1
|
Shi L, Wang J, Wei T, Liang Z, Zhang L, Li C, Liu T, Fan W, MinZhang. Analysis of research trends and hotspots in the primary treatment of end-stage renal disease. Int Urol Nephrol 2025; 57:1513-1531. [PMID: 39589637 DOI: 10.1007/s11255-024-04290-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/11/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND There is a lack of bibliometric analysis in research regarding primary therapy for end-stage renal disease (ESRD). This study aims to analyze the literature on ESRD therapy published over the past decade to understand current conditions and study trends for future research. METHODS Data were collected from the Web of Science Core Collection (WOSCC) database. Tools like CiteSpace 6.2.R4, 6.1.R6, VOSviewer 1.6.18, and Bibliometrix R4.1.1 were used to reveal research trends and hotspots. In addition, KEGG/GO analysis examined the probable functionalities of genes implicated in ESRD therapy to guide future research. RESULTS The bibliometric analysis presented in this paper indicates that the number of publications has remained relatively stable since 2013. The level of international collaboration is notably high, with the United States serving as the dominant research hub in this field. The University of California system is the most prolific institution, and Transplantation Proceedings is the most frequently published journal in this area. Kalantar-Zadeh, Kamyar is recognized as the most published and cited author. Keywords such as "secondary hyperparathyroidism," "uremic toxins," "cyclosporine," "mycophenolate mofetil," and "biomarkers" have seen a surge in interest recently, reflecting emerging research trends. Furthermore, inflammation and stem cell research have been identified as promising new therapeutic avenues for ESRD. CONCLUSION This study identifies major areas, frontiers, and trends in research on primary treatments for ESRD, providing significant implications for future research.
Collapse
Affiliation(s)
- LuYao Shi
- Department of Nephrology, First Affiliated Hospital, Kunming Medical University, No.295, Xichang Road, Kunming, 650032, Yunnan Province, China
| | - Jian Wang
- Department of Nephrology, The Second People's Hospital of Baoshan City, Baoshan, Yunnan Province, China
| | - Tao Wei
- Library, Kunming Medical University, Kunming, Yunnan Province, China
| | - Zhang Liang
- Department of Science and Technology, Kunming Medical University, Kunming, Yunnan Province, China
| | - Le Zhang
- Institute for Integrative Genome Biology, University of California Riverside, Riverside, CA, USA
| | - ChangYan Li
- Department of Nephrology, First Affiliated Hospital, Kunming Medical University, No.295, Xichang Road, Kunming, 650032, Yunnan Province, China
| | - Tao Liu
- Organ Transplantation Center, First Affiliated Hospital, Kunming Medical University, Kunming, Yunnan Province, China
| | - WenXing Fan
- Department of Nephrology, First Affiliated Hospital, Kunming Medical University, No.295, Xichang Road, Kunming, 650032, Yunnan Province, China.
| | - MinZhang
- Clinical Pharmacy Center, First Affiliated Hospital, Kunming Medical University, No.295, Xichang Road, Kunming, 650032, Yunnan Province, China.
| |
Collapse
|
2
|
Wagner CA, Frey-Wagner I, Ortiz A, Unwin R, Liabeuf S, Suzumoto Y, Iervolino A, Stasi A, Di Marzo V, Gesualdo L, Massy ZA. The role of the intestinal microbiome in cognitive decline in patients with kidney disease. Nephrol Dial Transplant 2025; 40:ii4-ii17. [PMID: 40080091 PMCID: PMC11905753 DOI: 10.1093/ndt/gfae253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Indexed: 03/15/2025] Open
Abstract
Cognitive decline is frequently seen in patients with chronic kidney disease (CKD). The causes of cognitive decline in these patients are likely to be multifactorial, including vascular disease, uraemic toxins, blood-brain barrier leakage, and metabolic and endocrine changes. Gut dysbiosis is common in patients with CKD and contributes to the increase in uraemic toxins. However, the gut microbiome modulates local and systemic levels of several metabolites such as short-chain fatty acids or derivatives of tryptophan metabolism, neurotransmitters, endocannabinoid-like mediators, bile acids, hormones such as glucagon-like peptide 1 (GLP1) or cholecystokinin (CCK). These factors can affect gut function, immunity, autonomic nervous system activity and various aspects of brain function. Key areas include blood-brain barrier integrity, nerve myelination and survival/proliferation, appetite, metabolism and thermoregulation, mood, anxiety and depression, stress and local inflammation. Alterations in the composition of the gut microbiota and the production of biologically active metabolites in patients with CKD are well documented and are favoured by low-fiber diets, elevated urea levels, sedentary lifestyles, slow stool transit times and polypharmacy. In turn, dysbiosis can modulate brain function and cognitive processes, as discussed in this review. Thus, the gut microbiome may contribute to alterations in cognition in patients with CKD and may be a target for therapeutic interventions using diet, prebiotics and probiotics.
Collapse
Affiliation(s)
- Carsten A Wagner
- Institute of Physiology and Zurich Kidney Center, University of Zurich, Switzerland
| | | | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, RICORS2040, Madrid, Spain
| | - Robert Unwin
- Department of Renal Medicine, University College London, London, UK
| | - Sophie Liabeuf
- Pharmacoepidemiology Unit, Department of Clinical Pharmacology, Amiens-Picardie University Medical Center, Amiens, France
- MP3CV Laboratory, Jules Verne University of Picardie, Amiens, France
| | - Yoko Suzumoto
- Biogem, Molecular Biology and Genetics Research Institute, Ariano Irpino, Italy
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Naples, Italy
| | - Anna Iervolino
- Biogem, Molecular Biology and Genetics Research Institute, Ariano Irpino, Italy
- University of Campania “L. Vanvitelli”, Naples, Italy
| | - Alessandra Stasi
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J) Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, Bari, Italy
| | - Vincenzo Di Marzo
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, CRIUCPQ and INAF, Centre NUTRISS, Faculties of Medicine and Agriculture and Food Sciences, Université Laval, Québec City, Canada
- Joint International Research Unit for Chemical and Biomolecular Research on the Microbiome and its impact on Metabolic Health and Nutrition (JIRU-MicroMeNu) between Université Laval Québec, Canada and Consiglio Nazionale delle Ricerche, Institute of Biomolecular Chemistry, Pozzuoli, Italy
| | - Loreto Gesualdo
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J) Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, Bari, Italy
| | - Ziad A Massy
- INSERM Unit 1018, Team 5, CESP, Hôpital Paul Brousse, Paris-Saclay University and Versailles Saint-Quentin-en-Yvelines University (UVSQ), Villejuif, France
- Association pour l'Utilisation du Rein Artificiel dans la région parisienne (AURA) Paris, France and Ambroise Paré University Hospital, APHP, Department of Nephrology Boulogne-Billancourt, Paris, France
| |
Collapse
|
3
|
Malaweera A, Huang L, McMahon L. Benefits and Pitfalls of Uraemic Toxin Measurement in Peritoneal Dialysis. J Clin Med 2025; 14:1395. [PMID: 40004925 PMCID: PMC11857055 DOI: 10.3390/jcm14041395] [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: 01/22/2025] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
Chronic kidney disease is a global health burden with a rising incidence and prevalence in developed and developing nations. Once established, it results in a progressive accumulation of a myriad of uraemic toxins. Peritoneal dialysis (PD) uses the body's peritoneal membrane to remove these toxins across a semipermeable membrane to restore and maintain homeostasis. Traditionally, dialysis adequacy has been measured through clearance of urea and creatinine. However, numerous studies have shown marginal links comparing the clearance of urea and creatinine with clinical outcomes reflected in the recent changes to the ISPD guidelines on dialysis adequacy. Instead, attention has focused on protein-bound uraemic toxins (PBTs). Produced by gut bacteria, these molecules are highly protein-bound and poorly removed by either dialysis or absorptive agents. Elevated concentrations of molecules such as p-cresyl sulfate and indoxyl sulfate have been associated with abnormal cellular function and poor patient outcomes. However, widespread use of these measures to determine dialysis adequacy has been limited by the need for specialized techniques required for measurement. Altering the gut microbiome to reduce generation of PBTs through increased dietary fiber might be an alternate approach to better patient outcomes, with some initial positive reports. This report explores advantages and limitations of measuring uraemic toxins in PD, now and in the foreseeable future.
Collapse
Affiliation(s)
- Aruni Malaweera
- Department of Renal Medicine, Eastern Health, 5, Arnold Street, Box Hill, Melbourne, VIC 3128, Australia; (L.H.); (L.M.)
| | | | | |
Collapse
|
4
|
Delrue C, Speeckaert MM. Decoding Kidney Pathophysiology: Omics-Driven Approaches in Precision Medicine. J Pers Med 2024; 14:1157. [PMID: 39728069 DOI: 10.3390/jpm14121157] [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: 11/01/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
Chronic kidney disease (CKD) is a major worldwide health concern because of its progressive nature and complex biology. Traditional diagnostic and therapeutic approaches usually fail to account for disease heterogeneity, resulting in low efficacy. Precision medicine offers a novel approach to studying kidney disease by combining omics technologies such as genomics, transcriptomics, proteomics, metabolomics, and epigenomics. By identifying discrete disease subtypes, molecular biomarkers, and therapeutic targets, these technologies pave the way for personalized treatment approaches. Multi-omics integration has enhanced our understanding of CKD by revealing intricate molecular linkages and pathways that contribute to treatment resistance and disease progression. While pharmacogenomics offers insights into expected responses to personalized treatments, single-cell and spatial transcriptomics can be utilized to investigate biological heterogeneity. Despite significant development, challenges persist, including data integration concerns, high costs, and ethical quandaries. Standardized data protocols, collaborative data-sharing frameworks, and advanced computational tools such as machine learning and causal inference models are required to address these challenges. With the advancement of omics technology, nephrology may benefit from improved diagnostic accuracy, risk assessment, and personalized care. By overcoming these barriers, precision medicine has the potential to develop novel techniques for improving patient outcomes in kidney disease treatment.
Collapse
Affiliation(s)
- Charlotte Delrue
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Marijn M Speeckaert
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium
- Research Foundation-Flanders (FWO), 1000 Brussels, Belgium
| |
Collapse
|
5
|
Coll E, Cigarran S, Portolés J, Cases A. Gut Dysbiosis and Its Role in the Anemia of Chronic Kidney Disease. Toxins (Basel) 2024; 16:495. [PMID: 39591250 PMCID: PMC11598790 DOI: 10.3390/toxins16110495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
The gut dysbiosis present in chronic kidney disease (CKD) has been associated with anemia. Factors such as the accumulation of gut-derived uremic toxins, increased gut barrier permeability-induced inflammation, and a reduced intestinal production of short-chain fatty acids (SCFAs), all associated with changes in the intestinal microbiota composition in CKD, may lead to the development or worsening of anemia in renal patients. Understanding and addressing these mechanisms related to gut dysbiosis in CKD patients can help to delay the development of anemia and improve its control in this population. One approach is to avoid or reduce the use of drugs linked to gut dysbiosis in CKD, such as phosphate binders, oral iron supplementation, antibiotics, and others, unless they are indispensable. Another approach involves introducing dietary changes that promote a healthier microbiota and/or using prebiotics, probiotics, or symbiotics to improve gut dysbiosis in this setting. These measures can increase the presence of SCFA-producing saccharolytic bacteria and reduce proteolytic bacteria, thereby lowering the production of gut-derived uremic toxins and inflammation. By ameliorating CKD-related gut dysbiosis, these strategies can also improve the control of renal anemia and enhance the response to erythropoiesis-stimulating agents (ESAs) in ESA-resistant patients. In this review, we have explored the relationship between gut dysbiosis in CKD and renal anemia and propose feasible solutions, both those already known and potential future treatments.
Collapse
Affiliation(s)
- Elisabet Coll
- Servei de Nefrologia, Fundacio Puigvert, 08025 Barcelona, Spain
- Anemia Working Group of the Spanish Society of Nephrology, 39008 Santander, Spain; (J.P.); (A.C.)
| | | | - Jose Portolés
- Anemia Working Group of the Spanish Society of Nephrology, 39008 Santander, Spain; (J.P.); (A.C.)
- Ressearch Net RICORS 2030 Instituto de Salud Carlos III ISCIII, 28029 Madrid, Spain
- Nephrology Department, Hospital Universitario Puerta de Hierro Majadahonda, 28222 Madrid, Spain
- Medicine Department, Facultad de Medicina, Research Institute Puerta de Hierro Segovia de Arana (IDIPHISA), Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Aleix Cases
- Anemia Working Group of the Spanish Society of Nephrology, 39008 Santander, Spain; (J.P.); (A.C.)
- Nephrology Unit, Hospital Clinic, 08036 Barcelona, Spain
| |
Collapse
|
6
|
Wang Q, Munteanu B, Marker A, Luo Y, Holz C, Kane JL, Kuntzweiler T, Poulton EJ, Sedic M, Jayyosi Z, Riedel J, Fretland J. The Simultaneous Inhibition of Solute Carrier Family 6 Member 19 and Breast Cancer Resistance Protein Transporters Leads to an Increase of Indoxyl Sulfate (a Uremic Toxin) in Plasma and Kidney. Drug Metab Dispos 2024; 52:1288-1296. [PMID: 39251367 DOI: 10.1124/dmd.124.001813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/05/2024] [Accepted: 08/26/2024] [Indexed: 09/11/2024] Open
Abstract
Solute carrier family 6 member 19 (SLC6A19) inhibitors are being studied as therapeutic agents for phenylketonuria. In this work, a potent SLC6A19 inhibitor (RA836) elevated rat kidney uremic toxin indoxyl sulfate (IDS) levels by intensity (arbitrary unit) of 13.7 ± 7.7 compared with vehicle 0.3 ± 0.1 (P = 0.01) as determined by tissue mass spectrometry imaging analysis. We hypothesized that increased plasma and kidney levels of IDS could be caused by the simultaneous inhibition of both Slc6a19 and a kidney IDS transporter responsible for excretion of IDS into urine. To test this, we first confirmed the formation of IDS through tryptophan metabolism by feeding rats a Trp-free diet. Inhibiting Slc6a19 with RA836 led to increased IDS in these rats. Next, RA836 and its key metabolites were evaluated in vitro for inhibiting kidney transporters such as organic anion transporter (OAT)1, OAT3, and breast cancer resistance protein (BCRP). RA836 inhibits BCRP with an IC50 of 0.045 μM but shows no significant inhibition of OAT1 or OAT3. Finally, RA836 analogs with either potent or no inhibition of SLC6A19 and/or BCRP were synthesized and administered to rats fed a normal diet. Plasma and kidney samples were collected to quantify IDS using liquid chromatography-mass spectrometry. Neither a SLC6A19 inactive but potent BCRP inhibitor nor a SLC6A19 active but weak BCRP inhibitor raised IDS levels, whereas compounds inhibiting both transporters caused IDS accumulation in rat plasma and kidney, supporting the hypothesis that rat Bcrp contributes to the excretion of IDS. In summary, we identified that inhibiting Slc6a19 increases IDS formation, while simultaneously inhibiting Bcrp results in IDS accumulation in the kidney and plasma. SIGNIFICANCE STATEMENT: This is the first publication to decipher the mechanism for accumulation of indoxyl sulfate (IDS) (a uremic toxin) in rats via inhibition of both Slc6a19 and Bcrp. Specifically, inhibition of Slc6a19 in the gastrointestinal track increases IDS formation, and inhibition of Bcrp in the kidney blocks IDS excretion. Therefore, we should avoid inhibiting both solute carrier family 6 member 19 and breast cancer resistance protein simultaneously in humans to prevent accumulation of IDS, a known risk factor for cardiovascular disease, psychic anxiety, and mortality in chronic kidney disease patients.
Collapse
Affiliation(s)
- Qingping Wang
- Department of Drug Metabolism and Pharmacokinetics (Q.W., Y.L., J.F.), Integrated Drug Discovery (J.L.K., T.K.), and Preclinical Safety Research (E.-J.P., M.S., Z.J.), Sanofi, Cambridge, Massachusetts; Department of Drug Metabolism and Pharmacokinetics, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany (B.M., A.M., C.H., J.R.)
| | - Bogdan Munteanu
- Department of Drug Metabolism and Pharmacokinetics (Q.W., Y.L., J.F.), Integrated Drug Discovery (J.L.K., T.K.), and Preclinical Safety Research (E.-J.P., M.S., Z.J.), Sanofi, Cambridge, Massachusetts; Department of Drug Metabolism and Pharmacokinetics, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany (B.M., A.M., C.H., J.R.)
| | - Alexander Marker
- Department of Drug Metabolism and Pharmacokinetics (Q.W., Y.L., J.F.), Integrated Drug Discovery (J.L.K., T.K.), and Preclinical Safety Research (E.-J.P., M.S., Z.J.), Sanofi, Cambridge, Massachusetts; Department of Drug Metabolism and Pharmacokinetics, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany (B.M., A.M., C.H., J.R.)
| | - Yongyi Luo
- Department of Drug Metabolism and Pharmacokinetics (Q.W., Y.L., J.F.), Integrated Drug Discovery (J.L.K., T.K.), and Preclinical Safety Research (E.-J.P., M.S., Z.J.), Sanofi, Cambridge, Massachusetts; Department of Drug Metabolism and Pharmacokinetics, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany (B.M., A.M., C.H., J.R.)
| | - Constanze Holz
- Department of Drug Metabolism and Pharmacokinetics (Q.W., Y.L., J.F.), Integrated Drug Discovery (J.L.K., T.K.), and Preclinical Safety Research (E.-J.P., M.S., Z.J.), Sanofi, Cambridge, Massachusetts; Department of Drug Metabolism and Pharmacokinetics, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany (B.M., A.M., C.H., J.R.)
| | - John L Kane
- Department of Drug Metabolism and Pharmacokinetics (Q.W., Y.L., J.F.), Integrated Drug Discovery (J.L.K., T.K.), and Preclinical Safety Research (E.-J.P., M.S., Z.J.), Sanofi, Cambridge, Massachusetts; Department of Drug Metabolism and Pharmacokinetics, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany (B.M., A.M., C.H., J.R.)
| | - Theresa Kuntzweiler
- Department of Drug Metabolism and Pharmacokinetics (Q.W., Y.L., J.F.), Integrated Drug Discovery (J.L.K., T.K.), and Preclinical Safety Research (E.-J.P., M.S., Z.J.), Sanofi, Cambridge, Massachusetts; Department of Drug Metabolism and Pharmacokinetics, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany (B.M., A.M., C.H., J.R.)
| | - Emma-Jane Poulton
- Department of Drug Metabolism and Pharmacokinetics (Q.W., Y.L., J.F.), Integrated Drug Discovery (J.L.K., T.K.), and Preclinical Safety Research (E.-J.P., M.S., Z.J.), Sanofi, Cambridge, Massachusetts; Department of Drug Metabolism and Pharmacokinetics, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany (B.M., A.M., C.H., J.R.)
| | - Maja Sedic
- Department of Drug Metabolism and Pharmacokinetics (Q.W., Y.L., J.F.), Integrated Drug Discovery (J.L.K., T.K.), and Preclinical Safety Research (E.-J.P., M.S., Z.J.), Sanofi, Cambridge, Massachusetts; Department of Drug Metabolism and Pharmacokinetics, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany (B.M., A.M., C.H., J.R.)
| | - Zaid Jayyosi
- Department of Drug Metabolism and Pharmacokinetics (Q.W., Y.L., J.F.), Integrated Drug Discovery (J.L.K., T.K.), and Preclinical Safety Research (E.-J.P., M.S., Z.J.), Sanofi, Cambridge, Massachusetts; Department of Drug Metabolism and Pharmacokinetics, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany (B.M., A.M., C.H., J.R.)
| | - Jens Riedel
- Department of Drug Metabolism and Pharmacokinetics (Q.W., Y.L., J.F.), Integrated Drug Discovery (J.L.K., T.K.), and Preclinical Safety Research (E.-J.P., M.S., Z.J.), Sanofi, Cambridge, Massachusetts; Department of Drug Metabolism and Pharmacokinetics, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany (B.M., A.M., C.H., J.R.)
| | - Jennifer Fretland
- Department of Drug Metabolism and Pharmacokinetics (Q.W., Y.L., J.F.), Integrated Drug Discovery (J.L.K., T.K.), and Preclinical Safety Research (E.-J.P., M.S., Z.J.), Sanofi, Cambridge, Massachusetts; Department of Drug Metabolism and Pharmacokinetics, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany (B.M., A.M., C.H., J.R.)
| |
Collapse
|
7
|
Rodríguez-Espinosa D, Cuadrado-Payán E, Rico N, Torra M, Fernández RM, Gómez M, Morantes L, Casals G, Rodriguez-Garcia M, Maduell F, Broseta JJ. Comparative Effects of Acetate- and Citrate-Based Dialysates on Dialysis Dose and Protein-Bound Uremic Toxins in Hemodiafiltration Patients: Exploring the Impact of Calcium and Magnesium Concentrations. Toxins (Basel) 2024; 16:426. [PMID: 39453202 PMCID: PMC11511190 DOI: 10.3390/toxins16100426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024] Open
Abstract
Modern hemodialysis employs weak acids as buffers to prevent bicarbonate precipitation with calcium or magnesium. Acetate, the most used acid, is linked to chronic inflammation and poor dialysis tolerance. Citrate has emerged as a potential alternative, though its effect on dialysis efficiency is not clear. This study aims to compare the efficacy of acetate- and citrate-based dialysates, focusing on protein-bound uremic toxins and dialysis doses. This single-center prospective crossover study includes prevalent patients participating in a thrice-weekly online hemodiafiltration program. Four dialysates were tested: two acetate-based (1.25 and 1.5 mmol/L calcium) and two citrate-based (1.5 mmol/L calcium with 0.5 and 0.75 mmol/L magnesium). Pre- and post-dialysis blood samples of eighteen patients were analyzed for urea, creatinine, p-cresyl sulfate, indoxyl sulfate, and albumin. Statistical significance was assessed using paired t-tests and repeated measures of ANOVA. There were no significant differences in dialysis dose (Kt), urea, creatinine, or indoxyl sulfate reduction ratios between acetate- and citrate-based dialysates. However, a significant decrease in the reduction ratio of p-cresyl sulfate was observed with the acetate dialysate containing 1.25 mmol/L calcium and the citrate dialysate with 0.5 mmol/L magnesium compared to the acetate dialysate containing 1.5 mmol/L calcium and the citrate dialysate with 0.75 mmol/L magnesium (51.56 ± 4.75 and 53.02 ± 4.52 vs. 65.25 ± 3.38 and 58.66 ± 4.16, p 0.007). No differences in dialysis dose were found between acetate- and citrate-based dialysates. However, citrate dialysates with lower calcium and magnesium concentrations may reduce the albumin displacement of p-cresyl sulfate. Further studies are needed to understand the observed differences and optimize the dialysate composition for the better clearance of protein-bound uremic toxins.
Collapse
Affiliation(s)
- Diana Rodríguez-Espinosa
- Nephrology and Renal Transplantation, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (D.R.-E.); (E.C.-P.); (M.G.); (L.M.); (F.M.)
| | - Elena Cuadrado-Payán
- Nephrology and Renal Transplantation, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (D.R.-E.); (E.C.-P.); (M.G.); (L.M.); (F.M.)
| | - Naira Rico
- Biochemistry and Molecular Genetics Department-CDB, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (N.R.); (M.T.); (R.M.F.); (G.C.); (M.R.-G.)
| | - Mercè Torra
- Biochemistry and Molecular Genetics Department-CDB, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (N.R.); (M.T.); (R.M.F.); (G.C.); (M.R.-G.)
| | - Rosa María Fernández
- Biochemistry and Molecular Genetics Department-CDB, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (N.R.); (M.T.); (R.M.F.); (G.C.); (M.R.-G.)
| | - Miquel Gómez
- Nephrology and Renal Transplantation, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (D.R.-E.); (E.C.-P.); (M.G.); (L.M.); (F.M.)
| | - Laura Morantes
- Nephrology and Renal Transplantation, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (D.R.-E.); (E.C.-P.); (M.G.); (L.M.); (F.M.)
| | - Gregori Casals
- Biochemistry and Molecular Genetics Department-CDB, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (N.R.); (M.T.); (R.M.F.); (G.C.); (M.R.-G.)
| | - Maria Rodriguez-Garcia
- Biochemistry and Molecular Genetics Department-CDB, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (N.R.); (M.T.); (R.M.F.); (G.C.); (M.R.-G.)
| | - Francisco Maduell
- Nephrology and Renal Transplantation, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (D.R.-E.); (E.C.-P.); (M.G.); (L.M.); (F.M.)
| | - José Jesús Broseta
- Nephrology and Renal Transplantation, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (D.R.-E.); (E.C.-P.); (M.G.); (L.M.); (F.M.)
| |
Collapse
|
8
|
Snauwaert E, De Buyser S, Desloovere A, Van Biesen W, Raes A, Glorieux G, Collard L, Van Hoeck K, Van Dyck M, Godefroid N, Vande Walle J, Eloot S. Assessment of Within- and Inter-Patient Variability of Uremic Toxin Concentrations in Children with CKD. Toxins (Basel) 2024; 16:349. [PMID: 39195759 PMCID: PMC11359554 DOI: 10.3390/toxins16080349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 08/29/2024] Open
Abstract
To promote improved trial design in upcoming randomized clinical trials in childhood chronic kidney disease (CKD), insight in the within- and inter-patient variability of uremic toxins with its nutritional, treatment- and patient-related confounding factors is of utmost importance. In this study, the within- and inter-patient variability of a selection of uremic toxins in a longitudinal cohort of children diagnosed with CKD was assessed, using the intraclass correlation coefficient (ICC) and the within-patient coefficient of variation (CV). Subsequently, the contribution of anthropometry, estimated glomerular filtration rate (eGFR), dietary fiber and protein, and use of (prophylactic) antibiotics to uremic toxin variability was evaluated. Based on 403 observations from 62 children (median seven visits per patient; 9.4 ± 5.3 years; 68% males; eGFR 38.5 [23.1; 64.0] mL/min/1.73 m2) collected over a maximum of 2 years, we found that the within-patient variability is high for especially protein-bound uremic toxins (PBUTs) (ICC < 0.7; within-patient CV 37-67%). Moreover, eGFR was identified as a predominant contributor to the within- and inter-patient variability for the majority of solutes, while the impact of the child's anthropometry, fiber and protein intake, and antibiotics on the variability of uremic toxin concentrations was limited. Based on these findings, we would recommend future intervention studies that attempt to decrease uremic toxin levels to select a (non-dialysis) CKD study population with a narrow eGFR range. As the expected effect of the selected intervention should exceed the inter-patient variability of the selected uremic toxins, a narrow eGFR range might aid in improving the trial design.
Collapse
Affiliation(s)
- Evelien Snauwaert
- Ghent University Hospital, 9000 Ghent, Belgium; (A.D.); (W.V.B.); (A.R.); (G.G.); (J.V.W.); (S.E.)
| | - Stefanie De Buyser
- Biostatistics Unit, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium;
| | - An Desloovere
- Ghent University Hospital, 9000 Ghent, Belgium; (A.D.); (W.V.B.); (A.R.); (G.G.); (J.V.W.); (S.E.)
| | - Wim Van Biesen
- Ghent University Hospital, 9000 Ghent, Belgium; (A.D.); (W.V.B.); (A.R.); (G.G.); (J.V.W.); (S.E.)
| | - Ann Raes
- Ghent University Hospital, 9000 Ghent, Belgium; (A.D.); (W.V.B.); (A.R.); (G.G.); (J.V.W.); (S.E.)
| | - Griet Glorieux
- Ghent University Hospital, 9000 Ghent, Belgium; (A.D.); (W.V.B.); (A.R.); (G.G.); (J.V.W.); (S.E.)
| | | | | | | | | | - Johan Vande Walle
- Ghent University Hospital, 9000 Ghent, Belgium; (A.D.); (W.V.B.); (A.R.); (G.G.); (J.V.W.); (S.E.)
| | - Sunny Eloot
- Ghent University Hospital, 9000 Ghent, Belgium; (A.D.); (W.V.B.); (A.R.); (G.G.); (J.V.W.); (S.E.)
| |
Collapse
|
9
|
Bakinowska E, Olejnik-Wojciechowska J, Kiełbowski K, Skoryk A, Pawlik A. Pathogenesis of Sarcopenia in Chronic Kidney Disease-The Role of Inflammation, Metabolic Dysregulation, Gut Dysbiosis, and microRNA. Int J Mol Sci 2024; 25:8474. [PMID: 39126043 PMCID: PMC11313360 DOI: 10.3390/ijms25158474] [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: 05/31/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Chronic kidney disease (CKD) is a progressive disorder associated with a decline in kidney function. Consequently, patients with advanced stages of CKD require renal replacement therapies, such as dialysis and kidney transplantation. Various conditions lead to the development of CKD, including diabetes mellitus, hypertension, and glomerulonephritis, among others. The disease is associated with metabolic and hormonal dysregulation, including uraemia and hyperparathyroidism, as well as with low-grade systemic inflammation. Altered homeostasis increases the risk of developing severe comorbidities, such as cardiovascular diseases or sarcopenia, which increase mortality. Sarcopenia is defined as a progressive decline in muscle mass and function. However, the precise mechanisms that link CKD and the development of sarcopenia are poorly understood. Knowledge about these linking mechanisms might lead to the introduction of precise treatment strategies that could prevent muscle wasting. This review discusses inflammatory mediators, metabolic and hormonal dysregulation, gut microbiota dysbiosis, and non-coding RNA alterations that could link CKD and sarcopenia.
Collapse
Affiliation(s)
- Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (E.B.); (J.O.-W.); (K.K.); (A.S.)
| | - Joanna Olejnik-Wojciechowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (E.B.); (J.O.-W.); (K.K.); (A.S.)
- Independent Laboratory of Community Nursing, Pomeranian Medical University, 71-210 Szczecin, Poland
| | - Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (E.B.); (J.O.-W.); (K.K.); (A.S.)
| | - Anastasiia Skoryk
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (E.B.); (J.O.-W.); (K.K.); (A.S.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (E.B.); (J.O.-W.); (K.K.); (A.S.)
| |
Collapse
|
10
|
Al-Dajani AR, Hou QK, Kiang TKL. Liquid Chromatography-Mass Spectrometry Analytical Methods for the Quantitation of p-Cresol Sulfate and Indoxyl Sulfate in Human Matrices: Biological Applications and Diagnostic Potentials. Pharmaceutics 2024; 16:743. [PMID: 38931865 PMCID: PMC11206749 DOI: 10.3390/pharmaceutics16060743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/20/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Indoxyl sulfate (IxS) and p-cresyl sulfate (pCS) are toxic uremic compounds with documented pathological outcomes. This review critically and comprehensively analyzes the available liquid chromatography-mass spectrometry methods quantifying IxS and pCS in human matrices and the biological applications of these validated assays. Embase, Medline, PubMed, Scopus, and Web of Science were searched until December 2023 to identify assays with complete analytical and validation data (N = 23). Subsequently, citation analysis with PubMed and Scopus was utilized to identify the biological applications for these assays (N = 45). The extraction methods, mobile phase compositions, chromatography, and ionization methods were evaluated with respect to overall assay performance (e.g., sensitivity, separation, interference). Most of the assays focused on human serum/plasma, utilizing acetonitrile or methanol (with ammonium acetate/formate or formic/acetic acid), liquid-liquid extraction, reverse phase (e.g., C18) chromatography, and gradient elution for analyte separation. Mass spectrometry conditions were also consistent in the identified papers, with negative electrospray ionization, select multiple reaction monitoring transitions and deuterated internal standards being the most common approaches. The validated biological applications indicated IxS and/or pCS were correlated with renal disease progression and cardiovascular outcomes, with limited data on central nervous system disorders. Methods for reducing IxS and/or pCS concentrations were also identified (e.g., drugs, natural products, diet, dialysis, transplantation) where inconsistent findings have been reported. The clinical monitoring of IxS and pCS is gaining significant interest, and this review will serve as a useful compendium for scientists and clinicians.
Collapse
Affiliation(s)
| | | | - Tony K. L. Kiang
- Katz Group Centre for Pharmacy and Health Research, Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada; (A.R.A.-D.); (Q.K.H.)
| |
Collapse
|
11
|
Cha RH. Pharmacologic therapeutics in sarcopenia with chronic kidney disease. Kidney Res Clin Pract 2024; 43:143-155. [PMID: 38389147 PMCID: PMC11016676 DOI: 10.23876/j.krcp.23.094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/25/2023] [Accepted: 10/24/2023] [Indexed: 02/24/2024] Open
Abstract
Inflammation, metabolic acidosis, renin-angiotensin system activation, insulin resistance, and impaired perfusion to skeletal muscles, among others, are possible causes of uremic sarcopenia. These conditions induce the activation of the nuclear factor-kappa B and mitogen-activated protein kinase pathways, adenosine triphosphate ubiquitin-proteasome system, and reactive oxygen species system, resulting in protein catabolism. Strategies for the prevention and treatment of sarcopenia in chronic kidney disease (CKD) are aerobic and resistance exercises along with nutritional interventions. Anabolic hormones have shown beneficial effects. Megestrol acetate increased weight, protein catabolic rate, and albumin concentration, and it increased intracellular water component and muscle mass. Vitamin D supplementation showed improvement in physical function, muscle strength, and muscle mass. Correction of metabolic acidosis showed an increase in protein intake, serum albumin levels, body weight, and mid-arm circumference. The kidney- gut-muscle axis indicates that dysbiosis and changes in gut-derived uremic toxins and short-chain fatty acids affect muscle mass, composition, strength, and functional capacity. Biotic supplements, AST-120 administration, hemodiafiltration, and preservation of residual renal function are alleged to reduce uremic toxins, including indoxyl sulfate (IS) and p-cresyl sulfate (PCS). Synbiotics reversed the microbiota change in CKD patients and decreased uremic toxins. AST-120 administration changed the overall gut microbiota composition in CKD. AST-120 prevented IS and PCS tissue accumulation, ameliorated muscle atrophy, improved exercise capacity and mitochondrial biogenesis, restored epithelial tight junction proteins, and reduced plasma endotoxin levels and markers of oxidative stress and inflammation. In a human study, the addition of AST-120 to standard treatment had modest beneficial effects on gait speed change and quality of life.
Collapse
Affiliation(s)
- Ran-hui Cha
- Department of Internal Medicine, National Medical Center, Seoul, Republic of Korea
| |
Collapse
|
12
|
Akl EM, El-Eraki JM, Elfallah AA, Mohamed NH, Maher AM, Mansour AE, Abdelsalam OH. Does Indoxyl Sulfate Have a Role in Uremic Pruritus? A Laboratory and Interventional Study. J Cutan Med Surg 2024; 28:44-50. [PMID: 38156627 DOI: 10.1177/12034754231220935] [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] [Indexed: 01/03/2024]
Abstract
BACKGROUND Pruritus is a common complaint in patients with end-stage renal disease. Indoxyl sulfate (IS) is a tryptophan end metabolite extremely renal excreted. Activated charcoal can interfere with IS intestinal absorption. OBJECTIVES To evaluate the serum level of IS and the effect of activated charcoal on uremic pruritus. MATERIALS AND METHODS In all, 135 participants were divided into 2 main groups. In total, 45 normal and healthy individuals as a control group and 90 patients on regular hemodialysis; 45 of these patients had uremic pruritus and the other 45 were not complaining of uremic pruritus. Serum IS was measured. Activated charcoal was used by patients with uremic pruritus. The severity of pruritus and Dermatology Life Quality Index (DLQI) were assessed. RESULTS The serum IS was significantly elevated in uremic patients than in control subjects (P < .001) and significantly elevated in uremic patients without pruritus (P < .001). Furthermore, there were positive significant correlations between the serum IS and both severity of pruritus (P < .001) and DLQI (P < .001). After activated charcoal usage, there was a significant decrease in IS level with the improvement of pruritus and quality of life of patients. CONCLUSIONS IS may play a role in uremic pruritus. Activated charcoal could be considered a treatment for uremic pruritus.
Collapse
Affiliation(s)
- Essam Mohamed Akl
- Department of Dermatology and Andrology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Jeylan M El-Eraki
- Specialist of Dermatology and Andrology, Ministry of Health and Population, Cairo, Egypt
| | - Assma A Elfallah
- Department of Clinical Pathology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Nelly H Mohamed
- Specialist of Clinical Pathology, Ministry of Health and Population, Benha, Egypt
| | - Amr M Maher
- Specialist of Internal Medicine, Ministry of Health and Population, Cairo, Egypt
| | - Ahmed E Mansour
- Department of Internal Medicine, Faculty of Medicine, Benha University, Benha, Egypt
| | - Osama H Abdelsalam
- Department of Dermatology and Andrology, Faculty of Medicine, Benha University, Benha, Egypt
| |
Collapse
|
13
|
Pantazi AC, Kassim MAK, Nori W, Tuta LA, Mihai CM, Chisnoiu T, Balasa AL, Mihai L, Lupu A, Frecus CE, Lupu VV, Chirila SI, Badescu AG, Hangan LT, Cambrea SC. Clinical Perspectives of Gut Microbiota in Patients with Chronic Kidney Disease and End-Stage Kidney Disease: Where Do We Stand? Biomedicines 2023; 11:2480. [PMID: 37760920 PMCID: PMC10525496 DOI: 10.3390/biomedicines11092480] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/26/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
The gut microbiota (GM) plays a vital role in human health, with increasing evidence linking its imbalance to chronic kidney disease and end-stage kidney disease. Although the exact methods underlying kidney-GM crosstalk are not fully understood, interventions targeting GM were made and lay in three aspects: diagnostic, predictive, and therapeutic interventions. While these interventions show promising results in reducing uremic toxins and inflammation, challenges remain in the form of patient-specific GM variability, potential side effects, and safety concerns. Our understanding of GMs role in kidney disease is still evolving, necessitating further research to elucidate the causal relationship and mechanistic interactions. Personalized interventions focusing on specific GM signatures could enhance patient outcomes. However, comprehensive clinical trials are needed to validate these approaches' safety, efficacy, and feasibility.
Collapse
Affiliation(s)
| | | | - Wassan Nori
- College of Medicine, Mustansiriyah University, Baghdad 10052, Iraq;
| | - Liliana Ana Tuta
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania (L.A.T.)
- Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Cristina Maria Mihai
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania (L.A.T.)
- Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Tatiana Chisnoiu
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania (L.A.T.)
- Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Adriana Luminita Balasa
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania (L.A.T.)
- Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Larisia Mihai
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania (L.A.T.)
- Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Ancuta Lupu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Corina Elena Frecus
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania (L.A.T.)
- Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Vasile Valeriu Lupu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Sergiu Ioachim Chirila
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania (L.A.T.)
| | | | - Laurentiu-Tony Hangan
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania (L.A.T.)
| | - Simona Claudia Cambrea
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania (L.A.T.)
| |
Collapse
|
14
|
Esposito P, Verzola D, Saio M, Picciotto D, Frascio M, Laudon A, Zanetti V, Brunori G, Garibotto G, Viazzi F. The Contribution of Muscle Innate Immunity to Uremic Cachexia. Nutrients 2023; 15:2832. [PMID: 37447158 PMCID: PMC10343562 DOI: 10.3390/nu15132832] [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: 05/25/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
Protein energy wasting (PEW) is a common complication both in chronic kidney disease (CKD) and end-stage kidney disease (ESKD). Of note, PEW is one of the stronger predictors of morbidity and mortality in this patient population. The pathogenesis of PEW involves several mechanisms, including anorexia, insulin resistance, acidosis and low-grade inflammation. In addition, "sterile" muscle inflammation contributes to PEW at an advanced CKD stage. Both immune and resident muscle cells can activate innate immunity; thus, they have critical roles in triggering "sterile" tissue inflammation. Toll-like receptor 4 (TLR4) can detect endogenous danger-associated molecular patterns generated or retained in blood in uremia and induce a sterile muscle inflammatory response via NF-κB in myocytes. In addition, TLR4, though the activation of the NLRP3 inflammasome, links the sensing of metabolic uremic stress in muscle to the activation of pro-inflammatory cascades, which lead to the production of IL-1β and IL-18. Finally, uremia-induced accelerated cell senescence is associated with a secretory phenotype that favors fibrosis in muscle. Targeting these innate immune pathways could lead to novel therapies for CKD-related PEW.
Collapse
Affiliation(s)
- Pasquale Esposito
- Division of Nephrology, Dialysis and Transplantation, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (P.E.); (M.S.); (D.P.); (V.Z.); (F.V.)
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy;
| | - Daniela Verzola
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy;
| | - Michela Saio
- Division of Nephrology, Dialysis and Transplantation, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (P.E.); (M.S.); (D.P.); (V.Z.); (F.V.)
| | - Daniela Picciotto
- Division of Nephrology, Dialysis and Transplantation, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (P.E.); (M.S.); (D.P.); (V.Z.); (F.V.)
| | - Marco Frascio
- Division of Surgery, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
- Department of Surgical Sciences and Integrated Diagnostics, University of Genova, 16132 Genova, Italy
| | - Alessandro Laudon
- Division of Nephrology, Ospedale Santa Chiara, 38122 Trento, Italy; (A.L.); (G.B.)
| | - Valentina Zanetti
- Division of Nephrology, Dialysis and Transplantation, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (P.E.); (M.S.); (D.P.); (V.Z.); (F.V.)
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy;
| | - Giuliano Brunori
- Division of Nephrology, Ospedale Santa Chiara, 38122 Trento, Italy; (A.L.); (G.B.)
| | - Giacomo Garibotto
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy;
| | - Francesca Viazzi
- Division of Nephrology, Dialysis and Transplantation, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (P.E.); (M.S.); (D.P.); (V.Z.); (F.V.)
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy;
| |
Collapse
|
15
|
Gut microbiome studies in CKD: opportunities, pitfalls and therapeutic potential. Nat Rev Nephrol 2023; 19:87-101. [PMID: 36357577 DOI: 10.1038/s41581-022-00647-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2022] [Indexed: 11/12/2022]
Abstract
Interest in gut microbiome dysbiosis and its potential association with the development and progression of chronic kidney disease (CKD) has increased substantially in the past 6 years. In parallel, the microbiome field has matured considerably as the importance of host-related and environmental factors is increasingly recognized. Past research output in the context of CKD insufficiently considered the myriad confounding factors that are characteristic of the disease. Gut microbiota-derived metabolites remain an interesting therapeutic target to decrease uraemic (cardio)toxicity. However, future studies on the effect of dietary and biotic interventions will require harmonization of relevant readouts to enable an in-depth understanding of the underlying beneficial mechanisms. High-quality standards throughout the entire microbiome analysis workflow are also of utmost importance to obtain reliable and reproducible results. Importantly, investigating the relative composition and abundance of gut bacteria, and their potential association with plasma uraemic toxins levels is not sufficient. As in other fields, the time has come to move towards in-depth quantitative and functional exploration of the patient's gut microbiome by relying on confounder-controlled quantitative microbial profiling, shotgun metagenomics and in vitro simulations of microorganism-microorganism and host-microorganism interactions. This step is crucial to enable the rational selection and monitoring of dietary and biotic intervention strategies that can be deployed as a personalized intervention in CKD.
Collapse
|
16
|
Caggiano G, Stasi A, Franzin R, Fiorentino M, Cimmarusti MT, Deleonardis A, Palieri R, Pontrelli P, Gesualdo L. Fecal Microbiota Transplantation in Reducing Uremic Toxins Accumulation in Kidney Disease: Current Understanding and Future Perspectives. Toxins (Basel) 2023; 15:toxins15020115. [PMID: 36828429 PMCID: PMC9965504 DOI: 10.3390/toxins15020115] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/21/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
During the past decades, the gut microbiome emerged as a key player in kidney disease. Dysbiosis-related uremic toxins together with pro-inflammatory mediators are the main factors in a deteriorating kidney function. The toxicity of uremic compounds has been well-documented in a plethora of pathophysiological mechanisms in kidney disease, such as cardiovascular injury (CVI), metabolic dysfunction, and inflammation. Accumulating data on the detrimental effect of uremic solutes in kidney disease supported the development of many strategies to restore eubiosis. Fecal microbiota transplantation (FMT) spread as an encouraging treatment for different dysbiosis-associated disorders. In this scenario, flourishing studies indicate that fecal transplantation could represent a novel treatment to reduce the uremic toxins accumulation. Here, we present the state-of-the-art concerning the application of FMT on kidney disease to restore eubiosis and reverse the retention of uremic toxins.
Collapse
|
17
|
van Ham WB, Cornelissen CM, van Veen TAB. Uremic toxins in chronic kidney disease highlight a fundamental gap in understanding their detrimental effects on cardiac electrophysiology and arrhythmogenesis. Acta Physiol (Oxf) 2022; 236:e13888. [PMID: 36148604 PMCID: PMC9787632 DOI: 10.1111/apha.13888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/09/2022] [Accepted: 09/18/2022] [Indexed: 01/29/2023]
Abstract
Chronic kidney disease (CKD) and cardiovascular disease (CVD) have an estimated 700-800 and 523 million cases worldwide, respectively, with CVD being the leading cause of death in CKD patients. The pathophysiological interplay between the heart and kidneys is defined as the cardiorenal syndrome (CRS), in which worsening of kidney function is represented by increased plasma concentrations of uremic toxins (UTs), culminating in dialysis patients. As there is a high incidence of CVD in CKD patients, accompanied by arrhythmias and sudden cardiac death, knowledge on electrophysiological remodeling would be instrumental for understanding the CRS. While the interplay between both organs is clearly of importance in CRS, the involvement of UTs in pro-arrhythmic remodeling is only poorly investigated, especially regarding the mechanistic background. Currently, the clinical approach against potential arrhythmic events is mainly restricted to symptom treatment, stressing the need for fundamental research on UT in relation to electrophysiology. This review addresses the existing knowledge of UTs and cardiac electrophysiology, and the experimental research gap between fundamental research and clinical research of the CRS. Clinically, mainly absorbents like ibuprofen and AST-120 are studied, which show limited safe and efficient usability. Experimental research shows disturbances in cardiac electrical activation and conduction after inducing CKD or exposure to UTs, but are scarcely present or focus solely on already well-investigated UTs. Based on UTs data derived from CKD patient cohort studies, a clinically relevant overview of physiological and pathological UTs concentrations is created. Using this, future experimental research is stimulated to involve electrophysiologically translatable animals, such as rabbits, or in vitro engineered heart tissues.
Collapse
Affiliation(s)
- Willem B. van Ham
- Department of Medical Physiology, Division Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Carlijn M. Cornelissen
- Department of Medical Physiology, Division Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Toon A. B. van Veen
- Department of Medical Physiology, Division Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| |
Collapse
|
18
|
Madella AM, Van Bergenhenegouwen J, Garssen J, Masereeuw R, Overbeek SA. Microbial-Derived Tryptophan Catabolites, Kidney Disease and Gut Inflammation. Toxins (Basel) 2022; 14:toxins14090645. [PMID: 36136583 PMCID: PMC9505404 DOI: 10.3390/toxins14090645] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Uremic metabolites, molecules either produced by the host or from the microbiota population existing in the gastrointestinal tract that gets excreted by the kidneys into urine, have significant effects on both health and disease. Tryptophan-derived catabolites are an important group of bacteria-produced metabolites with an extensive contribution to intestinal health and, eventually, chronic kidney disease (CKD) progression. The end-metabolite, indoxyl sulfate, is a key contributor to the exacerbation of CKD via the induction of an inflammatory state and oxidative stress affecting various organ systems. Contrastingly, other tryptophan catabolites positively contribute to maintaining intestinal homeostasis and preventing intestinal inflammation—activities signaled through nuclear receptors in particular—the aryl hydrocarbon receptor (AhR) and the pregnane X receptor (PXR). This review discusses the origins of these catabolites, their effect on organ systems, and how these can be manipulated therapeutically in the future as a strategy to treat CKD progression and gut inflammation management. Furthermore, the use of biotics (prebiotics, probiotics, synbiotics) as a means to increase the presence of beneficial short-chain fatty acids (SCFAs) to achieve intestinal homeostasis is discussed.
Collapse
Affiliation(s)
- Avra Melina Madella
- Department of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
- Correspondence: (A.M.M.); or (S.A.O.); Tel.: +31-30-209-5000 (S.A.O.)
| | - Jeroen Van Bergenhenegouwen
- Department of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
- Danone Nutricia Research, Uppsalalaan 12, Utrecht Science Park, 3584 CT Utrecht, The Netherlands
| | - Johan Garssen
- Department of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
- Danone Nutricia Research, Uppsalalaan 12, Utrecht Science Park, 3584 CT Utrecht, The Netherlands
| | - Rosalinde Masereeuw
- Department of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Saskia Adriana Overbeek
- Department of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
- Danone Nutricia Research, Uppsalalaan 12, Utrecht Science Park, 3584 CT Utrecht, The Netherlands
- Correspondence: (A.M.M.); or (S.A.O.); Tel.: +31-30-209-5000 (S.A.O.)
| |
Collapse
|
19
|
Goto M, Kobira Y, Kaneko S, Arima H, Michihara A, Azuma K, Higashi T, Motoyama K, Watanabe H, Maruyama T, Kadowaki D, Otagiri M, Iohara D, Hirayama F, Anraku M. The Effects of Sacran, a Sulfated Polysaccharide, on Gut Microbiota Using Chronic Kidney Disease Model Rats. Biol Pharm Bull 2022; 45:576-582. [DOI: 10.1248/bpb.b21-00897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Miwa Goto
- Faculty of Pharmaceutical Sciences, Sojo University
| | - Yusei Kobira
- Faculty of Pharmaceutical Sciences, Sojo University
| | | | | | - Akihiro Michihara
- Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University
| | - Kazuo Azuma
- Department of Veterinary Clinical Medicine, Tottori University
| | - Taishi Higashi
- Graduate School of Pharmaceutical Sciences, Kumamoto University
| | | | | | - Toru Maruyama
- Graduate School of Pharmaceutical Sciences, Kumamoto University
| | | | | | | | | | | |
Collapse
|
20
|
Cha RH, Kang SH, Han MY, An WS, Kim SH, Kim JC. Effects of AST-120 on muscle health and quality of life in chronic kidney disease patients: results of RECOVERY study. J Cachexia Sarcopenia Muscle 2022; 13:397-408. [PMID: 34862753 PMCID: PMC8818653 DOI: 10.1002/jcsm.12874] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/28/2021] [Accepted: 10/30/2021] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND The prevalence of sarcopenia is increased with declining renal function. Elevated serum indoxyl sulfate levels are associated with poor skeletal muscle conditions. We aimed to determine the effects of AST-120, the oral adsorbent of indoxyl sulfate, on sarcopenia and sarcopenia-associated factors in chronic kidney disease patients. METHODS This was a 48 week, randomized controlled, parallel group, open-label, multicentre trial (n = 150). The participants were randomly assigned in a 1:1 ratio to the control (CON) and AST-120 (Renamezin®, REN) groups. Outcome measurements were performed at baseline and every 24 weeks for 48 weeks. The primary outcome was gait speed difference ≥0.1 m/s between the two groups, and secondary outcomes included hand grip strength, muscle mass, and health-related quality of life. RESULTS A difference of gait speed ≥0.1 m/s was not observed during the study period. The mean dynamic-start gait speed in the REN group increased from baseline to 48 weeks (1.04 ± 0.31 to 1.08 ± 0.32 m/s, P = 0.019). The static-start gait speed changed by -0.024 and 0.04 m/s (P = 0.049) in the CON and REN groups over 48 weeks, respectively. Hand grip strength decreased during the first 24 weeks and did not significantly change over the next 24 weeks in either group. The proportion of low muscle mass or sarcopenia at baseline was larger in the REN group than in the CON group, but the difference attenuated over the study period [low muscle mass and sarcopenia in the CON and REN groups at baseline, 4.0% vs. 18.9% (P = 0.004) and 2.7% vs. 13.5% (P = 0.017); at 24 weeks, 2.9% vs. 13.6% (P = 0.021) and 1.4% vs. 10.5% (P = 0.029); and at 48 weeks, 7.6% vs. 12.9% (P = 0.319) and 4.5% vs. 8.1% (P = 0.482), respectively]. Bodily pain, vitality, symptoms/problems, and cognitive function in the REN group improved, while the quality of social interactions and the kidney disease effects in the CON group aggravated from baseline to 48 weeks. Interaction between time and group was evident only in symptoms/problems, cognitive function, and kidney disease effects. CONCLUSIONS The addition of AST-120 to standard treatment in chronic kidney disease patients did not make a significant difference in gait speed, although AST-120 modestly had beneficial effects on gait speed change and quality of life and showed the potential to improve sarcopenia. (clinicaltrials.gov: NCT03788252).
Collapse
Affiliation(s)
- Ran-Hui Cha
- Department of Internal Medicine, National Medical Center, Seoul, Republic of Korea
| | - Seok Hui Kang
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Mi Yeun Han
- Department of Internal Medicine, Hallym University Hangang Sacred Heart Hospital, Seoul, Republic of Korea
| | - Won Suk An
- Department of Internal Medicine, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Su-Hyun Kim
- Department of Internal Medicine, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Jun Chul Kim
- Department of Internal Medicine, CHA Gumi Medical Center, CHA University, Gumi, Republic of Korea
| |
Collapse
|
21
|
Takkavatakarn K, Puapatanakul P, Phannajit J, Sukkumme W, Chariyavilaskul P, Sitticharoenchai P, Leelahavanichkul A, Katavetin P, Praditpornsilpa K, Eiam-Ong S, Susantitaphong P. Protein-Bound Uremic Toxins Lowering Effect of Sevelamer in Pre-Dialysis Chronic Kidney Disease Patients with Hyperphosphatemia: A Randomized Controlled Trial. Toxins (Basel) 2021; 13:toxins13100688. [PMID: 34678981 PMCID: PMC8539528 DOI: 10.3390/toxins13100688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 11/16/2022] Open
Abstract
P-cresyl sulfate and indoxyl sulfate are strongly associated with cardiovascular events and all-cause mortality in chronic kidney disease (CKD). This randomized controlled trial was conducted to compare the effects between sevelamer and calcium carbonate on protein-bound uremic toxins in pre-dialysis CKD patients with hyperphosphatemia. Forty pre-dialysis CKD patients with persistent hyperphosphatemia were randomly assigned to receive either 2400 mg of sevelamer daily or 1500 mg of calcium carbonate daily for 24 weeks. A significant decrease of total serum p-cresyl sulfate was observed in sevelamer therapy compared to calcium carbonate therapy (mean difference between two groups −5.61 mg/L; 95% CI −11.01 to −0.27 mg/L; p = 0.04). There was no significant difference in serum indoxyl sulfate levels (p = 0.36). Sevelamer had effects in terms of lowering fibroblast growth factor 23 (p = 0.01) and low-density lipoprotein cholesterol levels (p = 0.04). Sevelamer showed benefits in terms of retarding CKD progression. Changes in vascular stiffness were not found in this study.
Collapse
Affiliation(s)
- Kullaya Takkavatakarn
- Division of Nephrology, Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok 10330, Thailand; (K.T.); (P.P.); (J.P.); (A.L.); (P.K.); (K.P.); (S.E.-O.)
| | - Pongpratch Puapatanakul
- Division of Nephrology, Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok 10330, Thailand; (K.T.); (P.P.); (J.P.); (A.L.); (P.K.); (K.P.); (S.E.-O.)
| | - Jeerath Phannajit
- Division of Nephrology, Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok 10330, Thailand; (K.T.); (P.P.); (J.P.); (A.L.); (P.K.); (K.P.); (S.E.-O.)
| | - Warumphon Sukkumme
- Clinical Pharmacokinetics and Pharmacogenomics Research Unit, Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (W.S.); (P.C.)
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pajaree Chariyavilaskul
- Clinical Pharmacokinetics and Pharmacogenomics Research Unit, Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (W.S.); (P.C.)
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Patita Sitticharoenchai
- Division of Cardiology, Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Asada Leelahavanichkul
- Division of Nephrology, Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok 10330, Thailand; (K.T.); (P.P.); (J.P.); (A.L.); (P.K.); (K.P.); (S.E.-O.)
- Department of Microbiology, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pisut Katavetin
- Division of Nephrology, Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok 10330, Thailand; (K.T.); (P.P.); (J.P.); (A.L.); (P.K.); (K.P.); (S.E.-O.)
| | - Kearkiat Praditpornsilpa
- Division of Nephrology, Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok 10330, Thailand; (K.T.); (P.P.); (J.P.); (A.L.); (P.K.); (K.P.); (S.E.-O.)
| | - Somchai Eiam-Ong
- Division of Nephrology, Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok 10330, Thailand; (K.T.); (P.P.); (J.P.); (A.L.); (P.K.); (K.P.); (S.E.-O.)
| | - Paweena Susantitaphong
- Division of Nephrology, Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok 10330, Thailand; (K.T.); (P.P.); (J.P.); (A.L.); (P.K.); (K.P.); (S.E.-O.)
- Research Unit for Metabolic Bone Disease in CKD Patients, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: ; Tel.: +(662)-256-4251
| |
Collapse
|
22
|
Maheshwari V, Tao X, Thijssen S, Kotanko P. Removal of Protein-Bound Uremic Toxins Using Binding Competitors in Hemodialysis: A Narrative Review. Toxins (Basel) 2021; 13:toxins13090622. [PMID: 34564626 PMCID: PMC8473190 DOI: 10.3390/toxins13090622] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/28/2021] [Accepted: 08/31/2021] [Indexed: 11/16/2022] Open
Abstract
Removal of protein-bound uremic toxins (PBUTs) during conventional dialysis is insufficient. PBUTs are associated with comorbidities and mortality in dialysis patients. Albumin is the primary carrier for PBUTs and only a small free fraction of PBUTs are dialyzable. In the past, we proposed a novel method where a binding competitor is infused upstream of a dialyzer into an extracorporeal circuit. The competitor competes with PBUTs for their binding sites on albumin and increases the free PBUT fraction. Essentially, binding competitor-augmented hemodialysis is a reactive membrane separation technique and is a paradigm shift from conventional dialysis therapies. The proposed method has been tested in silico, ex vivo, and in vivo, and has proven to be very effective in all scenarios. In an ex vivo study and a proof-of-concept clinical study with 18 patients, ibuprofen was used as a binding competitor; however, chronic ibuprofen infusion may affect residual kidney function. Binding competition with free fatty acids significantly improved PBUT removal in pre-clinical rat models. Based on in silico analysis, tryptophan can also be used as a binding competitor; importantly, fatty acids or tryptophan may have salutary effects in HD patients. More chemoinformatics research, pre-clinical, and clinical studies are required to identify ideal binding competitors before routine clinical use.
Collapse
Affiliation(s)
- Vaibhav Maheshwari
- Renal Research Institute, New York, NY 10065, USA; (X.T.); (S.T.); (P.K.)
- Correspondence:
| | - Xia Tao
- Renal Research Institute, New York, NY 10065, USA; (X.T.); (S.T.); (P.K.)
| | - Stephan Thijssen
- Renal Research Institute, New York, NY 10065, USA; (X.T.); (S.T.); (P.K.)
| | - Peter Kotanko
- Renal Research Institute, New York, NY 10065, USA; (X.T.); (S.T.); (P.K.)
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
23
|
Kumar P, Lee JH, Lee J. Diverse roles of microbial indole compounds in eukaryotic systems. Biol Rev Camb Philos Soc 2021; 96:2522-2545. [PMID: 34137156 PMCID: PMC9290978 DOI: 10.1111/brv.12765] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 02/06/2023]
Abstract
Indole and its derivatives are widespread across different life forms, functioning as signalling molecules in prokaryotes and with more diverse roles in eukaryotes. A majority of indoles found in the environment are attributed to bacterial enzymes converting tryptophan into indole and its derivatives. The involvement of indoles among lower organisms as an interspecies and intraspecies signal is well known, with many reports showing that inter‐kingdom interactions involving microbial indole compounds are equally important as they influence defence systems and even the behaviour of higher organisms. This review summarizes recent advances in our understanding of the functional properties of indole and indole derivatives in diverse eukaryotes. Furthermore, we discuss current perspectives on the role of microbial indoles in human diseases such as diabetes, obesity, atherosclerosis, and cancers. Deciphering the function of indoles as biomarkers of metabolic state will facilitate the formulation of diet‐based treatments and open unique therapeutic opportunities.
Collapse
Affiliation(s)
- Prasun Kumar
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541, Republic of Korea
| | - Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541, Republic of Korea
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541, Republic of Korea
| |
Collapse
|
24
|
Cosola C, Rocchetti MT, di Bari I, Acquaviva PM, Maranzano V, Corciulo S, Di Ciaula A, Di Palo DM, La Forgia FM, Fontana S, De Angelis M, Portincasa P, Gesualdo L. An Innovative Synbiotic Formulation Decreases Free Serum Indoxyl Sulfate, Small Intestine Permeability and Ameliorates Gastrointestinal Symptoms in a Randomized Pilot Trial in Stage IIIb-IV CKD Patients. Toxins (Basel) 2021; 13:334. [PMID: 34063068 PMCID: PMC8147955 DOI: 10.3390/toxins13050334] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 02/07/2023] Open
Abstract
Proteolytic dysbiosis of the gut microbiota has been recognized as both a typical feature of chronic kidney disease (CKD) and a risk factor for its progression. Blood accumulation of gut-derived uremic toxins (UTs) like indoxyl sulfate (IS) and p-cresyl sulfate (PCS), intestinal permeability and constipation are typical features accompanying CKD progression and triggering chronic inflammation. In order to verify the efficacy of the innovative synbiotic formulation NATUREN G® in modulating the levels of circulating UTs, intestinal permeability and gastrointestinal symptoms, we set up a randomized, single-blind, placebo-controlled, pilot trial in stage IIIb-IV CKD patients and in healthy controls. Two-month administration of the synbiotic resulted in a decrease of free IS, as compared with the placebo-treated arm, only in the CKD group. The other UTs did not significantly change, although different trends in time (increase in the placebo arm and decrease in the synbiotic arm) were observed. Moreover, after supplementation, reduction of small intestinal permeability and amelioration of abdominal pain and constipation syndromes were observed only in the CKD group. The obtained results suggest the specificity of action of NATUREN G® in CKD and justify further validation in a wider study population.
Collapse
Affiliation(s)
- Carmela Cosola
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari “Aldo Moro”, 70124 Bari, Italy; (C.C.); (I.d.B.); (P.M.A.); (V.M.); (S.C.)
| | - Maria Teresa Rocchetti
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| | - Ighli di Bari
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari “Aldo Moro”, 70124 Bari, Italy; (C.C.); (I.d.B.); (P.M.A.); (V.M.); (S.C.)
| | - Paola Maria Acquaviva
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari “Aldo Moro”, 70124 Bari, Italy; (C.C.); (I.d.B.); (P.M.A.); (V.M.); (S.C.)
| | - Valentina Maranzano
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari “Aldo Moro”, 70124 Bari, Italy; (C.C.); (I.d.B.); (P.M.A.); (V.M.); (S.C.)
| | - Simone Corciulo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari “Aldo Moro”, 70124 Bari, Italy; (C.C.); (I.d.B.); (P.M.A.); (V.M.); (S.C.)
| | - Agostino Di Ciaula
- Department of Biomedical Sciences and Human Oncology, Clinica Medica “A. Murri”, Medical School, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (P.P.)
| | - Domenica Maria Di Palo
- Department of Biomedical Sciences and Human Oncology, Clinica Medica “A. Murri”, Medical School, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (P.P.)
| | - Flavia Maria La Forgia
- Centro Studi e Ricerche Dr. Sergio Fontana (1900–1982), 76012 Canosa, Italy; (F.M.L.F.); (S.F.)
| | - Sergio Fontana
- Centro Studi e Ricerche Dr. Sergio Fontana (1900–1982), 76012 Canosa, Italy; (F.M.L.F.); (S.F.)
| | - Maria De Angelis
- Department of Soil, Plant and Food Science, University of Bari “Aldo Moro”, 70126 Bari, Italy;
| | - Piero Portincasa
- Department of Biomedical Sciences and Human Oncology, Clinica Medica “A. Murri”, Medical School, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (P.P.)
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari “Aldo Moro”, 70124 Bari, Italy; (C.C.); (I.d.B.); (P.M.A.); (V.M.); (S.C.)
| |
Collapse
|
25
|
Rong Y, Kiang TKL. Characterization of human sulfotransferases catalyzing the formation of p-cresol sulfate and identification of mefenamic acid as a potent metabolism inhibitor and potential therapeutic agent for detoxification. Toxicol Appl Pharmacol 2021; 425:115553. [PMID: 33915121 DOI: 10.1016/j.taap.2021.115553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/03/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022]
Abstract
p-Cresol sulfate, the primary metabolite of p-cresol, is a uremic toxin that has been associated with toxicities and mortalities. The study objectives were to i) characterize the contributions of human sulfotransferases (SULT) catalyzing p-cresol sulfate formation using multiple recombinant SULT enzymes (including the polymorphic variant SULT1A1*2), pooled human liver cytosols, and pooled human kidney cytosols; and ii) determine the potencies and mechanisms of therapeutic inhibitors capable of attenuating the production of p-cresol sulfate. Human recombinant SULT1A1 was the primary enzyme responsible for the formation of p-cresol sulfate (Km = 0.19 ± 0.02 μM [with atypical kinetic behavior at lower substrate concentrations; see text discussion], Vmax = 789.5 ± 101.7 nmol/mg/min, Ksi = 2458.0 ± 332.8 μM, mean ± standard deviation, n = 3), while SULT1A3, SULT1B1, SULT1E1, and SULT2A1 contributed negligible or minor roles at toxic p-cresol concentrations. Moreover, human recombinant SULT1A1*2 exhibited reduced enzyme activities (Km = 81.5 ± 31.4 μM, Vmax = 230.6 ± 17.7 nmol/mg/min, Ksi = 986.0 ± 434.4 μM) compared to the wild type. The sulfonation of p-cresol was characterized by Michaelis-Menten kinetics in liver cytosols (Km = 14.8 ± 3.4 μM, Vmax = 1.5 ± 0.2 nmol/mg/min) and substrate inhibition in kidney cytosols (Km = 0.29 ± 0.02 μM, Vmax = 0.19 ± 0.05 nmol/mg/min, Ksi = 911.7 ± 278.4 μM). Of the 14 investigated therapeutic inhibitors, mefenamic acid (Ki = 2.4 ± 0.1 nM [liver], Ki = 1.2 ± 0.3 nM [kidney]) was the most potent in reducing the formation of p-cresol sulfate, exhibiting noncompetitive inhibition in human liver cytosols and recombinant SULT1A1, and mixed inhibition in human kidney cytosols. Our novel findings indicated that SULT1A1 contributed an important role in p-cresol sulfonation (hence it can be considered a probe reaction) in liver and kidneys, and mefenamic acid may be utilized as a potential therapeutic agent to attenuate the generation of p-cresol sulfate as an approach to detoxification.
Collapse
Affiliation(s)
- Yan Rong
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| | - Tony K L Kiang
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
26
|
Cupisti A, Bolasco P, D’Alessandro C, Giannese D, Sabatino A, Fiaccadori E. Protection of Residual Renal Function and Nutritional Treatment: First Step Strategy for Reduction of Uremic Toxins in End-Stage Kidney Disease Patients. Toxins (Basel) 2021; 13:toxins13040289. [PMID: 33921862 PMCID: PMC8073165 DOI: 10.3390/toxins13040289] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 02/06/2023] Open
Abstract
The retention of uremic toxins and their pathological effects occurs in the advanced phases of chronic kidney disease (CKD), mainly in stage 5, when the implementation of conventional thrice-weekly hemodialysis is the prevalent and life-saving treatment. However, the start of hemodialysis is associated with both an acceleration of the loss of residual kidney function (RKF) and the shift to an increased intake of proteins, which are precursors of uremic toxins. In this phase, hemodialysis treatment is the only way to remove toxins from the body, but it can be largely inefficient in the case of high molecular weight and/or protein-bound molecules. Instead, even very low levels of RKF are crucial for uremic toxins excretion, which in most cases are protein-derived waste products generated by the intestinal microbiota. Protection of RKF can be obtained even in patients with end-stage kidney disease (ESKD) by a gradual and soft shift to kidney replacement therapy (KRT), for example by combining a once-a-week hemodialysis program with a low or very low-protein diet on the extra-dialysis days. This approach could represent a tailored strategy aimed at limiting the retention of both inorganic and organic toxins. In this paper, we discuss the combination of upstream (i.e., reduced production) and downstream (i.e., increased removal) strategies to reduce the concentration of uremic toxins in patients with ESKD during the transition phase from pure conservative management to full hemodialysis treatment.
Collapse
Affiliation(s)
- Adamasco Cupisti
- Department of Clinical and Experimental Medicine, University of Pisa, 56121 Pisa, Italy; (C.D.); (D.G.)
- “Conservative Treatment of Chronic Kidney Disease” Project Group of the Italian Society of Nephrology, 00185 Rome, Italy;
- Correspondence:
| | - Piergiorgio Bolasco
- “Conservative Treatment of Chronic Kidney Disease” Project Group of the Italian Society of Nephrology, 00185 Rome, Italy;
| | - Claudia D’Alessandro
- Department of Clinical and Experimental Medicine, University of Pisa, 56121 Pisa, Italy; (C.D.); (D.G.)
- “Conservative Treatment of Chronic Kidney Disease” Project Group of the Italian Society of Nephrology, 00185 Rome, Italy;
| | - Domenico Giannese
- Department of Clinical and Experimental Medicine, University of Pisa, 56121 Pisa, Italy; (C.D.); (D.G.)
| | - Alice Sabatino
- Department of Medicine and Surgery, University of Parma, Nephrology Unit, Parma University Hospital, 43121 Parma, Italy; (A.S.); (E.F.)
| | - Enrico Fiaccadori
- Department of Medicine and Surgery, University of Parma, Nephrology Unit, Parma University Hospital, 43121 Parma, Italy; (A.S.); (E.F.)
| |
Collapse
|