- Renal, Vascular and Diabetes Research Laboratory, Av Reyes Catolicos 2, Madrid, E-28040, Spain
| | - Sebastian Mas
- Renal, Vascular and Diabetes Research Laboratory, Av Reyes Catolicos 2, Madrid, E-28040, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Spain
| | - Esther Civantos
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Spain
| | - Pedro Abaigar
- Division of Nephrology, Hospital Universitario de Burgos, Burgos, Spain
| | - Vanesa Camarero
- Division of Nephrology, Hospital Universitario de Burgos, Burgos, Spain
| | - Alberto Ruiz-Priego
- Renal, Vascular and Diabetes Research Laboratory, Av Reyes Catolicos 2, Madrid, E-28040, Spain
| | - Alberto Ortiz
- Renal, Vascular and Diabetes Research Laboratory, Av Reyes Catolicos 2, Madrid, E-28040, Spain
- Division of Nephrology and Hypertension, IIS-Fundación Jimenez Diaz UAM, Madrid, Spain
- Department of Medicine, UAM, Madrid, Spain
- Kidney Research Network (REDINREN), Madrid, Spain
| | - Jesus Egido
- Renal, Vascular and Diabetes Research Laboratory, Av Reyes Catolicos 2, Madrid, E-28040, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Spain
- Division of Nephrology and Hypertension, IIS-Fundación Jimenez Diaz UAM, Madrid, Spain
- Department of Medicine, UAM, Madrid, Spain
| | - Emilio González-Parra
- Renal, Vascular and Diabetes Research Laboratory, Av Reyes Catolicos 2, Madrid, E-28040, Spain
- Division of Nephrology and Hypertension, IIS-Fundación Jimenez Diaz UAM, Madrid, Spain
- Department of Medicine, UAM, Madrid, Spain
- Kidney Research Network (REDINREN), Madrid, Spain
| |
Collapse
|
31
|
Albalate Ramón M, Martínez Miguel P, Bohorquez L, de Sequera P, Bouarich H, Pérez-García R, Rodríguez Puyol D, Barril G, Sánchez Tomero JA, Giorgi M, Ramirez Chamond MR. Asymmetric cellulose triacetate is a safe and effective alternative for online haemodiafiltration. Nefrologia 2018; 38:315-320. [PMID: 29454540 DOI: 10.1016/j.nefro.2017.11.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/04/2017] [Accepted: 11/28/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND In post-dilution haemodiafiltration only synthetic membranes have been used to date. Asymmetric cellulose triacetate (ATA™) is now available, whose characteristics are suitable for this technique. OBJECTIVES To describe the in vivo performance and behaviour of this membrane, to identify its depurative effectiveness, use in clinical practice and its biocompatibility, both acute and after one month of treatment. METHODS Observational prospective study of 23 patients who were dialysed for 4 weeks using an ATA™ membrane and who maintained their prior regimen. RESULTS A total of 287 sessions were performed and 264 complete sessions were collected. With an effective time of 243.7 (17.6) min and a mean blood flow of 371.7 (23) ml/min, an average Kt of 56.3 (5.3) l was observed, as well as a convection volume of 27.1 (4.2) l, a filtration fraction of 29.9 (3.7) %, a urea reduction ratio (RR) of 81 (5.2) %, a creatinine RR of 74.7 (4.6) %, a β2-microglobulin RR of 76.5 (4.8) % and a retinol binding protein RR of 18.6 (7.6) %. There were no technical problems or alarms. Changing the heparin dosage was not necessary. No increases in C3a or C5a concentrations or leukopenia were observed in the first 30min of the session. Neither the monocyte subpopulations nor IL-β1 or IL-6 were significantly altered after one month of treatment. CONCLUSIONS The new ATA™ membrane achieves adequate Kt and convection volume, without technical problems and with good biocompatibility and inflammatory profiles. It is therefore a valid option for post-dilution haemodiafiltration, particularly in patients allergic to synthetic membranes.
Collapse
Affiliation(s)
| | - Patricia Martínez Miguel
- Servicio de Nefrología, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, España
| | - Lourdes Bohorquez
- Laboratorio de Fisiología, Universidad de Alcalá, Alcalá de Henares, Madrid, España
| | - Patricia de Sequera
- Servicio de Nefrología, Hospital Universitario Infanta Leonor, Madrid, España
| | - Hanane Bouarich
- Servicio de Nefrología, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, España
| | - Rafael Pérez-García
- Servicio de Nefrología, Hospital Universitario Infanta Leonor, Madrid, España
| | - Diego Rodríguez Puyol
- Servicio de Nefrología, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, España
| | | | | | - Martin Giorgi
- Servicio de Nefrología, Hospital de La Princesa, Madrid, España
| | | |
Collapse
|
32
|
Vanholder R, Pletinck A, Schepers E, Glorieux G. Biochemical and Clinical Impact of Organic Uremic Retention Solutes: A Comprehensive Update. Toxins (Basel) 2018; 10:toxins10010033. [PMID: 29316724 PMCID: PMC5793120 DOI: 10.3390/toxins10010033] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 12/21/2017] [Accepted: 12/23/2017] [Indexed: 02/07/2023] Open
Abstract
In this narrative review, the biological/biochemical impact (toxicity) of a large array of known individual uremic retention solutes and groups of solutes is summarized. We classified these compounds along their physico-chemical characteristics as small water-soluble compounds or groups, protein bound compounds and middle molecules. All but one solute (glomerulopressin) affected at least one mechanism with the potential to contribute to the uremic syndrome. In general, several mechanisms were influenced for each individual solute or group of solutes, with some impacting up to 7 different biological systems of the 11 considered. The inflammatory, cardio-vascular and fibrogenic systems were those most frequently affected and they are one by one major actors in the high morbidity and mortality of CKD but also the mechanisms that have most frequently been studied. A scoring system was built with the intention to classify the reviewed compounds according to the experimental evidence of their toxicity (number of systems affected) and overall experimental and clinical evidence. Among the highest globally scoring solutes were 3 small water-soluble compounds [asymmetric dimethylarginine (ADMA); trimethylamine-N-oxide (TMAO); uric acid], 6 protein bound compounds or groups of protein bound compounds [advanced glycation end products (AGEs); p-cresyl sulfate; indoxyl sulfate; indole acetic acid; the kynurenines; phenyl acetic acid;] and 3 middle molecules [β2-microglobulin; ghrelin; parathyroid hormone). In general, more experimental data were provided for the protein bound molecules but for almost half of them clinical evidence was missing in spite of robust experimental data. The picture emanating is one of a complex disorder, where multiple factors contribute to a multisystem complication profile, so that it seems of not much use to pursue a decrease of concentration of a single compound.
Collapse
Affiliation(s)
- Raymond Vanholder
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
| | - Anneleen Pletinck
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
| | - Eva Schepers
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
| | - Griet Glorieux
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
| |
Collapse
|
33
|
Tangvoraphonkchai K, Davenport A. Enhancing dialyser clearance-from target to development. Pediatr Nephrol 2017; 32:2225-2233. [PMID: 28401301 DOI: 10.1007/s00467-017-3647-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 03/04/2017] [Accepted: 03/08/2017] [Indexed: 01/18/2023]
Abstract
Products of metabolism accumulate in kidney failure and potentially have toxic effects. Traditionally these uraemic toxins are classified as small, middle-sized and protein-bound toxins, and clearance during dialysis is affected by diffusion, convection and adsorption. As current dialysis practice effectively clears small solutes, increasing evidence supports a toxic effect for middle-sized and protein-bound toxins. Therefore, newer approaches to standard dialysis practice are required to look beyond urea clearance. Current dialysers have been developed to effectively clear small solutes and secondly to increase middle-sized toxin clearances. However, there is no ideal dialyser which can effectively clear all uraemic toxins. Advances in nanotechnology have led to improvements in manufacturing, with the production of smoother membrane surfaces and uniformity of pore size. The introduction of haemodiafiltration has led to changes in dialyser design to improve convective clearances. Both diffusional and convectional clearances can be increased by changing dialyser designs to alter blood and dialysate flows, and novel dialyser designs using microfluidics offer more efficient solute clearances. Adjusting surface hydrophilicity and charge alter adsorptive properties, and greater clearance of protein-bound toxins can be achieved by adding carbon or other absorptive monoliths into the circuit or by developing composite dialyser membranes. Other strategies to increase protein-bound toxins clearances have centred on disrupting binding and so displacing toxins from proteins. Just as the hollow fibre design replaced the flat plate dialyser, we are now entering a new era of dialyser designs aimed to increase the spectrum of uraemic toxins which can be cleared by dialysis.
Collapse
Affiliation(s)
| | - Andrew Davenport
- UCL Centre for Nephrology, Royal Free Hospital, University College London Medical School, Rowland Hill Street, London, NW3 2PF, UK.
| |
Collapse
|
34
|
Roumelioti ME, Trietley G, Nolin TD, Ng YH, Xu Z, Alaini A, Figueroa R, Unruh ML, Argyropoulos CP. Beta-2 microglobulin clearance in high-flux dialysis and convective dialysis modalities: a meta-analysis of published studies. Nephrol Dial Transplant 2017; 33:1025-1039. [DOI: 10.1093/ndt/gfx311] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 10/04/2017] [Indexed: 01/01/2023] Open
Affiliation(s)
- Maria-Eleni Roumelioti
- Nephrology Division, Department of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Gregory Trietley
- Department of Pharmacy and Therapeutics, Renal-Electrolyte Division, University of Pittsburgh Schools of Pharmacy and Medicine, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh Schools of Pharmacy and Medicine, Pittsburgh, PA, USA
| | - Thomas D Nolin
- Department of Pharmacy and Therapeutics, Renal-Electrolyte Division, University of Pittsburgh Schools of Pharmacy and Medicine, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh Schools of Pharmacy and Medicine, Pittsburgh, PA, USA
| | - Yue-Harn Ng
- Nephrology Division, Department of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Zhi Xu
- Nephrology Division, Department of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Ahmed Alaini
- Nephrology Division, Department of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Rocio Figueroa
- Nephrology Division, Department of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Mark L Unruh
- Nephrology Division, Department of Medicine, University of New Mexico, Albuquerque, NM, USA
| | | |
Collapse
|
35
|
Pavlenko D, Giasafaki D, Charalambopoulou G, van Geffen E, Gerritsen KGF, Steriotis T, Stamatialis D. Carbon Adsorbents With Dual Porosity for Efficient Removal of Uremic Toxins and Cytokines from Human Plasma. Sci Rep 2017; 7:14914. [PMID: 29097715 PMCID: PMC5668241 DOI: 10.1038/s41598-017-15116-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/20/2017] [Indexed: 11/28/2022] Open
Abstract
The number of patients with chronic kidney disease increases while the number of available donor organs stays at approximately the same level. Unavoidable accumulation of the uremic toxins and cytokines for these patients comes as the result of malfunctioning kidneys and their high levels in the blood result in high morbidity and mortality. Unfortunately, the existing methods, like hemodialysis and hemofiltration, provide only partial removal of uremic toxins and/or cytokines from patients' blood. Consequently, there is an increasing need for the development of the extracorporeal treatments which will enable removal of broad spectrum of uremic toxins that are usually removed by healthy kidneys. Therefore, in this work we developed and tested ordered mesoporous carbons as new sorbents with dual porosity (micro/meso) that provide selective and efficient removal of a broad range of uremic toxins from human plasma. The new sorbents, CMK-3 are developed by nanocasting methods and have two distinct pore domains, i.e. micropores and mesopores, therefore show high adsorption capacity towards small water soluble toxins (creatinine), protein-bound molecules (indoxyl sulfate and hippuric acid), middle molecules (β-2-microglobulin) and cytokines of different size (IL-6 and IL-8). Our results show that small amounts of CMK-3 could provide selective and complete blood purification.
Collapse
Affiliation(s)
- D Pavlenko
- (Bio)artificial organs, Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Engineering and Technical Medicine, University of Twente, P.O. Box 217, 7500 AE, Enschede, The Netherlands
| | - D Giasafaki
- National Center for Scientific Research "Demokritos", Agia Paraskevi Attikis, 15341, Athens, Greece
| | - G Charalambopoulou
- National Center for Scientific Research "Demokritos", Agia Paraskevi Attikis, 15341, Athens, Greece
| | - E van Geffen
- (Bio)artificial organs, Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Engineering and Technical Medicine, University of Twente, P.O. Box 217, 7500 AE, Enschede, The Netherlands
| | - K G F Gerritsen
- Department of Nephrology and Hypertension, University Medical Centre Utrecht, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
| | - T Steriotis
- National Center for Scientific Research "Demokritos", Agia Paraskevi Attikis, 15341, Athens, Greece
| | - D Stamatialis
- (Bio)artificial organs, Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Engineering and Technical Medicine, University of Twente, P.O. Box 217, 7500 AE, Enschede, The Netherlands.
| |
Collapse
|
36
|
Florens N, Yi D, Juillard L, Soulage CO. Using binding competitors of albumin to promote the removal of protein-bound uremic toxins in hemodialysis: Hope or pipe dream? Biochimie 2017; 144:1-8. [PMID: 28987629 DOI: 10.1016/j.biochi.2017.09.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 09/28/2017] [Indexed: 02/08/2023]
Abstract
Chronic kidney disease is associated with the accumulation of a large range of uremic retention solutes as referred to as uremic toxins. Some of these compounds belong to the group of Protein Bound Uremic Toxins (PBUT) due to their tight interactions with plasma proteins and especially serum albumin. These PBUT therefore exist in the bloodstream into two forms: a major bound (and non-diffusible) fraction and a minor free fraction. As a result, these compounds are poorly removed by most of the renal replacement therapies (such as hemodialysis) and their concentration can hardly be decreased in end-stage renal disease patients. An increase of the free fraction of PBUT could be achieved using chemical displacers that could compete with PBUT for binding to serum albumin. This review summarizes and discusses the interest of chemicals displacers as a valuable option to enhance PBUT removal in CKD patients.
Collapse
Affiliation(s)
- Nans Florens
- Univ. Lyon, CarMeN, INSERM U1060, INSA de Lyon, INRA U1397, F-69621, Villeurbanne, France; Hospices Civils de Lyon, Department of Nephrology, Hôpital E. Herriot, Lyon, F-69003, France.
| | - Dan Yi
- Univ. Lyon, CarMeN, INSERM U1060, INSA de Lyon, INRA U1397, F-69621, Villeurbanne, France
| | - Laurent Juillard
- Univ. Lyon, CarMeN, INSERM U1060, INSA de Lyon, INRA U1397, F-69621, Villeurbanne, France; Hospices Civils de Lyon, Department of Nephrology, Hôpital E. Herriot, Lyon, F-69003, France
| | - Christophe O Soulage
- Univ. Lyon, CarMeN, INSERM U1060, INSA de Lyon, INRA U1397, F-69621, Villeurbanne, France
| |
Collapse
|
37
|
Maheshwari V, Thijssen S, Tao X, Fuertinger D, Kappel F, Kotanko P. A novel mathematical model of protein-bound uremic toxin kinetics during hemodialysis. Sci Rep 2017; 7:10371. [PMID: 28871178 PMCID: PMC5583320 DOI: 10.1038/s41598-017-10981-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/10/2017] [Indexed: 11/17/2022] Open
Abstract
Protein-bound uremic toxins (PBUTs) are difficult to remove by conventional hemodialysis; a high degree of protein binding reduces the free fraction of toxins and decreases their diffusion across dialyzer membranes. Mechanistic understanding of PBUT kinetics can open new avenues to improve their dialytic removal. We developed a comprehensive model of PBUT kinetics that comprises: (1) a three-compartment patient model, (2) a dialyzer model. The model accounts for dynamic equilibrium between protein, toxin, and the protein-toxin complex. Calibrated and validated using clinical and experimental data from the literature, the model predicts key aspects of PBUT kinetics, including the free and bound concentration profiles for PBUTs and the effects of dialysate flow rate and dialyzer size on PBUT removal. Model simulations suggest that an increase in dialysate flow rate improves the reduction ratio (and removal) of strongly protein-bound toxins, namely, indoxyl sulfate and p-cresyl sulfate, while for weakly bound toxins, namely, indole-3-acetic acid and p-cresyl glucuronide, an increase in blood flow rate is advantageous. With improved dialyzer performance, removal of strongly bound PBUTs improves gradually, but marginally. The proposed model can be used for optimizing the dialysis regimen and for in silico testing of novel approaches to enhance removal of PBUTs.
Collapse
Affiliation(s)
| | | | - Xia Tao
- Renal Research Institute, New York, USA
| | | | - Franz Kappel
- Institute for Mathematics and Scientific Computing, University of Graz, Graz, Austria
| | - Peter Kotanko
- Renal Research Institute, New York, USA.,Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
38
|
Tangvoraphonkchai K, Davenport A. Increasing Haemodialytic Clearances as Residual Renal Function Declines: An Incremental Approach. Blood Purif 2017; 44:217-226. [DOI: 10.1159/000475458] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/02/2017] [Indexed: 11/19/2022]
Abstract
Many patients with chronic kidney disease start undergoing thrice-weekly haemodialysis (HD), aiming for an HD sessional dialyzer urea clearance target, irrespective of whether they have residual renal function (RRF). While increasing sessional dialyzer urea clearance above a target of 1.2 has not been shown to improve patient survival, it has been shown that the preservation of RRF improves patient self-reported outcomes and survival. Observational studies have suggested that initiating twice-weekly HD schedules leads to greater preservation of RRF. This has led to the concept of following an incremental approach to initiating HD, steadily increasing the amount of weekly dialyzer clearance as RRF decreases. Incremental dialysis practice requires the regular assessment of RRF to prevent inadequate delivery of dialysis treatment. Once RRF is lost, then the dialysis schedule and modality need to be adjusted to try to increase the middle-sized solute clearance and protein-bound toxins.
Collapse
|
39
|
Disposition and clinical implications of protein-bound uremic toxins. Clin Sci (Lond) 2017; 131:1631-1647. [DOI: 10.1042/cs20160191] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 04/10/2017] [Accepted: 04/11/2017] [Indexed: 12/11/2022]
Abstract
In patients with chronic kidney disease (CKD), adequate renal clearance is compromised, resulting in the accumulation of a plethora of uremic solutes. These uremic retention solutes, also named uremic toxins, are a heterogeneous group of organic compounds with intrinsic biological activities, many of which are too large to be filtered and/or are protein bound. The renal excretion of protein-bound toxins depends largely on active tubular secretion, which shifts the binding and allows for active secretion of the free fraction. To facilitate this process, renal proximal tubule cells are equipped with a range of transporters that co-operate in basolateral uptake and luminal excretion. Many of these transporters have been characterized as mediators of drug disposition, but have recently been recognized for their importance in the proximal renal tubular transport of uremic toxins as well. This also indicates that during uremia, drug disposition may be severely affected as a result of drug–uremic toxin interaction. In addition, CKD patients receive various drugs to treat their complications potentially resulting in drug–drug interactions (DDIs), also for drugs that are non-renally excreted. This review discusses the current knowledge on formation, disposition and removal of protein-bound uremic toxins. Furthermore, implications associated with drug treatment in kidney failure, as well as innovative renal replacement therapies targetting the protein-bound uremic toxins are being discussed. It will become clear that the complex problems associated with uremia warrant a transdisciplinary approach that unites research experts in the area of fundamental biomedical research with their colleagues in clinical nephrology.
Collapse
|
40
|
Medium cut-off membranes - closer to the natural kidney removal function. Int J Artif Organs 2017; 40:328-334. [PMID: 28574113 PMCID: PMC6159847 DOI: 10.5301/ijao.5000603] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2017] [Indexed: 01/31/2023]
|
41
|
Canaud B, Chénine L, Leray-Moraguès H, Patrier L, Rodriguez A, Gontier-Picard A, Moréna M. [Online hemodiafiltration: Practical aspects, safety and efficacy]. Nephrol Ther 2017; 13:189-201. [PMID: 28483384 DOI: 10.1016/j.nephro.2017.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Purification of high molecular uremic toxins by conventional hemodialysis is limited. It remains associated with a high morbidity and excessively high mortality. Online hemodiafiltration using a high permeability hemodiafilter, an ultrapure dialysate, and which tends to maximize substitution volumes, provides a high efficiency and low bio-incompatibility renal supplementation. Regular use of online hemodiafiltration is associated with reduced morbidity (reduction of intradialytic hypotension episodes, improved blood pressure control, reduced inflammatory profile, better anemia correction and prevention of β2-microglobulin-associated amyloidosis). Recently, several cohort studies have shown that hemodiafiltration with high substitution volume was associated with a significant reduction in mortality. Randomized studies have been conducted in Europe to confirm these facts. The high safety of online hemodiafiltration has been confirmed in clinical practice by prospective studies. Online hemodiafiltration has reached its full maturity phase and is expected to represent the new standard of renal replacement therapy.
Collapse
Affiliation(s)
- Bernard Canaud
- Néphrologie, dialyse et soins intensifs, hôpital Lapeyronie, CHRU de Montpellier, 375, avenue du Doyen-Giraud, 34925 Montpellier cedex 5, France; Association pour l'installation à domicile des épurations rénales (Aider), 746, rue de la Croix-de-Lavit, 34192 Montpellier, France; Institut de recherche et formation en dialyse (IRFD), hôpital Lapeyronie, CHRU de Montpellier, 375, avenue du Doyen-Giraud, 34295 Montpellier, France.
| | - Leïla Chénine
- Néphrologie, dialyse et soins intensifs, hôpital Lapeyronie, CHRU de Montpellier, 375, avenue du Doyen-Giraud, 34925 Montpellier cedex 5, France; Institut de recherche et formation en dialyse (IRFD), hôpital Lapeyronie, CHRU de Montpellier, 375, avenue du Doyen-Giraud, 34295 Montpellier, France
| | - Hélène Leray-Moraguès
- Néphrologie, dialyse et soins intensifs, hôpital Lapeyronie, CHRU de Montpellier, 375, avenue du Doyen-Giraud, 34925 Montpellier cedex 5, France; Association pour l'installation à domicile des épurations rénales (Aider), 746, rue de la Croix-de-Lavit, 34192 Montpellier, France; Institut de recherche et formation en dialyse (IRFD), hôpital Lapeyronie, CHRU de Montpellier, 375, avenue du Doyen-Giraud, 34295 Montpellier, France
| | - Laure Patrier
- Néphrologie, dialyse et soins intensifs, hôpital Lapeyronie, CHRU de Montpellier, 375, avenue du Doyen-Giraud, 34925 Montpellier cedex 5, France
| | - Annie Rodriguez
- Institut de recherche et formation en dialyse (IRFD), hôpital Lapeyronie, CHRU de Montpellier, 375, avenue du Doyen-Giraud, 34295 Montpellier, France
| | - A Gontier-Picard
- Néphrologie, dialyse et soins intensifs, hôpital Lapeyronie, CHRU de Montpellier, 375, avenue du Doyen-Giraud, 34925 Montpellier cedex 5, France; Institut de recherche et formation en dialyse (IRFD), hôpital Lapeyronie, CHRU de Montpellier, 375, avenue du Doyen-Giraud, 34295 Montpellier, France
| | - Marion Moréna
- Institut de recherche et formation en dialyse (IRFD), hôpital Lapeyronie, CHRU de Montpellier, 375, avenue du Doyen-Giraud, 34295 Montpellier, France
| |
Collapse
|
42
|
Panichi V, Rocchetti MT, Scatena A, Rosati A, Migliori M, Pizzarelli F, Gesualdo L. Long term variation of serum levels of uremic toxins in patients treated by post-dilution high volume on-line hemodiafiltration in comparison to standard low-flux bicarbonate dialysis: results from the REDERT study. J Nephrol 2017; 30:583-591. [PMID: 28337716 DOI: 10.1007/s40620-017-0381-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 02/06/2017] [Indexed: 11/29/2022]
Abstract
PURPOSE OF THE STUDY Little information have been provided till now regarding the effect of high volume HDF (hv-OL-HDF) in respect to standard bicarbonate dialysis (BHD) in medium-long term protein-bound toxins removal. PROCEDURES A randomised cross-over multicentre study (REDERT study) was designed to compare the effects of hv-OL-HDF and low-flux BHD on uremic toxins serum levels in 36 chronic dialysis patients followed for 13 months. Group 1 patients were treated with BHD (Treatment A) for 6 months, and afterwards, they were transferred to hv-OL-HDF for a further 6 months (Treatment B). Group 2 patients were treated with Treatment B for 6 months, and afterwards, they were transferred to Treatment A for a further 6 months. Total and free pre-dialysis indoxyl-sulfate (IS) and p-cresyl-sulfate (pCS) were determined starting a midweek dialysis session at baseline and after six months of hv-OL-HDF or BHD. IS and pCS, were simultaneously measured, by liquid chromatography/electrospray ionization-tandem mass spectrometry, Kt/v and pre and post-dialysis b-2microglobulin (b2MG) levels were measured every three months. RESULTS Kt/V was significantly increased in hv-OL-HDF (from 1.47 ± 0.24 to 1.49 ± 0.16; p < 0.01) and was reduced in BHD (from 1.51 ± 0.2 to 1.36 ± 0.21; p < 0.001). The mean infusion volume in HDF was 20.9 ± 2.1 L with a mean total convective volume of 23.8 ± 2.3 L and a significant removal of b2MG was obtained in hv-OL-HDF at month 3 and month 6. Both free and total levels of IS and pCS were significantly reduced in hv-OL-HDF at month 6 in respect to BHD. CONCLUSIONS In the present study we confirm the assumption that post-HDF is an effective technique in small and protein-bound uremic toxins removal. INTRODUCTION
Collapse
Affiliation(s)
- Vincenzo Panichi
- Nephrology and Dialysis USL Nord Ovest Toscana, Versilia Hospital, Toscana, Italy.
| | - Maria Teresa Rocchetti
- Department of Emergency and Organ Transplantation, Section of Nephrology, University of Bari 'Aldo Moro', Bari, Italy
| | - Alessia Scatena
- Nephrology and Dialysis USL Nord Ovest Toscana, Versilia Hospital, Toscana, Italy
| | - Alberto Rosati
- Nephrology and Dialysis USL Centro Toscana, Toscana, Italy
| | | | | | - Loreto Gesualdo
- Department of Emergency and Organ Transplantation, Section of Nephrology, University of Bari 'Aldo Moro', Bari, Italy
| | | |
Collapse
|
43
|
Gayrard N, Ficheux A, Duranton F, Guzman C, Szwarc I, Vetromile F, Cazevieille C, Brunet P, Servel MF, Argilés À, Le Quintrec M. Consequences of increasing convection onto patient care and protein removal in hemodialysis. PLoS One 2017; 12:e0171179. [PMID: 28166268 PMCID: PMC5293266 DOI: 10.1371/journal.pone.0171179] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/17/2017] [Indexed: 11/24/2022] Open
Abstract
INTRODUCTION Recent randomised controlled trials suggest that on-line hemodiafiltration (OL-HDF) improves survival, provided that it reaches high convective volumes. However, there is scant information on the feasibility and the consequences of modifying convection volumes in clinics. METHODS Twelve stable dialysis patients were treated with high-flux 1.8 m2 polysulphone dialyzers and 4 levels of convection flows (QUF) based on GKD-UF monitoring of the system, for 1 week each. The consequences on dialysis delivery (transmembrane pressure (TMP), number of alarms, % of achieved prescribed convection) and efficacy (mass removal of low and high molecular weight compounds) were analysed. RESULTS TMP increased exponentially with QUF (p<0.001 for N >56,000 monitoring values). Beyond 21 L/session, this resulted into frequent TMP alarms requiring nursing staff interventions (mean ± SEM: 10.3 ± 2.2 alarms per session, p<0.001 compared to lower convection volumes). Optimal convection volumes as assessed by GKD-UF-max were 20.6 ± 0.4 L/session, whilst 4 supplementary litres were obtained in the maximum situation (24.5 ± 0.6 L/session) but the proportion of sessions achieving the prescribed convection volume decreased from 94% to only 33% (p<0.001). Convection increased high molecular weight compound removal and shifted the membrane cut-off towards the higher molecular weight range. CONCLUSIONS Reaching high convection volumes as recommended by the recent RCTs (> 20L) is feasible by setting an HDF system at its optimal conditions based upon the GKD-UF monitoring. Prescribing higher convection volumes resulted in instability of the system, provoked alarms, was bothersome for the nursing staff and the patients, rarely achieved the prescribed convection volumes and increased removal of high molecular weight compounds, notably albumin.
Collapse
Affiliation(s)
- Nathalie Gayrard
- RD–Néphrologie and EA7288, University of Montpellier, Montpellier, France
| | - Alain Ficheux
- RD–Néphrologie and EA7288, University of Montpellier, Montpellier, France
| | - Flore Duranton
- RD–Néphrologie and EA7288, University of Montpellier, Montpellier, France
| | - Caroline Guzman
- RD–Néphrologie and EA7288, University of Montpellier, Montpellier, France
| | - Ilan Szwarc
- Centre de dialyse Néphrologie Dialyse St Guilhem, Sète, France
| | | | | | - Philippe Brunet
- Service de Néphrologie, Hôpital de La Conception–Université Aix-Marseille, Marseille, France
- European Uraemic Toxin Working Group of ESAO, endorsed by ERA-EDTA (EUTox), Krems, Austria
| | | | - Àngel Argilés
- RD–Néphrologie and EA7288, University of Montpellier, Montpellier, France
- Centre de dialyse Néphrologie Dialyse St Guilhem, Sète, France
- European Uraemic Toxin Working Group of ESAO, endorsed by ERA-EDTA (EUTox), Krems, Austria
| | - Moglie Le Quintrec
- Service de Néphrologie et Transplantation, Hôpital Lapeyronie CHU Montpellier, Montpellier, France
| |
Collapse
|
44
|
Krieter DH, Devine E, Körner T, Rüth M, Wanner C, Raine M, Jankowski J, Lemke HD. Haemodiafiltration at increased plasma ionic strength for improved protein-bound toxin removal. Acta Physiol (Oxf) 2017; 219:510-520. [PMID: 27259463 DOI: 10.1111/apha.12730] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 03/01/2016] [Accepted: 05/31/2016] [Indexed: 11/27/2022]
Abstract
AIM Protein-bound uraemic toxin accumulation causes uraemia-associated cardiovascular morbidity. Enhancing the plasma ionic strength releases toxins from protein binding and makes them available for removal during dialysis. This concept was implemented through high sodium concentrations ([Na+ ]) in the substituate of pre-dilution haemodiafiltration at increased plasma ionic strength (HDF-IPIS). METHODS Ex vivo HDF-IPIS with blood tested increasing [Na+ ] to demonstrate efficacy and haemocompatibility. Haemocompatibility was further assessed in sheep using two different HDF-IPIS set-ups and [Na+ ] between 350 and 600 mmol L-1 . Safety and efficacy of para-cresyl sulphate (pCS) and indoxyl sulphate (IS) removal was further investigated in a randomized clinical pilot trial comparing HDF-IPIS to HD and HDF. RESULTS Compared to [Na+ ] of 150 mmol L-1 , ex vivo HDF-IPIS at 500 mmol L-1 demonstrated up to 50% higher IS removal. Haemolysis in sheep was low even at [Na+ ] of 600 mmol L-1 (free Hb 0.016 ± 0.001 g dL-1 ). In patients, compared to HD, a [Na+ ] of 240 mmol L-1 in HDF-IPIS resulted in 40% greater reduction (48.7 ± 23.6 vs. 67.8 ± 7.9%; P = 0.013) in free IS. Compared to HD and HDF (23.0 ± 14.8 and 25.4 ± 10.5 mL min-1 ), the dialytic clearance of free IS was 31.6 ± 12.8 mL min-1 (P = 0.017) in HDF-IPIS, but [Na+ ] in arterial blood increased from 132 ± 2 to 136 ± 3 mmol L-1 (0 vs. 240 min; P < 0.001). CONCLUSION HDF-IPIS is technically and clinically feasible. More effective HDF-IPIS requires higher temporary plasma [Na+ ], but dialysate [Na+ ] has to be appropriately adapted to avoid sodium accumulation.
Collapse
Affiliation(s)
- D. H. Krieter
- Division of Nephrology; Department of Medicine; University Hospital Würzburg; Würzburg Germany
| | | | - T. Körner
- Division of Nephrology; Department of Medicine; University Hospital Würzburg; Würzburg Germany
| | - M. Rüth
- eXcorLab GmbH; Obernburg Germany
| | - C. Wanner
- Division of Nephrology; Department of Medicine; University Hospital Würzburg; Würzburg Germany
| | - M. Raine
- eXcorLab GmbH; Obernburg Germany
| | - J. Jankowski
- Institute of Molecular Cardiovascular Research; University Hospital RWTH Aachen; Aachen Germany
| | | |
Collapse
|
45
|
Gryp T, Vanholder R, Vaneechoutte M, Glorieux G. p-Cresyl Sulfate. Toxins (Basel) 2017; 9:toxins9020052. [PMID: 28146081 PMCID: PMC5331431 DOI: 10.3390/toxins9020052] [Citation(s) in RCA: 251] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/12/2017] [Accepted: 01/23/2017] [Indexed: 12/16/2022] Open
Abstract
If chronic kidney disease (CKD) is associated with an impairment of kidney function, several uremic solutes are retained. Some of these exert toxic effects, which are called uremic toxins. p-Cresyl sulfate (pCS) is a prototype protein-bound uremic toxin to which many biological and biochemical (toxic) effects have been attributed. In addition, increased levels of pCS have been associated with worsening outcomes in CKD patients. pCS finds its origin in the intestine where gut bacteria metabolize aromatic amino acids, such as tyrosine and phenylalanine, leading to phenolic end products, of which pCS is one of the components. In this review we summarize the biological effects of pCS and its metabolic origin in the intestine. It appears that, according to in vitro studies, the intestinal bacteria generating phenolic compounds mainly belong to the families Bacteroidaceae, Bifidobacteriaceae, Clostridiaceae, Enterobacteriaceae, Enterococcaceae, Eubacteriaceae, Fusobacteriaceae, Lachnospiraceae, Lactobacillaceae, Porphyromonadaceae, Staphylococcaceae, Ruminococcaceae, and Veillonellaceae. Since pCS remains difficult to remove by dialysis, the gut microbiota could be a future target to decrease pCS levels and its toxicity, even at earlier stages of CKD, aiming at slowing down the progression of the disease and decreasing the cardiovascular burden.
Collapse
Affiliation(s)
- Tessa Gryp
- Department of Internal Medicine, Nephrology Division, Ghent University Hospital, 9000 Ghent, Belgium.
- Laboratory for Bacteriology Research, Department of Clinical Chemistry, Microbiology & Immunology, Ghent University, 9000 Ghent, Belgium.
| | - Raymond Vanholder
- Department of Internal Medicine, Nephrology Division, Ghent University Hospital, 9000 Ghent, Belgium.
| | - Mario Vaneechoutte
- Laboratory for Bacteriology Research, Department of Clinical Chemistry, Microbiology & Immunology, Ghent University, 9000 Ghent, Belgium.
| | - Griet Glorieux
- Department of Internal Medicine, Nephrology Division, Ghent University Hospital, 9000 Ghent, Belgium.
| |
Collapse
|
46
|
Medium Cut-Off (MCO) Membranes Reduce Inflammation in Chronic Dialysis Patients-A Randomized Controlled Clinical Trial. PLoS One 2017; 12:e0169024. [PMID: 28085888 PMCID: PMC5234772 DOI: 10.1371/journal.pone.0169024] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 12/08/2016] [Indexed: 11/26/2022] Open
Abstract
Background To increase the removal of middle-sized uremic toxins a new membrane with enhanced permeability and selectivity, called Medium Cut-Off membrane (MCO-Ci) has been developed that at the same time ensures the retention of albumin. Because many middle-sized substances may contribute to micro-inflammation we hypothesized that the use of MCO-Ci influences the inflammatory state in hemodialysis patients. Methods The randomized crossover trial in 48 patients compared MCO-Ci dialysis to High-flux dialysis of 4 weeks duration each plus 8 weeks extension phase. Primary endpoint was the gene expression of TNF-α and IL-6 in peripheral blood mononuclear cells (PBMCs), secondary endpoints were plasma levels of specified inflammatory mediators and cytokines. Results After four weeks of MCO-Ci the expression of TNF-α mRNA (Relative quantification (RQ) from 0.92 ± 0.34 to 0.75 ± 0.31, -18.5%, p<0.001)-α and IL-6 mRNA (RQ from 0.78 ± 0.80 to 0.60 ± 0.43, -23.1%, p<0.01) was reduced to a significantly greater extent than with High-flux dialyzers (TNF mRNA-RQ: -14.3%; IL-6 mRNA-RQ: -3.5%). After retransformation of logarithmically transformed data, measurements after MCO were reduced to 82% of those after HF (95% CI 74%–91%). 4 weeks use of MCO-Ci resulted in long-lasting change in plasma levels of several cytokines and other substances with a significant decrease for sTNFR1, kappa and lambda free light chains, urea and an increase for Lp-PLA2 (PLA2G7) compared to High-flux. Albumin levels dropped significantly after 4 weeks of MCO dialysis but increased after additional 8 weeks of MCO dialysis. Twelve weeks treatment with MCO-Ci was well tolerated regarding the number of (S)AEs. In the extension period levels of CRP, TNF-α-mRNA and IL-6 mRNA remained stable in High-flux as well as in MCO-Ci. Conclusions MCO-Ci dialyzers modulate inflammation in chronic HD patients to a greater extent compared to High-flux dialyzers. Transcription of pro-inflammatory cytokines in peripheral leukocytes is markedly reduced and removal of soluble mediators is enhanced with MCO dialysis. Serum albumin concentrations stabilize after an initial drop. These results encourage further trials with longer treatment periods and clinical endpoints.
Collapse
|
47
|
Esposito P, La Porta E, Calatroni M, Grignano MA, Milanesi S, Verzola D, Battaglia Y, Gregorini M, Libetta C, Garibotto G, Rampino T. Modulation of Myostatin/Hepatocyte Growth Factor Balance by Different Hemodialysis Modalities. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7635459. [PMID: 28459069 PMCID: PMC5387812 DOI: 10.1155/2017/7635459] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/20/2017] [Indexed: 02/07/2023]
Abstract
Background. In this study we investigated the relevance of myostatin and Hepatocyte Growth Factor (HGF) in patients undergoing hemodialysis HD and the influence of different HD modalities on their levels. Methods. We performed a prospective crossover study in which HD patients were randomized to undergo 3-month treatment periods with bicarbonate hemodialysis (BHD) followed by online hemodiafiltration (HDF). Clinical data, laboratory parameters, and myostatin and HGF serum levels were collected and compared. Results. Ten patients and six controls (C) were evaluated. In any experimental condition myostatin and HGF levels were higher in HD than in C. At enrollment and after BHD there were not significant correlations, whereas at the end of the HDF treatment period myostatin and HGF were inversely correlated (r -0.65, p < 0.05), myostatin serum levels inversely correlated with transferrin (r -0.73, p < 0.05), and HGF levels that resulted positively correlated with BMI (r 0.67, p < 0.05). Moving from BHD to HDF, clinical and laboratory parameters were unchanged, as well as serum HGF, whereas myostatin levels significantly decreased (6.3 ± 4.1 versus 4.3 ± 3.1 ng/ml, p < 0.05). Conclusions. Modulation of myostatin levels and myostatin/HGF balance by the use of different HD modalities might represent a novel approach to the prevention and treatment of HD-related muscle wasting syndrome.
Collapse
Affiliation(s)
- Pasquale Esposito
- 1Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
- *Pasquale Esposito:
| | - Edoardo La Porta
- 1Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Marta Calatroni
- 1Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Maria Antonietta Grignano
- 1Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Samantha Milanesi
- 1Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Daniela Verzola
- 2Department of Internal Medicine, Istituto Nazionale per la Ricerca sul Cancro, University of Genoa and IRCCS Azienda Ospedaliera Universitaria San Martino-IST, Genoa, Italy
| | - Yuri Battaglia
- 3Nephrology and Dialysis Unit, St. Anna University Hospital, Ferrara, Italy
| | - Marilena Gregorini
- 1Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Carmelo Libetta
- 1Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Giacomo Garibotto
- 2Department of Internal Medicine, Istituto Nazionale per la Ricerca sul Cancro, University of Genoa and IRCCS Azienda Ospedaliera Universitaria San Martino-IST, Genoa, Italy
| | - Teresa Rampino
- 1Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| |
Collapse
|
48
|
Abad S, Vega A, Quiroga B, Arroyo D, Panizo N, Reque JE, López-Gómez JM. Toxinas unidas a proteínas: valor añadido en su eliminación con altos volúmenes convectivos. Nefrologia 2016; 36:637-642. [DOI: 10.1016/j.nefro.2016.05.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 04/06/2016] [Accepted: 05/04/2016] [Indexed: 12/19/2022] Open
|
49
|
Leypoldt JK, Meijers BKI. Effect of Treatment Duration and Frequency on Uremic Solute Kinetics, Clearances and Concentrations. Semin Dial 2016; 29:463-470. [PMID: 27578429 DOI: 10.1111/sdi.12531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The kinetics of uremic solute clearances are discussed based on two categories of uremic solutes, namely those that are and those that are not derived directly from nutrient intake, particularly dietary protein intake. This review highlights dialysis treatments that are more frequent and longer (high-dose hemodialysis) than conventional thrice weekly therapy. It is proposed that the dialysis dose measures based on urea as a marker uremic solute, such as Kt/V and stdKt/V, be referred to as measures of dialysis inadequacy, not dialysis adequacy. For uremic solutes derived directly from nutrient intake, it is suggested that inorganic phosphorus and protein-bound uremic solutes be considered as markers in the development of alternative measures of dialysis dose for high-dose hemodialysis prescriptions. As the current gap in understanding the detailed kinetics of protein-bound uremic solutes, it is proposed that normalization of serum phosphorus concentration with a minimum (or preferably without a) need for oral-phosphorus binders be targeted as a measure of dialysis adequacy in high-dose hemodialysis. For large uremic solutes not derived directly from nutrient intake (middle molecules), use of extracorporeal clearances for β2 -microglobulin that are higher than currently available during thrice weekly therapy is unlikely to reduce predialysis serum β2 -microglobulin concentrations. High-dose hemodialysis prescriptions will lead to reductions in predialysis serum β2 -microglobulin concentrations, but such reductions are also limited by significant residual kidney clearance. Kinetic data regarding middle molecules larger than β2 -microglobulin are scarce; additional studies on such uremic solutes are of high interest to better understand improved methods for prescribing high-dose hemodialysis prescriptions to improve patient outcomes.
Collapse
Affiliation(s)
- John K Leypoldt
- Renal Therapeutic Area and Medical Affairs, Baxter Healthcare Corporation, Deerfield, Illinois.
| | - Björn K I Meijers
- Division of Nephrology, Department of Microbiology and Immunology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
50
|
Pavlenko D, van Geffen E, van Steenbergen MJ, Glorieux G, Vanholder R, Gerritsen KGF, Stamatialis D. New low-flux mixed matrix membranes that offer superior removal of protein-bound toxins from human plasma. Sci Rep 2016; 6:34429. [PMID: 27703258 PMCID: PMC5050520 DOI: 10.1038/srep34429] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 09/07/2016] [Indexed: 11/09/2022] Open
Abstract
Hemodialysis is a widely available and well-established treatment for patients with End Stage Renal Disease (ESRD). However, although life-sustaining, patient mortality rates are very high. Several recent studies corroborated the link between dialysis patients' outcomes and elevated levels of protein-bound uremic toxins (PBUT) that are poorly removed by conventional hemodialysis. Therefore, new treatments are needed to improve their removal. Recently, our group showed that the combination of dialysis and adsorption on one membrane, the mixed matrix membrane (MMM), can effectively remove those toxins from human plasma. However, these first MMMs were rather large in diameter and their mass transport characteristics needed improvement before application in the clinical setting. Therefore, in this study we developed a new generation of MMMs that have a smaller diameter and optimized characteristics offering superior ability in removing the PBUT indoxyl sulfate (IS) and p-cresyl sulfate (pCS) in comparison to first generation MMMs (30 and 125% respectively), as well as, a commercial dialysis membrane (more than 100% better removal).
Collapse
Affiliation(s)
- Denys Pavlenko
- Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Engineering and Technical Medicine, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Esmée van Geffen
- Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Engineering and Technical Medicine, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
- Department of Nephrology and Hypertension, University Medical Centre Utrecht, P.O. Box 85500, 3508 GA Utrecht, The Netherlands
| | - Mies J. van Steenbergen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Griet Glorieux
- Ghent University Hospital, Department of Internal Medicine, Nephrology Division, 9000 Ghent, Belgium
| | - Raymond Vanholder
- Ghent University Hospital, Department of Internal Medicine, Nephrology Division, 9000 Ghent, Belgium
| | - Karin G. F. Gerritsen
- Department of Nephrology and Hypertension, University Medical Centre Utrecht, P.O. Box 85500, 3508 GA Utrecht, The Netherlands
| | - Dimitrios Stamatialis
- Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Engineering and Technical Medicine, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| |
Collapse
|