1
|
Arai S, Yasukawa M, Shibata S. Role of selenium in the pathophysiology of cardiorenal anaemia syndrome. ESC Heart Fail 2024. [PMID: 39223820 DOI: 10.1002/ehf2.14893] [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: 11/09/2023] [Revised: 05/02/2024] [Accepted: 05/20/2024] [Indexed: 09/04/2024] Open
Abstract
Chronic kidney disease (CKD) and cardiovascular disease (CVD) have multiple bidirectional mechanisms, and anaemia is one of the critical factors that are associated with the progression of the two disorders [referred to as cardiorenal anaemia syndrome (CRAS)]. Several lines of evidence indicate that CRAS confers a worse prognosis, suggesting the need to clarify the underlying pathophysiology. Among the micronutrients (trace elements) that are essential to humans, inadequate iron status has previously been implicated in the pathogenesis of CRAS; however, the roles of other trace elements remain unclear. Selenium critically regulates the function of selenoproteins, in which selenocysteine is present at the active centres. The human genome encodes 25 selenoproteins, and accumulating data indicate that they regulate diverse physiological processes, including cellular redox homeostasis, calcium flux, thyroid hormone activity and haematopoiesis, all of which directly or indirectly influence cardiac function. The essential role of selenium in human health is underscored by the fact that its deficiency results in multiple disorders, among which are cardiomyopathy and abnormal erythrocyte morphology. Studies have shown that selenium deficiency is not uncommon in CKD patients with poor nutritional status, suggesting that it may be an under-recognized cause of anaemia and cardiovascular disorders in these patients. In this review, we discuss the role of selenium in the pathophysiology of CKD, particularly in the context of the interconnection among CKD, cardiac dysfunction and anaemia. Given that selenium deficiency is associated with treatment-resistant anaemia and an increased risk of CVD, its role as a key modulator of CRAS merits future investigation.
Collapse
Affiliation(s)
- Shigeyuki Arai
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Minoru Yasukawa
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Shigeru Shibata
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| |
Collapse
|
2
|
Almeida PP, Brito ML, Thomasi B, Mafra D, Fouque D, Knauf C, Tavares-Gomes AL, Stockler-Pinto MB. Is the enteric nervous system a lost piece of the gut-kidney axis puzzle linked to chronic kidney disease? Life Sci 2024; 351:122793. [PMID: 38848938 DOI: 10.1016/j.lfs.2024.122793] [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: 02/12/2024] [Revised: 05/20/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
The enteric nervous system (ENS) regulates numerous functional and immunological attributes of the gastrointestinal tract. Alterations in ENS cell function have been linked to intestinal outcomes in various metabolic, intestinal, and neurological disorders. Chronic kidney disease (CKD) is associated with a challenging intestinal environment due to gut dysbiosis, which further affects patient quality of life. Although the gut-related repercussions of CKD have been thoroughly investigated, the involvement of the ENS in this puzzle remains unclear. ENS cell dysfunction, such as glial reactivity and alterations in cholinergic signaling in the small intestine and colon, in CKD are associated with a wide range of intestinal pathways and responses in affected patients. This review discusses how the ENS is affected in CKD and how it is involved in gut-related outcomes, including intestinal permeability, inflammation, oxidative stress, and dysmotility.
Collapse
Affiliation(s)
| | - Michele Lima Brito
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | - Beatriz Thomasi
- Department of Physiology, Neuroscience Program, Michigan State University (MSU), East Lansing, MI, USA
| | - Denise Mafra
- Graduate Program in Biological Sciences - Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Denis Fouque
- Department of Nephrology, Centre Hopitalier Lyon Sud, INSERM 1060, CENS, Université de Lyon, France
| | - Claude Knauf
- INSERM U1220 Institut de Recherche en Santé Digestive, CHU Purpan, Université Toulouse III Paul Sabatier Toulouse, Toulouse, France
| | - Ana Lúcia Tavares-Gomes
- Neurosciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | - Milena Barcza Stockler-Pinto
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, Brazil; INSERM U1220 Institut de Recherche en Santé Digestive, CHU Purpan, Université Toulouse III Paul Sabatier Toulouse, Toulouse, France
| |
Collapse
|
3
|
Biruete A, Shin A, Kistler BM, Moe SM. Feeling gutted in chronic kidney disease (CKD): Gastrointestinal disorders and therapies to improve gastrointestinal health in individuals CKD, including those undergoing dialysis. Semin Dial 2024; 37:334-349. [PMID: 34708456 PMCID: PMC9043041 DOI: 10.1111/sdi.13030] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 09/21/2021] [Indexed: 12/15/2022]
Abstract
Chronic kidney disease (CKD) affects 9.1% of the population worldwide. CKD may lead to structural and functional gastrointestinal alterations, including impairment in the intestinal barrier, digestion and absorption of nutrients, motility, and changes to the gut microbiome. These changes can lead to increased gastrointestinal symptoms in people with CKD, even in early grades of kidney dysfunction. Gastrointestinal symptoms have been associated with lower quality of life and reduced nutritional status. Therefore, there has been considerable interest in improving gastrointestinal health in this clinical population. Gastrointestinal health can be influenced by lifestyle and medications, particularly in advanced grades of kidney dysfunction. Therapies focused on gastrointestinal health have been studied, including the use of probiotics, prebiotics, and synbiotics, yielding limited and conflicting results. This review summarizes the alterations in the gastrointestinal tract structure and function and provides an overview of potential nutritional interventions that kidney disease professionals can provide to improve gastrointestinal health in individuals with CKD.
Collapse
Affiliation(s)
- Annabel Biruete
- Department of Nutrition and Dietetics, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, USA
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Andrea Shin
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Brandon M. Kistler
- Department of Nutrition and Health Science, Ball State University, Muncie, Indiana, USA
| | - Sharon M. Moe
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Anatomy, Cell Biology, and Anatomy, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
4
|
Vandecruys M, De Smet S, De Beir J, Renier M, Leunis S, Van Criekinge H, Glorieux G, Raes J, Vanden Wyngaert K, Nagler E, Calders P, Monbaliu D, Cornelissen V, Evenepoel P, Van Craenenbroeck AH. Revitalizing the Gut Microbiome in Chronic Kidney Disease: A Comprehensive Exploration of the Therapeutic Potential of Physical Activity. Toxins (Basel) 2024; 16:242. [PMID: 38922137 PMCID: PMC11209503 DOI: 10.3390/toxins16060242] [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: 04/18/2024] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 06/27/2024] Open
Abstract
Both physical inactivity and disruptions in the gut microbiome appear to be prevalent in patients with chronic kidney disease (CKD). Engaging in physical activity could present a novel nonpharmacological strategy for enhancing the gut microbiome and mitigating the adverse effects associated with microbial dysbiosis in individuals with CKD. This narrative review explores the underlying mechanisms through which physical activity may favorably modulate microbial health, either through direct impact on the gut or through interorgan crosstalk. Also, the development of microbial dysbiosis and its interplay with physical inactivity in patients with CKD are discussed. Mechanisms and interventions through which physical activity may restore gut homeostasis in individuals with CKD are explored.
Collapse
Affiliation(s)
- Marieke Vandecruys
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium; (M.V.); or (P.E.)
| | - Stefan De Smet
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, 3000 Leuven, Belgium;
| | - Jasmine De Beir
- Department of Rehabilitation Sciences, Ghent University, 9000 Ghent, Belgium; (J.D.B.); (P.C.)
| | - Marie Renier
- Group Rehabilitation for Internal Disorders, Department of Rehabilitation Sciences, KU Leuven, 3000 Leuven, Belgium; (M.R.); (V.C.)
| | - Sofie Leunis
- Department of Microbiology, Immunology and Transplantation, Abdominal Transplantation, KU Leuven, 3000 Leuven, Belgium; (S.L.); (H.V.C.); (D.M.)
| | - Hanne Van Criekinge
- Department of Microbiology, Immunology and Transplantation, Abdominal Transplantation, KU Leuven, 3000 Leuven, Belgium; (S.L.); (H.V.C.); (D.M.)
| | - Griet Glorieux
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, 9000 Ghent, Belgium; (G.G.); (K.V.W.); (E.N.)
| | - Jeroen Raes
- Department of Microbiology and Immunology, Rega Institute for Medical Research, 3000 Leuven, Belgium;
- VIB-KU Leuven Center for Microbiology, 3000 Leuven, Belgium
| | - Karsten Vanden Wyngaert
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, 9000 Ghent, Belgium; (G.G.); (K.V.W.); (E.N.)
| | - Evi Nagler
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, 9000 Ghent, Belgium; (G.G.); (K.V.W.); (E.N.)
| | - Patrick Calders
- Department of Rehabilitation Sciences, Ghent University, 9000 Ghent, Belgium; (J.D.B.); (P.C.)
| | - Diethard Monbaliu
- Department of Microbiology, Immunology and Transplantation, Abdominal Transplantation, KU Leuven, 3000 Leuven, Belgium; (S.L.); (H.V.C.); (D.M.)
- Transplantoux Foundation, 3000 Leuven, Belgium
| | - Véronique Cornelissen
- Group Rehabilitation for Internal Disorders, Department of Rehabilitation Sciences, KU Leuven, 3000 Leuven, Belgium; (M.R.); (V.C.)
| | - Pieter Evenepoel
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium; (M.V.); or (P.E.)
- Department of Nephrology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Amaryllis H. Van Craenenbroeck
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium; (M.V.); or (P.E.)
- Department of Nephrology, University Hospitals Leuven, 3000 Leuven, Belgium
| |
Collapse
|
5
|
Fatani AMN, Suh JH, Auger J, Alabasi KM, Wang Y, Segal MS, Dahl WJ. Pea hull fiber supplementation does not modulate uremic metabolites in adults receiving hemodialysis: a randomized, double-blind, controlled trial. Front Nutr 2023; 10:1179295. [PMID: 37457968 PMCID: PMC10349378 DOI: 10.3389/fnut.2023.1179295] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/08/2023] [Indexed: 07/18/2023] Open
Abstract
Background Fiber is a potential therapeutic to suppress microbiota-generated uremic molecules. This study aimed to determine if fiber supplementation decreased serum levels of uremic molecules through the modulation of gut microbiota in adults undergoing hemodialysis. Methods A randomized, double-blinded, controlled crossover study was conducted. Following a 1-week baseline, participants consumed muffins with added pea hull fiber (PHF) (15 g/d) and control muffins daily, each for 4 weeks, separated by a 4-week washout. Blood and stool samples were collected per period. Serum p-cresyl sulfate (PCS), indoxyl sulfate (IS), phenylacetylglutamine (PAG), and trimethylamine N-oxide (TMAO) were quantified by LC-MS/MS, and fecal microbiota profiled by 16S rRNA gene amplicon sequencing and specific taxa of interest by qPCR. QIIME 2 sample-classifier was used to discover unique microbiota profiles due to the consumption of PHF. Results Intake of PHF contributed an additional 9 g/d of dietary fiber to the subjects' diet due to compliance. No significant changes from baseline were observed in serum PCS, IS, PAG, or TMAO, or for the relative quantification of Akkermansia muciniphila, Faecalibacterium prausnitzii, Bifidobacterium, or Roseburia, taxa considered health-enhancing. Dietary protein intake and IS (r = -0.5, p = 0.05) and slow transit stool form and PCS (r = 0.7, p < 0.01) were significantly correlated at baseline. PHF and control periods were not differentiated; however, using machine learning, taxa most distinguishing the microbiota composition during the PHF periods compared to usual diet alone were enriched Gemmiger, Collinsella, and depleted Lactobacillus, Ruminococcus, Coprococcus, and Mogibacteriaceae. Conclusion PHF supplementation did not mitigate serum levels of targeted microbial-generated uremic molecules. Given the high cellulose content, which may be resistant to fermentation, PHF may not exert sufficient effects on microbiota composition to modulate its activity at the dose consumed.
Collapse
Affiliation(s)
- Asmaa M. N. Fatani
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL, United States
- Food and Nutrition Department, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Joon Hyuk Suh
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL, United States
| | - Jérémie Auger
- Rosell Institute for Microbiome and Probiotics, Lallemand Health Solutions, Montréal, QC, Canada
| | - Karima M. Alabasi
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL, United States
- Foods and Nutrition Department, School of Health Science and Wellness, Northwest Missouri State University, Maryville, MO, United States
| | - Yu Wang
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL, United States
| | - Mark S. Segal
- Department of Nephrology, Hypertension and Renal Transplantation, College of Medicine, University of Florida, Gainesville, FL, United States
- North Florida South Georgia VHS, Gainesville, FL, United States
| | - Wendy J. Dahl
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL, United States
| |
Collapse
|
6
|
Abstract
Homeostasis is a prerequisite for health. When homeostasis becomes disrupted, dysfunction occurs. This is especially the case for the gut microbiota, which under normal conditions lives in symbiosis with the host. As there are as many microbial cells in and on our body as human cells, it is unlikely they would not contribute to health or disease. The gut bacterial metabolism generates numerous beneficial metabolites but also uremic toxins and their precursors, which are transported into the circulation. Barrier function in the intestine, the heart, and the kidneys regulates metabolite transport and concentration and plays a role in inter-organ and inter-organism communication via small molecules. This communication is analyzed from the perspective of the remote sensing and signaling theory, which emphasizes the role of a large network of multispecific, oligospecific, and monospecific transporters and enzymes in regulating small-molecule homeostasis. The theory provides a systems biology framework for understanding organ cross talk and microbe-host communication involving metabolites, signaling molecules, nutrients, antioxidants, and uremic toxins. This remote small-molecule communication is critical for maintenance of homeostasis along the gut-heart-kidney axis and for responding to homeostatic perturbations. Chronic kidney disease is characterized by gut dysbiosis and accumulation of toxic metabolites. This slowly impacts the body, affecting the cardiovascular system and contributing to the progression of kidney dysfunction, which in its turn influences the gut microbiota. Preserving gut homeostasis and barrier functions or restoring gut dysbiosis and dysfunction could be a minimally invasive way to improve patient outcomes and quality of life in many diseases, including cardiovascular and kidney disease.
Collapse
Affiliation(s)
- Griet Glorieux
- Nephrology Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Gent, Belgium (G.G., R.V., F.V.)
| | - Sanjay K Nigam
- Department of Pediatrics (S.K.N.), University of California San Diego, La Jolla, CA
- Division of Nephrology, Department of Medicine (S.K.N.), University of California San Diego, La Jolla, CA
| | - Raymond Vanholder
- Nephrology Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Gent, Belgium (G.G., R.V., F.V.)
| | - Francis Verbeke
- Nephrology Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Gent, Belgium (G.G., R.V., F.V.)
| |
Collapse
|
7
|
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: 28] [Impact Index Per Article: 28.0] [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
|
8
|
Teixeira RR, de Andrade LS, Pereira NBF, Montenegro H, Hoffmann C, Cuppari L. Gut microbiota profile of patients on peritoneal dialysis: comparison with household contacts. Eur J Clin Nutr 2023; 77:90-97. [PMID: 35906334 DOI: 10.1038/s41430-022-01190-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 07/12/2022] [Accepted: 07/18/2022] [Indexed: 01/28/2023]
Abstract
BACKGROUND Differences in patients gut microbiota composition with the potential for dysbiosis have been associated with chronic kidney disease (CKD). However, factors other than the disease itself, such as diet and cohabitation, have not been evaluated when gut microbiota of CKD patients was compared with that of healthy controls. The aim of this study was to compare the gut microbiota composition between patients on peritoneal dialysis (PD) and age-matched household contacts with normal renal function. METHODS Fecal samples were collected from 20 patients [men: 70%; age: 53.5 years (48.2-66; median and interquartile range); length on PD: 14 months (5.2-43.5) and 20 controls. The region V4 of the 16S ribosomal RNA gene was PCR-amplified and sequenced on Illumina MiSeq platform. Dietary intake and diet quality were assessed by a 3-day food record and a diet quality index, respectively. RESULTS No difference was found between the gut microbiota composition of patients and controls, assessed by alpha and beta diversities (p > 0.05) and genera differential abundance (p > 0.05). The most abundant phyla in both groups were Firmicutes (PD = 45%; Control: 47%; p = 0.65) and Bacteroidetes (PD = 41%; Control: 45%; p = 0.17). The phylum Proteobacteria, known as a potential marker of gut dysbiosis, was not different in proportions between groups (p > 0.05). No difference was observed regarding diet quality and dietary intake of fiber, protein and other nutrients (p > 0.05). CONCLUSION Gut microbiota of patients on PD did not differ from household contacts. This result suggests that cohabitation and dietary intake might have outweighed the disease influence on gut microbiota composition of our PD patients.
Collapse
Affiliation(s)
| | | | | | | | - Christian Hoffmann
- Food Research Center, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Lilian Cuppari
- Nutrition Program, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.
- Division of Nephrology, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.
| |
Collapse
|
9
|
Microbial Tryptophan Metabolism Tunes Host Immunity, Metabolism, and Extraintestinal Disorders. Metabolites 2022; 12:metabo12090834. [PMID: 36144238 PMCID: PMC9505266 DOI: 10.3390/metabo12090834] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
The trillions of commensal microorganisms comprising the gut microbiota have received growing attention owing to their impact on host physiology. Recent advances in our understandings of the host–microbiota crosstalk support a pivotal role of microbiota-derived metabolites in various physiological processes, as they serve as messengers in the complex dialogue between commensals and host immune and endocrine cells. In this review, we highlight the importance of tryptophan-derived metabolites in host physiology, and summarize the recent findings on the role of tryptophan catabolites in preserving intestinal homeostasis and fine-tuning immune and metabolic responses. Furthermore, we discuss the latest evidence on the effects of microbial tryptophan catabolites, describe their mechanisms of action, and discuss how perturbations of microbial tryptophan metabolism may affect the course of intestinal and extraintestinal disorders, including inflammatory bowel diseases, metabolic disorders, chronic kidney diseases, and cardiovascular diseases.
Collapse
|
10
|
Almeida PP, de Moraes Thomasi BB, Menezes ÁC, Da Cruz BO, da Silva Costa N, Brito ML, D'Avila Pereira A, Castañon CR, Degani VAN, Magliano DC, Knauf C, Tavares-Gomes AL, Stockler-Pinto MB. 5/6 nephrectomy affects enteric glial cells and promotes impaired antioxidant defense in the colonic neuromuscular layer. Life Sci 2022; 298:120494. [PMID: 35339510 DOI: 10.1016/j.lfs.2022.120494] [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: 12/13/2021] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 10/18/2022]
Abstract
AIMS Chronic kidney disease (CKD) produces multiple repercussions in the gastrointestinal tract (GIT), such as alterations in motility, gut microbiota, intestinal permeability, and increased oxidative stress. However, despite enteric glial cells (EGC) having important neural and immune features in GIT physiology, their function in CKD remains unknown. The present study investigates colonic glial markers, inflammation, and antioxidant parameters in a CKD model. MAIN METHODS A 5/6 nephrectomized rat model was used to induce CKD in rats and Sham-operated animals as a control to suppress. Biochemical measures in plasma and neuromuscular layer such as glutathione peroxidase (GPx) and superoxide dismutase (SOD) activity were carried out. Kidney histopathology was evaluated. Colon morphology analysis and glial fibrillary acid protein (GFAP), connexin-43 (Cx43), nuclear factor-kappa B (NF-κB) p65, and GPx protein expression were performed. KEY FINDINGS The CKD group exhibited dilated tubules and tubulointerstitial fibrosis in the reminiscent kidney (p = 0.0002). CKD rats showed higher SOD activity (p = 0.004) in plasma, with no differences in neuromuscular layer (p = 0.9833). However, GPx activity was decreased in the CKD group in plasma (p = 0.013) and neuromuscular layer (p = 0.0338). Morphological analysis revealed alterations in colonic morphometry with inflammatory foci in the submucosal layer and neuromuscular layer straightness in CKD rats (p = 0.0291). In addition, GFAP, Cx43, NF-κBp65 protein expression were increased, and GPx decreased in the neuromuscular layer of the CKD group (p < 0.05). SIGNIFICANCE CKD animals present alterations in colonic cytoarchitecture and decreased layer thickness. Moreover, CKD affects the enteric glial network of the neuromuscular layer, associated with decreased antioxidant activity and inflammation.
Collapse
Affiliation(s)
- Patricia Pereira Almeida
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, Brazil.
| | | | - Ágatha Cristie Menezes
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | - Beatriz Oliveira Da Cruz
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | - Nathalia da Silva Costa
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | - Michele Lima Brito
- Nutrition Graduation, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | | | - Cecília Ribeiro Castañon
- Clinic and Animal Reproduction Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | | | - D'Angelo Carlo Magliano
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, Brazil; Morphology Department, Biomedical Institute, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | - Claude Knauf
- Institut de Recherche en Santé Digestive, Université Paul Sabatier (UPS), Toulouse, France
| | - Ana Lúcia Tavares-Gomes
- Neuroscience Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | - Milena Barcza Stockler-Pinto
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, Brazil; Nutrition Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| |
Collapse
|
11
|
Lin X, Liang W, Li L, Xiong Q, He S, Zhao J, Guo X, Xiang S, Zhang P, Wang H, Ying C, Yao Y, Zuo X. The Accumulation of Gut Microbiome-derived Indoxyl Sulfate and P-Cresyl Sulfate in Patients With End-stage Renal Disease. J Ren Nutr 2021; 32:578-586. [PMID: 34736844 DOI: 10.1053/j.jrn.2021.09.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/15/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Indoxyl sulfate (IS) and p-cresyl sulfate (pCS) are two important gut microbiota-generated protein-bound uremic toxins. The present study aims to explore the alterations of serum IS and pCS concentrations, their production, and daily removal in end-stage renal disease (ESRD). METHODS A case-controlled study was conducted based on 11 patients with ESRD and 11 healthy volunteers. The metabolic processes for IS and pCS were compared in these two groups, including gut microbiome, fecal indole and p-cresol, indole-producing bacteria and p-cresol-producing bacteria, serum total IS and pCS concentrations, and their daily removal by urine and spent dialyzate. RESULTS Compared with healthy controls, patients with ESRD exhibited higher relative abundance of the indole-producing bacteria Escherichia coli (P < .001) and Bacteroides fragilis (P = .010) and p-cresol-producing bacteria Bacteroides fragilis (P = .010) and Bacteroides caccae (P = .047). The predicted functional profiles of gut microbiome based on 16S rRNA gene PhyloChip analysis showed that the microbial tryptophan metabolism pathway (map00380, P = .0006) was significantly enriched in patients with ESRD. However, the fecal precursors indole (P = .332) and p-cresol concentrations (P = .699) were comparable between the two groups. The serum IS (P < .001) and pCS (P < .001) concentrations were far higher in patients with ESRD than those in healthy controls, whereas the daily total removal by urine and dialyzate was much lower for the former than that for the latter (P = .019 for IS, P = .016 for pCS). CONCLUSIONS The present study showed serious IS and pCS accumulation in patients with ESRD, with significant expansion of indole-producing bacteria and p-cresol-producing bacteria, upregulation of the bacterial tryptophan metabolism pathway, and greatly increased serum IS and pCS concentrations, whereas significant decline of daily IS and pCS removal.
Collapse
Affiliation(s)
- Xuechun Lin
- Department of Clinical Nutrition, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wangqun Liang
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Li Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qianqian Xiong
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuiqing He
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jing Zhao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaolei Guo
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Siyun Xiang
- Department of Nutrition, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Piwei Zhang
- Department of Clinical Nutrition, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hong Wang
- Wuhan Institute for Food and Cosmetic Control, Wuhan, Hubei, China
| | - Chenjiang Ying
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ying Yao
- Department of Clinical Nutrition, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Xuezhi Zuo
- Department of Clinical Nutrition, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW This review will discuss recent studies showing that patients with chronic wasting diseases suffer from a variety of small intestinal impairments which might negatively impact the colonic microbiota and overall well-being. New insights will be addressed as well as novel approaches to assess intestinal function. RECENT FINDINGS Small intestinal dysfunction can enhance the amount and alter the composition of undigested food reaching the colon. As a result of reduced protein digestion and absorption, a large amount of undigested protein might reach the colon promoting the presence of pathogenic colonic bacteria and a switch from bacterial fiber fermentation to protein fermentation. While microbial metabolites of fiber fermentation, such as short-chain fatty acids (SCFA), are mainly considered beneficial for overall health, metabolites of protein fermentation, i.e. ammonia, branched SCFAs, hydrogen sulfide, polyamines, phenols, and indoles, can exert beneficial or deleterious effects on overall health. Substantial advances have been made in the assessment of small intestinal dysfunction in chronic diseases, but studies investigating the connection to colonic microbial metabolism are needed. A promising new stable isotope approach can enable the measurement of metabolite production by the colonic microbiota. SUMMARY Several studies have been conducted to assess intestinal function in chronic diseases. Impairments in intestinal barrier function, sugar absorption, protein digestion, and absorption, as well as small intestinal bacterial overgrowth were observed and possibly might negatively impact colonic bacterial metabolism. We suggest that improving these perturbations will improve overall patient health.
Collapse
Affiliation(s)
- Sarah K Kirschner
- Center for Translational Research in Aging & Longevity, Department of Health and Kinesiology, Texas A&M University, College Station, Texas, USA
| | | | | |
Collapse
|
13
|
Mosterd CM, Kanbay M, van den Born BJH, van Raalte DH, Rampanelli E. Intestinal microbiota and diabetic kidney diseases: the Role of microbiota and derived metabolites inmodulation of renal inflammation and disease progression. Best Pract Res Clin Endocrinol Metab 2021; 35:101484. [PMID: 33546983 DOI: 10.1016/j.beem.2021.101484] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Diabetic kidney disease (DKD) represents a growing public health burden and is the leading cause of end-stage kidney diseases. In recent years, host-gut microbiota interactions have emerged as an integral part for host homeostasis. In the context of nephropathies, mounting evidence supports a bidirectional microbiota-kidney crosstalk, which becomes particularly manifest during progressive kidney dysfunction. Indeed, in chronic kidney disease (CKD), the "healthy" microbiota structure is disrupted and intestinal microbes produce large quantities of uremic solutes responsible for renal damage; on the other hand, the uremic state, fueled by reduced renal clearance, causes shifts in microbial metabolism and composition, hence creating a vicious cycle in which dysbiosis and renal dysfunction are progressively worsened. In this review, we will summarize the evidence from clinical/experimental studies concerning the occurrence of gut dysbiosis in diabetic and non-diabetic CKD, discuss the functional consequences of dysbiosis for CKD progression and debate putative therapeutic interventions targeting the intestinal microbiome.
Collapse
Affiliation(s)
- C M Mosterd
- Department of Internal and Vascular Medicine, Amsterdam UMC, Location VUmc, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands.
| | - M Kanbay
- Department of Medicine, Division of Nephrology, Koc University School of Medicine, Istanbul, Turkey
| | - B J H van den Born
- Department of Nephrology and Vascular Medicine, Amsterdam UMC, Location AMC, the Netherlands
| | - D H van Raalte
- Diabetes Center, Department of Internal Medicine, Amsterdam UMC, Location VUmc, Diabetes Center, Amsterdam, the Netherlands
| | - E Rampanelli
- Department of Experimental Vascular Medicine, Amsterdam UMC, Location AMC, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands.
| |
Collapse
|
14
|
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: 12] [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: 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
|
15
|
Graboski AL, Redinbo MR. Gut-Derived Protein-Bound Uremic Toxins. Toxins (Basel) 2020; 12:toxins12090590. [PMID: 32932981 PMCID: PMC7551879 DOI: 10.3390/toxins12090590] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/17/2020] [Accepted: 09/08/2020] [Indexed: 12/11/2022] Open
Abstract
Chronic kidney disease (CKD) afflicts more than 500 million people worldwide and is one of the fastest growing global causes of mortality. When glomerular filtration rate begins to fall, uremic toxins accumulate in the serum and significantly increase the risk of death from cardiovascular disease and other causes. Several of the most harmful uremic toxins are produced by the gut microbiota. Furthermore, many such toxins are protein-bound and are therefore recalcitrant to removal by dialysis. We review the derivation and pathological mechanisms of gut-derived, protein-bound uremic toxins (PBUTs). We further outline the emerging relationship between kidney disease and gut dysbiosis, including the bacterial taxa altered, the regulation of microbial uremic toxin-producing genes, and their downstream physiological and neurological consequences. Finally, we discuss gut-targeted therapeutic strategies employed to reduce PBUTs. We conclude that targeting the gut microbiota is a promising approach for the treatment of CKD by blocking the serum accumulation of PBUTs that cannot be eliminated by dialysis.
Collapse
Affiliation(s)
- Amanda L. Graboski
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599-7365, USA;
| | - Matthew R. Redinbo
- Departments of Chemistry, Biochemistry, Microbiology and Genomics, University of North Carolina, Chapel Hill, NC 27599-3290, USA
- Correspondence:
| |
Collapse
|
16
|
Ondrussek-Sekac M, Navas-Carrillo D, Orenes-Piñero E. Intestinal microbiota alterations in chronic kidney disease and the influence of dietary components. Crit Rev Food Sci Nutr 2020; 61:1490-1502. [PMID: 32393049 DOI: 10.1080/10408398.2020.1761771] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
In chronic kidney disease, as in many other diseases, dysbiosis of intestinal microbiota has been reported as a disturbance or imbalance of the normal microbiome content that could disrupt the symbiotic relationship between the host and associated microbes, a disruption that can result in diseases. The disruption of gut barrier function allows the translocation of endotoxins and bacterial metabolites to the organism, thus contributing to uremic toxicity, inflammation and progression of chronic kidney disease. Increased intake of some nutrients and different nutritional strategies have been proposed to modulate gut microbiota, thus offering the opportunity for therapeutic interventions modifying the diet, decreasing uremic toxins production, increasing toxin excretion and finally modifying the normal microbiome content. The use of probiotics, prebiotics and low protein diets, among other approaches, could also improve this imbalance and/or decrease permeability of the intestinal barrier. In this review, the link between nutrients, microbiota and uremic toxins with chronic kidney disease progression has been studied thoroughly. Furthermore, this review outlines potential mechanisms of action and efficacy of probiotics, prebiotics and low protein diets as a new chronic kidney disease management tool.
Collapse
Affiliation(s)
- Mateo Ondrussek-Sekac
- Department of Biochemistry and Molecular Biology-A, University of Murcia, Murcia, Spain
| | | | - Esteban Orenes-Piñero
- Department of Biochemistry and Molecular Biology-A, University of Murcia, Murcia, Spain
| |
Collapse
|
17
|
Effect of probiotics on the intestinal microbiota of hemodialysis patients: a randomized trial. Eur J Nutr 2020; 59:3755-3766. [PMID: 32112136 DOI: 10.1007/s00394-020-02207-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 02/12/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Intestinal dysbiosis contributes to the progression of renal failure and cardiovascular diseases in patients with chronic kidney disease. Probiotics is a promising intervention to improving intestinal dysbiosis. A double-blind clinical trial to investigate the ability of probiotics to modulate gut microbiota compositions in patients receiving hemodialysis (HD) was undertaken. METHODS Fifty HD patients were enrolled and randomized, receiving either probiotics or placebo for 6 months. The responses to the interventions on gut microbiome, serum and fecal metabolome, serum albumin and endotoxin, endothelial activation markers and inflammatory markers were assessed. RESULTS Totally, 22 in the probiotics group (11 males; 14 non-diabetic) and 23 in the placebo group (13 males; 17 non-diabetic) completed the study. Compared to that in the placebo group, probiotics did not significantly alter species diversity of the fecal microbiome. Probiotics did, however, restore the community composition, with particular significance in non-diabetic HD patients (P = 0.007 by Adonis analysis). Specifically, according to the results of linear discriminate analysis effect size, probiotics raised the proportions of family Bacteroidaceae and Enterococcaceae, and reduced Ruminococcaceae, Halomonadaceae, Peptostreptococcaceae, Clostridiales Family XIII. Incertae Sedis and Erysipelotrichaceae in non-diabetic HD patients. Additionally, probiotics reduced the abundances of several uremic retention solutes in serum or feces, including indole-3-acetic acid-O-glucuronide, 3-guanidinopropionic acid, and 1-methylinosine (P < 0.05). In the probiotic arm, no significant changes were observed in other secondary outcomes. CONCLUSIONS Taken together, outcomes from this study suggest that probiotics do have benefits on improving intestinal imbalances and lowering exposure to several uremic toxins in HD patients.
Collapse
|
18
|
Gryp T, De Paepe K, Vanholder R, Kerckhof FM, Van Biesen W, Van de Wiele T, Verbeke F, Speeckaert M, Joossens M, Couttenye MM, Vaneechoutte M, Glorieux G. Gut microbiota generation of protein-bound uremic toxins and related metabolites is not altered at different stages of chronic kidney disease. Kidney Int 2020; 97:1230-1242. [PMID: 32317112 DOI: 10.1016/j.kint.2020.01.028] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/17/2020] [Accepted: 01/24/2020] [Indexed: 01/06/2023]
Abstract
Chronic kidney disease (CKD) is characterized by accumulation of protein-bound uremic toxins such as p-cresyl sulfate, p-cresyl glucuronide, indoxyl sulfate and indole-3-acetic acid, which originate in the gut. Intestinal bacteria metabolize aromatic amino acids into p-cresol and indole, (further conjugated in the colon mucosa and liver) and indole-3-acetic acid. Here we measured fecal, plasma and urine metabolite concentrations; the contribution of gut bacterial generation to plasma protein-bound uremic toxins accumulation; and influx into the gut of circulating protein-bound uremic toxins at different stages of CKD. Feces, blood and urine were collected from 14 control individuals and 141 patients with CKD. Solutes were quantified by ultra-high performance liquid chromatography. To assess the rate of bacterial generation of p-cresol, indole and indole-3-acetic acid, fecal samples were cultured ex vivo. With CKD progression, an increase in protein-bound uremic toxins levels was observed in plasma, whereas the levels of these toxins and their precursors remained the same in feces and urine. Anaerobic culture of fecal samples showed no difference in ex vivo p-cresol, indole and indole-3-acetic acid generation. Therefore, differences in plasma protein-bound uremic toxins levels between different CKD stages cannot be explained by differences in bacterial generation rates in the gut, suggesting retention due to impaired kidney function as the main contributor to their increased plasma levels. Thus, as fractional clearance decreased with the progression of CKD, tubular clearance appeared to be more affected than the glomerular filtration rate, and there was no net increase in protein-bound uremic toxins influx into the gut lumen with increased plasma levels.
Collapse
Affiliation(s)
- Tessa Gryp
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium; Department of Diagnostic Sciences, Laboratory Bacteriology Research, Ghent University, Ghent, Belgium; Department of Microbiology, Immunology and Transplantation, Molecular Microbiology-Microbiome Research Lab, KU Leuven, Leuven, Belgium.
| | - Kim De Paepe
- Department of Biotechnology, Center for Microbial Ecology and Technology, Ghent University, Ghent, Belgium
| | - Raymond Vanholder
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Frederiek-Maarten Kerckhof
- Department of Biotechnology, Center for Microbial Ecology and Technology, Ghent University, Ghent, Belgium
| | - Wim Van Biesen
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Tom Van de Wiele
- Department of Biotechnology, Center for Microbial Ecology and Technology, Ghent University, Ghent, Belgium
| | - Francis Verbeke
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Marijn Speeckaert
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Marie Joossens
- Department of Microbiology, Immunology and Transplantation, Molecular Microbiology-Microbiome Research Lab, KU Leuven, Leuven, Belgium
| | | | - Mario Vaneechoutte
- Department of Diagnostic Sciences, Laboratory Bacteriology Research, Ghent University, Ghent, Belgium
| | - Griet Glorieux
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
19
|
Costa-Moreira P, Vilas-Boas F, Teixeira Fraga A, Macedo G. Particular aspects of gastroenterological disorders in chronic kidney disease and end-stage renal disease patients: a clinically focused review. Scand J Gastroenterol 2020; 55:129-138. [PMID: 32027522 DOI: 10.1080/00365521.2020.1722217] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Besides renal disease, gastrointestinal (GI) disorders are frequently reported in patients with chronic kidney disease (CKD) and end-stage renal disease (ESRD). Related gastrointestinal symptoms tend to increase as the renal disease progresses. Also, in patients with ESRD, the modality of dialysis is related to particular forms of GI disorders.The kidney can interact with the digestive organs through functional endogenous systems such as the 'kidney-colon axis' and the 'kidney-liver axis'. Digestive diseases are one of the visible manifestations of the disturbance between hemostatic, hemodynamic and immunological balance in such patients.No clear management guidelines currently exist for many of the gastrointestinal problems that accompany renal failure. This review aims to describe the particular aspects of GI diseases present in CKD/ESRD. We focus our discussion in the specificities of epidemiology, diagnosis, and prognosis of such disorders between the different segments of the digestive system.
Collapse
Affiliation(s)
- Pedro Costa-Moreira
- Gastroenterology Department, Centro Hospitalar e Universitário São João, Porto, Portugal.,Faculty of Medicine, Medicine Department, University of Porto, Portugal
| | - Filipe Vilas-Boas
- Gastroenterology Department, Centro Hospitalar e Universitário São João, Porto, Portugal.,Faculty of Medicine, Medicine Department, University of Porto, Portugal
| | | | - Guilherme Macedo
- Gastroenterology Department, Centro Hospitalar e Universitário São João, Porto, Portugal.,Faculty of Medicine, Medicine Department, University of Porto, Portugal
| |
Collapse
|
20
|
Rahhal MN, Gharaibeh NE, Rahimi L, Ismail-Beigi F. Disturbances in Insulin-Glucose Metabolism in Patients With Advanced Renal Disease With and Without Diabetes. J Clin Endocrinol Metab 2019; 104:4949-4966. [PMID: 31162534 DOI: 10.1210/jc.2019-00286] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 05/29/2019] [Indexed: 02/03/2023]
Abstract
CONTEXT Use of insulin in patients with diabetes and advanced chronic kidney disease (CKD; stages 4 to 5) is challenging and shows great variability among individuals. We explored the mechanisms underlying this variability. EVIDENCE ACQUISITION PubMed was searched for articles in English from 1960 to 2018 for advanced CKD and diabetes, glucose and insulin metabolism, insulin clearance, secretion and resistance, plasma insulin concentration, glycemic control, hypoglycemia, insulin dosage, and continuous glucose monitoring (CGM) in CKD. EVIDENCE SYNTHESIS The evidence shows that in most patients the daily dose of insulin needs to be significantly reduced with a high degree of variability; in some the dose remains unchanged, and rarely it is increased. The premise that the marked reduction in insulin requirement is essentially attributable to decreased insulin clearance by kidneys leading to prolongation of its plasma half-life, elevated blood insulin concentration, and hypoglycemia is not entirely correct. Other factors including decreases in food intake, insulin secretion, insulin clearance by peripheral tissues, and renal gluconeogenesis play important roles. There is also heightened resistance to insulin due to metabolic acidosis, uremic toxins, inflammatory state, and vitamin D deficiency. Importantly, the magnitude of changes in each of these factors varies between individuals with the same degree of CKD. CONCLUSIONS In the presence of diabetes with advanced CKD, the insulin regimen should be individualized based on knowledge of the daily glucose patterns. The use of CGM is promising for safer glycemic control in patients with advanced CKD and diabetes and helps prevent extremes of hypoglycemia and hyperglycemia.
Collapse
Affiliation(s)
- Marie-Noel Rahhal
- Department of Medicine, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Naser Eddin Gharaibeh
- Department of Medicine, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Leili Rahimi
- Department of Medicine, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Faramarz Ismail-Beigi
- Department of Medicine, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| |
Collapse
|
21
|
Arshad Z, Rezapour-Firouzi S, Ebrahimifar M, Mosavi Jarrahi A, Mohammadian M. Association of Delta-6-Desaturase Expression with
Aggressiveness of Cancer, Diabetes Mellitus, and Multiple
Sclerosis: A Narrative Review. Asian Pac J Cancer Prev 2019; 20:1005-1018. [PMID: 31030467 PMCID: PMC6948902 DOI: 10.31557/apjcp.2019.20.4.1005] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background: The phosphatidylinositol 3-kinase/ protein kinase B /mammalian target of rapamycin (PI3K/Akt/
mTOR) signaling regulates multiple cellular processes and organizes cell proliferation, survival, and differentiation
with the available nutrients, in particular, fatty acids. Polyunsaturated fatty acids (PUFAs) are cytotoxic to cancer cells
and play a critical role in the treatment of multiple sclerosis (MS) and diabetes mellitus (DM). PUFAs are produced in
the body by desaturases and elongases from dietary essential fatty acids (EFAs), primarily involving delta-6-desaturase
(D6D). D6D is a rate-limiting enzyme for maintaining many aspects of lipid homeostasis and normal health. D6D is
important to recognize the mechanisms that regulate the expression of this enzyme in humans. A lower level of D6D was
seen in breast tumors compared to normal tissues. Interestingly, the elevated serum level of D6D was seen in MS and
DM, which explains the critical role of D6D in inflammatory diseases. Methods: We searched databases of PubMed,
Web of Science (WOS), Google Scholar, Scopus and related studies by predefined eligibility criteria. We assessed
their quality and extracted data. Results: Regarding the mTOR signaling pathway, there is remarkable contributions of
many inflammatory diseases to attention to common metabolic pathways are depicted. Of course, we need to have the
insights into each disorder and their pathological process. The first step in balancing the intake of EFAs is to prevent
the disruption of metabolism and expression of the D6D enzyme. Conclusions: The ω6 and ω3 pathways are two major
pathways in the biosynthesis of PUFAs. In both of these, D6D is a vital bifunctional enzyme desaturating linoleic acid
or alpha-linolenic acid. Therefore, if ω6 and ω3 EFAs are given together in a ratio of 2: 1, the D6D expression will be
down-regulated and normalized.
Collapse
Affiliation(s)
- Zhila Arshad
- Department of Pathology of Anatomy, School of medicine, Baku University of Medical Sciences, Baku, Azerbaijan
| | - Soheila Rezapour-Firouzi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran. ,
| | - Meysam Ebrahimifar
- Department of Toxicology, Faculty of Pharmacy, Islamic Azad University, Shahreza Branch, Shahreza, Iran
| | - Alireza Mosavi Jarrahi
- Department of Social Medicine, Medical School, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahshid Mohammadian
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
22
|
Fernandes R, Viana SD, Nunes S, Reis F. Diabetic gut microbiota dysbiosis as an inflammaging and immunosenescence condition that fosters progression of retinopathy and nephropathy. Biochim Biophys Acta Mol Basis Dis 2018; 1865:1876-1897. [PMID: 30287404 DOI: 10.1016/j.bbadis.2018.09.032] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/18/2018] [Accepted: 09/24/2018] [Indexed: 02/07/2023]
Abstract
The increased prevalence of type 2 diabetes mellitus (T2DM) and life expectancy of diabetic patients fosters the worldwide prevalence of retinopathy and nephropathy, two major microvascular complications that have been difficult to treat with contemporary glucose-lowering medications. The gut microbiota (GM) has become a lively field research in the last years; there is a growing recognition that altered intestinal microbiota composition and function can directly impact the phenomenon of ageing and age-related disorders. In fact, human GM, envisaged as a potential source of novel therapeutics, strongly modulates host immunity and metabolism. It is now clear that gut dysbiosis and their products (e.g. p-cresyl sulfate, trimethylamine‑N‑oxide) dictate a secretory associated senescence phenotype and chronic low-grade inflammation, features shared in the physiological process of ageing ("inflammaging") as well as in T2DM ("metaflammation") and in its microvascular complications. This review provides an in-depth look on the crosstalk between GM, host immunity and metabolism. Further, it characterizes human GM signatures of elderly and T2DM patients. Finally, a comprehensive scrutiny of recent molecular findings (e.g. epigenetic changes) underlying causal relationships between GM dysbiosis and diabetic retinopathy/nephropathy complications is pinpointed, with the ultimate goal to unravel potential pathophysiological mechanisms that may be explored, in a near future, as personalized disease-modifying therapeutic approaches.
Collapse
Affiliation(s)
- Rosa Fernandes
- Institute of Pharmacology & Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, CNC.IBILI Consortium & CIBB Consortium, University of Coimbra, Coimbra, Portugal
| | - Sofia D Viana
- Institute of Pharmacology & Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, CNC.IBILI Consortium & CIBB Consortium, University of Coimbra, Coimbra, Portugal; Polytechnic Institute of Coimbra, ESTESC-Coimbra Health School, Pharmacy, Coimbra, Portugal
| | - Sara Nunes
- Institute of Pharmacology & Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, CNC.IBILI Consortium & CIBB Consortium, University of Coimbra, Coimbra, Portugal
| | - Flávio Reis
- Institute of Pharmacology & Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, CNC.IBILI Consortium & CIBB Consortium, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
23
|
de Andrade LS, Ramos CI, Cuppari L. The cross-talk between the kidney and the gut: implications for chronic kidney disease. ACTA ACUST UNITED AC 2017. [DOI: 10.1186/s41110-017-0054-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
24
|
Mishima E, Fukuda S, Kanemitsu Y, Saigusa D, Mukawa C, Asaji K, Matsumoto Y, Tsukamoto H, Tachikawa T, Tsukimi T, Fukuda NN, Ho HJ, Kikuchi K, Suzuki C, Nanto F, Suzuki T, Ito S, Soga T, Tomioka Y, Abe T. Canagliflozin reduces plasma uremic toxins and alters the intestinal microbiota composition in a chronic kidney disease mouse model. Am J Physiol Renal Physiol 2017; 315:F824-F833. [PMID: 29167170 DOI: 10.1152/ajprenal.00314.2017] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Accumulation of uremic toxins, which exert deleterious effects in chronic kidney disease, is influenced by the intestinal environment; the microbiota contributes to the production of representative uremic toxins, including p-cresyl sulfate and indoxyl sulfate. Canagliflozin is a sodium-glucose cotransporter (SGLT) 2 inhibitor, and it also exerts a modest inhibitory effect on SGLT1. The inhibition of intestinal SGLT1 can influence the gastrointestinal environment. We examined the effect of canagliflozin on the accumulation of uremic toxins in chronic kidney disease using adenine-induced renal failure mice. Two-week canagliflozin (10 mg/kg po) treatment did not influence the impaired renal function; however, it significantly reduced the plasma levels of p-cresyl sulfate and indoxyl sulfate in renal failure mice (a 75% and 26% reduction, respectively, compared with the vehicle group). Additionally, canagliflozin significantly increased cecal short-chain fatty acids in the mice, suggesting the promotion of bacterial carbohydrate fermentation in the intestine. Analysis of the cecal microbiota showed that canagliflozin significantly altered microbiota composition in the renal failure mice. These results indicate that canagliflozin exerts intestinal effects that reduce the accumulation of uremic toxins including p-cresyl sulfate. Reduction of accumulated uremic toxins by canagliflozin could provide a potential therapeutic option in chronic kidney disease.
Collapse
Affiliation(s)
- Eikan Mishima
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan.,Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology , Kawasaki , Japan.,Transborder Medical Research Center, University of Tsukuba , Tsukuba , Japan.,PRESTO, Japan Science and Technology Agency, Kawaguchi, Japan
| | - Yoshitomi Kanemitsu
- Laboratory of Oncology, Pharmacy Practice and Sciences, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Daisuke Saigusa
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University , Sendai , Japan
| | - Chikahisa Mukawa
- Laboratory of Oncology, Pharmacy Practice and Sciences, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Kei Asaji
- Laboratory of Oncology, Pharmacy Practice and Sciences, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Yotaro Matsumoto
- Laboratory of Oncology, Pharmacy Practice and Sciences, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Hiroki Tsukamoto
- Laboratory of Oncology, Pharmacy Practice and Sciences, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Tatsuki Tachikawa
- Laboratory of Oncology, Pharmacy Practice and Sciences, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Tomoya Tsukimi
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Noriko N Fukuda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Hsin-Jung Ho
- Department of Medical Science, Tohoku University Graduate School of Biomedical Engineering , Sendai , Japan
| | - Koichi Kikuchi
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Chitose Suzuki
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Fumika Nanto
- Department of Medical Science, Tohoku University Graduate School of Biomedical Engineering , Sendai , Japan
| | - Takehiro Suzuki
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Sadayoshi Ito
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Yoshihisa Tomioka
- Laboratory of Oncology, Pharmacy Practice and Sciences, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Takaaki Abe
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine , Sendai , Japan.,Department of Medical Science, Tohoku University Graduate School of Biomedical Engineering , Sendai , Japan.,Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine , Sendai , Japan
| |
Collapse
|
25
|
DALLAS DAVIDC, SANCTUARY MEGANR, QU YUNYAO, KHAJAVI SHABNAMHAGHIGHAT, VAN ZANDT ALEXANDRIAE, DYANDRA MELISSA, FRESE STEVENA, BARILE DANIELA, GERMAN JBRUCE. Personalizing protein nourishment. Crit Rev Food Sci Nutr 2017; 57:3313-3331. [PMID: 26713355 PMCID: PMC4927412 DOI: 10.1080/10408398.2015.1117412] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Proteins are not equally digestible-their proteolytic susceptibility varies by their source and processing method. Incomplete digestion increases colonic microbial protein fermentation (putrefaction), which produces toxic metabolites that can induce inflammation in vitro and have been associated with inflammation in vivo. Individual humans differ in protein digestive capacity based on phenotypes, particularly disease states. To avoid putrefaction-induced intestinal inflammation, protein sources, and processing methods must be tailored to the consumer's digestive capacity. This review explores how food processing techniques alter protein digestibility and examines how physiological conditions alter digestive capacity. Possible solutions to improving digestive function or matching low digestive capacity with more digestible protein sources are explored. Beyond the ileal digestibility measurements of protein digestibility, less invasive, quicker and cheaper techniques for monitoring the extent of protein digestion and fermentation are needed to personalize protein nourishment. Biomarkers of protein digestive capacity and efficiency can be identified with the toolsets of peptidomics, metabolomics, microbial sequencing and multiplexed protein analysis of fecal and urine samples. By monitoring individual protein digestive function, the protein component of diets can be tailored via protein source and processing selection to match individual needs to minimize colonic putrefaction and, thus, optimize gut health.
Collapse
Affiliation(s)
- DAVID C. DALLAS
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
- Foods for Health Institute, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| | - MEGAN R. SANCTUARY
- Foods for Health Institute, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
- Department of Nutrition, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| | - YUNYAO QU
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| | - SHABNAM HAGHIGHAT KHAJAVI
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
- Department of Food Science and Technology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - ALEXANDRIA E. VAN ZANDT
- Department of Nutrition, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| | - MELISSA DYANDRA
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| | - STEVEN A. FRESE
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
- Foods for Health Institute, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| | - DANIELA BARILE
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
- Foods for Health Institute, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| | - J. BRUCE GERMAN
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
- Foods for Health Institute, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| |
Collapse
|
26
|
Liu WS, Chu DC, Chan HL, Li SY, Liu CK, Yang CY, Chen YW, Lee PC, Lai YT, Lin CC. Fixed dose of long-acting erythropoietic stimulating agents at higher frequency improves appetite, reduces inflammation and corrects anaemia in patients on haemodialysis. Clin Exp Pharmacol Physiol 2017; 43:875-82. [PMID: 27385380 DOI: 10.1111/1440-1681.12618] [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] [Received: 12/01/2015] [Revised: 06/14/2016] [Accepted: 07/03/2016] [Indexed: 12/21/2022]
Abstract
Anaemia is an important issue in patients undergoing haemodialysis. We aimed to identify a better dosing schedule of a fixed monthly dose of continuous erythropoietin receptor activator (CERA) in patients with chronic kidney disease (CKD) on haemodialysis. The CERA dosing schedule included 100 μg once monthly for 2 months, 50 μg twice monthly for 2 months and then 100 μg once monthly for two months. The effectiveness was determined by comparing haematocrit, nutritional status (serum protein and albumin) and inflammatory markers (tumour necrosis factor (TNF)-α, interleukin (IL)-1, IL-6 and Hepcidin) at the beginning of the study with those at the end of the study. Forty-seven out of 67 patients completed the trial. At the end, haematocrit was significantly higher (34.51 vs 33.22%, P=.004), levels of inflammatory markers were significantly lower (TNF-α (30.71 vs 35.67 ng/mL, P=.007), IL-6 (5.12 vs 7.95 ng/mL, P=.033), hepcidin (60.39 vs 74.39 ng/mL, P=.002)), blood glucose levels were significantly lower (112.40 vs 139.02 mg/dL, P=.003) and albumin was significantly higher (4.11 vs 3.98, P=.001). Patients with a better than average response had a lower initial number of red blood cells (3.3 vs 3.6 × 10(6) /mm(3) , P=.025) and a lower IL-1 (3.8 vs 12.9 ng/mL, P=.01). They also had significantly lower blood glucose levels at the end. (91.3 vs 124.0 mg/dL, P=.03). We demonstrate that a fixed monthly dose of CERA at a twice monthly dosing schedule improves nutrition, reduces the inflammation and corrects anaemia in patients on haemodialysis. This finding may provide a new strategy for treating CKD-related anaemia.
Collapse
Affiliation(s)
- Wen-Sheng Liu
- Division of Nephrology, Department of Medicine, Taipei City Hospital, Zhong-Xing Branch, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Environmental and Occupational Health Sciences, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,College of Science and Engineering, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Da-Chen Chu
- Institute of Public Health and Community Medicine Research Centre, National Yang-Ming University, Taipei, Taiwan.,Department of Health Care Management, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan.,Department of Neurosurgery, Taipei City Hospital, Taipei, Taiwan
| | - Hsiang-Lin Chan
- Department of Child Psychiatry, Chang Gung Memorial Hospital and University, Taoyuan, Taiwan
| | - Szu-Yuan Li
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Nephrology, and Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chih-Kuang Liu
- College of Medicine & Graduate Institute of Business Administration, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chih-Yu Yang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Nephrology, and Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yu-Wei Chen
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Nephrology, and Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Pui-Ching Lee
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Nephrology, and Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yen-Ting Lai
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan.,Department of Nursing, Yuanpei University, Hsinchu, Taiwan
| | - Chih-Ching Lin
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Nephrology, and Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
27
|
Sabatino A, Regolisti G, Cosola C, Gesualdo L, Fiaccadori E. Intestinal Microbiota in Type 2 Diabetes and Chronic Kidney Disease. Curr Diab Rep 2017; 17:16. [PMID: 28271466 DOI: 10.1007/s11892-017-0841-z] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF THE REVIEW Diabetes mellitus is a major cause of kidney disease [chronic kidney disease (CKD) and end-stage renal disease (ESRD)] and are both characterized by an increased risk of cardiovascular events. Diabetes and kidney disease are also commonly associated with a chronic inflammatory state, which is now considered a non-traditional risk factor for atherosclerosis. In the case of type 2 diabetes mellitus (T2DM), inflammation is mainly a consequence of visceral obesity, while in the case of CKD or ESRD patients on dialysis, inflammation is caused by multiple factors, classically grouped as dialysis-related and non-dialysis-related. More recently, a key role has been credited to the intestinal microbiota in the pathogenesis of chronic inflammation present in both disease states. While many recent data on the intestinal microbiota and its relationship to chronic inflammation are available for CKD patients, very little is known regarding T2DM and patients with diabetic nephropathy. The aim of this review is to summarize and discuss the main pathophysiological issues of intestinal microbiota in patients with T2DM and CKD/ESRD. RECENT FINDINGS The presence of intestinal dysbiosis, along with increased intestinal permeability and high circulating levels of lipopolysaccharides, a condition known as "endotoxemia," characterize T2DM, CKD, and ESRD on dialysis. The hallmark of intestinal dysbiosis is a reduction of saccharolytic microbes mainly producing short-chain fatty acids (SCFA) and, in the case of CKD/ESRD, an increase in proteolytic microbes that produce different substances possibly related to uremic toxicity. Dysbiosis is associated with endotoxemia and chronic inflammation, with disruption of the intestinal barrier and depletion of beneficial bacteria producing SCFAs. T2DM and CKD/ESRD, whose coexistence is increasingly found in clinical practice, share similar negative effects on both intestinal microbiota and function. More studies are needed to characterize specific alterations of the intestinal microbiota in diabetic nephropathy and to assess possible effects of probiotic and prebiotic treatments in this setting.
Collapse
Affiliation(s)
- Alice Sabatino
- Unità di Fisiopatologia dell'Insufficienza Renale Acuta e Cronica, Università degli Studi di Parma, Parma, Italy
| | - Giuseppe Regolisti
- Unità di Fisiopatologia dell'Insufficienza Renale Acuta e Cronica, Università degli Studi di Parma, Parma, Italy
| | - Carmela Cosola
- Dipartimento dell'Emergenza e dei Trapianti di Organi-Sezione di Nefrologia, Dialisi e Trapianti, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Loreto Gesualdo
- Dipartimento dell'Emergenza e dei Trapianti di Organi-Sezione di Nefrologia, Dialisi e Trapianti, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Enrico Fiaccadori
- Unità di Fisiopatologia dell'Insufficienza Renale Acuta e Cronica, Università degli Studi di Parma, Parma, Italy.
| |
Collapse
|
28
|
Grant CJ, Harrison LE, Hoad CL, Marciani L, Gowland PA, McIntyre CW. Patients with chronic kidney disease have abnormal upper gastro-intestinal tract digestive function: A study of uremic enteropathy. J Gastroenterol Hepatol 2017; 32:372-377. [PMID: 27222079 DOI: 10.1111/jgh.13458] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/19/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND AIM Chronic kidney disease (CKD) affects gastrointestinal (GI) function and results in numerous adaptive and maladaptive responses. Disruption of the colonic microbiome and its attendant consequences-the loss of gut barrier integrity and increased generation of uremic toxins-has become well-recognized. However, less attention has been paid to characterizing the mechanisms behind dysfunction of the upper GI tract, largely owing to the difficulty of studying small bowel function in vivo. This present study was designed to comprehensively describe upper GI function in those with advanced renal impairment. METHODS Thirty-five non-diabetic subjects (12 CKD stage 4/5 patients, 23 healthy controls) underwent detailed GI magnetic resonance imaging (MRI) in both fasted and fed states. Upper GI function was assessed by quantification of gastric emptying and intra-luminal small bowel water. Characterization of hydration and cardiovascular status was performed at baseline. Gut barrier integrity was assessed using serum endotoxin level. RESULTS Chronic kidney disease was associated with dysmotility (gastric half-emptying time 96 ± 32 vs 74 ± 27 min, P = 0.04) and reduced fasting and post-prandial small bowel water (36 ± 22 mL vs 78 ± 42 mL, P < 0.001), reflecting abnormal digestive secretion, and absorption. This was related to the degree of endotoxemia (r = -0.60, P = 0.04) and poorer symptom scores, but not to disease severity, arterial stiffness or hydration status. CONCLUSION Chronic kidney disease adversely affects digestive function. Abnormalities in digestive secretion and absorption may potentially have a broad impact in the prevention and treatment of both CKD and its complications. Further study is required to assess the factors that contribute to this dysfunction in a wider CKD population.
Collapse
Affiliation(s)
- Claire J Grant
- The Lilibeth Caberto Kidney Clinical Research Unit, London, Ontario, Canada
| | | | - Caroline L Hoad
- Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, UK
| | - Luca Marciani
- Nottingham Digestive Diseases Centre, University of Nottingham, Nottingham, UK
| | - Penny A Gowland
- Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, UK
| | | |
Collapse
|
29
|
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: 243] [Impact Index Per Article: 34.7] [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
|
30
|
Afaghi E, Tayebi A, Ebadi A, Sobhani V, Einollahi B, Tayebi M. The Effect of BCAA and ISO-WHEY Oral Nutritional Supplements on Dialysis Adequacy. Nephrourol Mon 2016; 8:e34993. [PMID: 27896236 PMCID: PMC5120366 DOI: 10.5812/numonthly.34993] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 12/31/2015] [Indexed: 11/25/2022] Open
Abstract
Background Protein-energy malnutrition is a common problem in hemodialysis patients and has different outcomes such as reduced quality of life, longer hospitalization time, lower dialysis adequacy, and higher mortality rate. Investigation of dialysis adequacy is an important method for assessing hemodialysis patients, and improving the dialysis adequacy is an important healthcare team goal. Objectives The present study aims to investigate and compare the effects of BCAA and ISO-WHEY oral nutritional supplements on dialysis adequacy. Methods In a clinical trial study, 66 hemodialysis patients were randomly divided into three groups: Group A (n = 22), Group B (n = 22), and Group C or the control group (n = 22). In Groups A and B, as prescribed and controlled by nutritionists and nephrologists, respectively, ISO-WHEY and BCAA protein powder were used for 2 months on a daily basis. For all groups, before intervention and 1 and 2 months after intervention, the dialysis adequacy was measured using URR and Kt/V. Finally, the data were analyzed using IBM SPSS Statistics Base 21.0 software. Results Out of 66 patients, 61 (19 in Group A, 20 in Group B, and 22 in Group C) completed the study period, and before intervention, all groups were equal in terms of quality and quantity variables (P > 0.05). After intervention, there was a significant difference between the three groups with regard to the variables of dialysis adequacy based on Kt/V and URR to independent-t test and repeated measures ANOVA (P < 0.05). Conclusions Results show that the intake of oral nutritional supplements leads to an improvement in the dialysis adequacy of hemodialysis patients. Therefore, the use of nutritional supplements along with patients' training and regular consultation will be helpful in improving the nutritional status, dialysis adequacy, and eventually the quality of life.
Collapse
Affiliation(s)
- Effat Afaghi
- Nursing Faculty, AJA University of Medical Sciences, Tehran, IR Iran
| | - Ali Tayebi
- Nephrology and Urology Research Center, Nursing Faculty, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
- Corresponding author: Ali Tayebi, Nephrology and Urology Research Center, Nursing Faculty, Baqiyatallah University of Medical Sciences, Tehran, IR Iran. Tel: +98-2122289941, Fax: +98-2126127237, E-mail:
| | - Abbas Ebadi
- Behavioral Sciences Research Center, Nursing Faculty, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
| | - Vahid Sobhani
- Exercise Physiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
| | - Behzad Einollahi
- Nephrology and Urology Research Center, Nursing Faculty, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
| | - Mehdi Tayebi
- Faculty of Physical Education and Sport Sciences, Shahid Rajaee Teacher Training University, Tehran, IR Iran
| |
Collapse
|
31
|
McCue MD, Welch KC. (13)C-Breath testing in animals: theory, applications, and future directions. J Comp Physiol B 2015; 186:265-85. [PMID: 26660654 DOI: 10.1007/s00360-015-0950-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/11/2015] [Accepted: 11/25/2015] [Indexed: 10/22/2022]
Abstract
The carbon isotope values in the exhaled breath of an animal mirror the carbon isotope values of the metabolic fuels being oxidized. The measurement of stable carbon isotopes in carbon dioxide is called (13)C-breath testing and offers a minimally invasive method to study substrate oxidation in vivo. (13)C-breath testing has been broadly used to study human exercise, nutrition, and pathologies since the 1970s. Owing to reduced use of radioactive isotopes and the increased convenience and affordability of (13)C-analyzers, the past decade has witnessed a sharp increase in the use of breath testing throughout comparative physiology--especially to answer questions about how and when animals oxidize particular nutrients. Here, we review the practical aspects of (13)C-breath testing and identify the strengths and weaknesses of different methodological approaches including the use of natural abundance versus artificially-enriched (13)C tracers. We critically compare the information that can be obtained using different experimental protocols such as diet-switching versus fuel-switching. We also discuss several factors that should be considered when designing breath testing experiments including extrinsic versus intrinsic (13)C-labelling and different approaches to model nutrient oxidation. We use case studies to highlight the myriad applications of (13)C-breath testing in basic and clinical human studies as well as comparative studies of fuel use, energetics, and carbon turnover in multiple vertebrate and invertebrate groups. Lastly, we call for increased and rigorous use of (13)C-breath testing to explore a variety of new research areas and potentially answer long standing questions related to thermobiology, locomotion, and nutrition.
Collapse
|
32
|
Rossi M, Johnson DW, Campbell KL. The Kidney–Gut Axis: Implications for Nutrition Care. J Ren Nutr 2015; 25:399-403. [DOI: 10.1053/j.jrn.2015.01.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 12/30/2014] [Accepted: 01/05/2015] [Indexed: 02/08/2023] Open
|
33
|
Sabatino A, Regolisti G, Brusasco I, Cabassi A, Morabito S, Fiaccadori E. Alterations of intestinal barrier and microbiota in chronic kidney disease. Nephrol Dial Transplant 2014; 30:924-33. [PMID: 25190600 DOI: 10.1093/ndt/gfu287] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 08/04/2014] [Indexed: 12/27/2022] Open
Abstract
Recent studies have highlighted the close relationship between the kidney and the gastrointestinal (GI) tract--frequently referred to as the kidney--gut axis--in patients with chronic kidney disease (CKD). In this regard, two important pathophysiological concepts have evolved: (i) production and accumulation of toxic end-products derived from increased bacterial fermentation of protein and other nitrogen-containing substances in the GI tract, (ii) translocation of endotoxins and live bacteria from gut lumen into the bloodstream, due to damage of the intestinal epithelial barrier and quantitative/qualitative alterations of the intestinal microbiota associated with the uraemic milieu. In both cases, these gut-centred alterations may have relevant systemic consequences in CKD patients, since they are able to trigger chronic inflammation, increase cardiovascular risk and worsen uraemic toxicity. The present review is thus focused on the kidney-gut axis in CKD, with special attention to the alterations of the intestinal barrier and the local microbiota (i.e. the collection of microorganisms living in a symbiotic coexistence with their host in the intestinal lumen) and their relationships to inflammation and uraemic toxicity in CKD. Moreover, we will summarize the most important clinical data suggesting the potential for nutritional modulation of gut-related inflammation and intestinal production of noxious by-products contributing to uraemic toxicity in CKD patients.
Collapse
Affiliation(s)
- Alice Sabatino
- Department of Clinical and Experimental Medicine, Acute and Chronic Renal Failure Unit, Parma University Hospital, Parma, Italy
| | - Giuseppe Regolisti
- Department of Clinical and Experimental Medicine, Acute and Chronic Renal Failure Unit, Parma University Hospital, Parma, Italy
| | - Irene Brusasco
- Department of Clinical and Experimental Medicine, Acute and Chronic Renal Failure Unit, Parma University Hospital, Parma, Italy
| | - Aderville Cabassi
- Department of Clinical and Experimental Medicine, Acute and Chronic Renal Failure Unit, Parma University Hospital, Parma, Italy
| | - Santo Morabito
- Department of Clinical and Experimental Medicine, Acute and Chronic Renal Failure Unit, Parma University Hospital, Parma, Italy
| | - Enrico Fiaccadori
- Department of Clinical and Experimental Medicine, Acute and Chronic Renal Failure Unit, Parma University Hospital, Parma, Italy
| |
Collapse
|
34
|
Heritability and clinical determinants of serum indoxyl sulfate and p-cresyl sulfate, candidate biomarkers of the human microbiome enterotype. PLoS One 2014; 9:e79682. [PMID: 24850265 PMCID: PMC4029585 DOI: 10.1371/journal.pone.0079682] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 09/23/2013] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Indoxyl sulfate and p-cresyl sulfate are unique microbial co-metabolites. Both co-metabolites have been involved in the pathogenesis of accelerated cardiovascular disease and renal disease progression. Available evidence suggests that indoxyl sulfate and p-cresyl sulfate may be considered candidate biomarkers of the human enterotype and may help to explain the link between diet and cardiovascular disease burden. OBJECTIVE AND DESIGN Information on clinical determinants and heritability of indoxyl sulfate and p-cresyl sulfate serum is non-existing. To clarify this issue, the authors determined serum levels of indoxyl sulfate and p-cresyl sulfate in 773 individuals, recruited in the frame of the Flemish Study on Environment, Genes and Health Outcomes (FLEMENGHO study). RESULTS Serum levels of indoxyl sulfate and p-cresyl sulfate amounted to 3.1 (2.4-4.3) and 13.0 (7.4-21.5) μM, respectively. Regression analysis identified renal function, age and sex as independent determinants of both co-metabolites. Both serum indoxyl sulfate (h2 = 0.17) and p-cresyl sulfate (h2 = 0.18) concentrations showed moderate but significant heritability after adjustment for covariables, with significant genetic and environmental correlations for both co-metabolites. LIMITATIONS Family studies cannot provide conclusive evidence for a genetic contribution, as confounding by shared environmental effects can never be excluded. CONCLUSIONS The heritability of indoxyl sulfate and p-cresyl sulfate is moderate. Besides genetic host factors and environmental factors, also renal function, sex and age influence the serum levels of these co-metabolites.
Collapse
|
35
|
Mafra D, Barros AF, Fouque D. Dietary protein metabolism by gut microbiota and its consequences for chronic kidney disease patients. Future Microbiol 2014; 8:1317-23. [PMID: 24059921 DOI: 10.2217/fmb.13.103] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The breakdown of proteins and peptides by colonic microorganisms yields a great diversity of end products, including short-chain fatty acids, ammonia, amines, phenols, indoles, thiols, CO2, H2 and H2S, many of which have toxic properties. An increase of the dietary protein load in healthy individuals results in enhanced generation of these toxins, many of which are rapidly cleared by the kidneys. In this regard, the impact upon the colonic microbiota of controlled changes in the dietary protein has not been examined in chronic kidney disease patients. This review focuses on the impact of dietary proteins on the intestinal microbiota and its possible consequences for chronic kidney disease patients.
Collapse
Affiliation(s)
- Denise Mafra
- Cardiovascular Sciences Graduate Program, Universidade Federal Fluminense, Niterói-RJ, Brazil
| | | | | |
Collapse
|
36
|
Koppe L, Pelletier CC, Alix PM, Kalbacher E, Fouque D, Soulage CO, Guebre-Egziabher F. Insulin resistance in chronic kidney disease: new lessons from experimental models. Nephrol Dial Transplant 2013; 29:1666-74. [PMID: 24286973 DOI: 10.1093/ndt/gft435] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Insulin resistance (IR) is a common feature of chronic kidney disease (CKD), but the underlying mechanisms still remain unclear. A growing body of evidence suggests that IR and its associated metabolic disorders are important contributors for the cardiovascular burden of these patients. In recent years, the modification of the intestinal flora and activation of inflammation pathways have been implicated in the pathogenesis of IR in obese and diabetic patients. All these pathways ultimately lead to lipid accumulation in ectopic sites and impair insulin signalling. These important discoveries have led to major advances in understanding the mechanisms of uraemia-induced IR. Indeed, recent studies show impairment of the intestinal barrier function and changes in the composition of the gut microbiome during CKD that can contribute to the prevailing inflammation, and the production and absorption of toxins generated from bacterial metabolism. The specific role of individual uraemic toxins in the pathogenesis of IR has been highlighted in rodents. Moreover, correcting some uraemia-associated factors by modulating the intestinal flora improves insulin sensitivity. This review outlines potential mechanisms by which important modifications of body homeostasis induced by the decline in kidney function can affect insulin sensitivity, and the relevance of recent advances in the field to provide novel therapeutic approaches to reduce IR associated cardiovascular mortality.
Collapse
Affiliation(s)
- Laetitia Koppe
- Université de Lyon, INSERM U1060, CarMeN, INSA de Lyon, Univ Lyon-1, Villeurbanne, France Department of Nephrology, Hospices Civils de Lyon, Hôpital E Herriot, Lyon, France
| | - Caroline C Pelletier
- Université de Lyon, INSERM U1060, CarMeN, INSA de Lyon, Univ Lyon-1, Villeurbanne, France Department of Nephrology, Hospices Civils de Lyon, Hôpital E Herriot, Lyon, France
| | - Pascaline M Alix
- Université de Lyon, INSERM U1060, CarMeN, INSA de Lyon, Univ Lyon-1, Villeurbanne, France Department of Nephrology, Hospices Civils de Lyon, Hôpital E Herriot, Lyon, France
| | - Emilie Kalbacher
- Université de Lyon, INSERM U1060, CarMeN, INSA de Lyon, Univ Lyon-1, Villeurbanne, France Department of Nephrology, Hospices Civils de Lyon, Hôpital E Herriot, Lyon, France
| | - Denis Fouque
- Université de Lyon, INSERM U1060, CarMeN, INSA de Lyon, Univ Lyon-1, Villeurbanne, France Department of Nephrology, Hospices Civils de Lyon, Hôpital E Herriot, Lyon, France
| | - Christophe O Soulage
- Department of Nephrology, Hospices Civils de Lyon, Hôpital E Herriot, Lyon, France
| | - Fitsum Guebre-Egziabher
- Université de Lyon, INSERM U1060, CarMeN, INSA de Lyon, Univ Lyon-1, Villeurbanne, France Department of Nephrology, Hospices Civils de Lyon, Hôpital E Herriot, Lyon, France
| |
Collapse
|
37
|
Eloot S, Van Biesen W, Glorieux G, Neirynck N, Dhondt A, Vanholder R. Does the adequacy parameter Kt/V(urea) reflect uremic toxin concentrations in hemodialysis patients? PLoS One 2013; 8:e76838. [PMID: 24236005 PMCID: PMC3827207 DOI: 10.1371/journal.pone.0076838] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 08/31/2013] [Indexed: 11/18/2022] Open
Abstract
Hemodialysis aims at removing uremic toxins thus decreasing their concentrations. The present study investigated whether Kt/Vurea, used as marker of dialysis adequacy, is correlated with these concentrations. Predialysis blood samples were taken before a midweek session in 71 chronic HD patients. Samples were analyzed by colorimetry, HPLC, or ELISA for a broad range of uremic solutes. Solute concentrations were divided into four groups according to quartiles of Kt/Vurea, and also of different other parameters with potential impact, such as age, body weight (BW), Protein equivalent of Nitrogen Appearance (PNA), Residual Renal Function (RRF), and dialysis vintage. Dichotomic concentration comparisons were performed for gender and Diabetes Mellitus (DM). Analysis of Variance in quartiles of Kt/Vurea did not show significant differences for any of the solute concentrations. For PNA, however, concentrations showed significant differences for urea (P<0.001), uric acid (UA), p-cresylsulfate (PCS), and free PCS (all P<0.01), and for creatinine (Crea) and hippuric acid (HA) (both P<0.05). For RRF, concentrations varied for β2-microglobulin (P<0.001), HA, free HA, free indoxyl sulfate, and free indole acetic acid (all P<0.01), and for p-cresylglucuronide (PCG), 3-carboxy-4-methyl-5-propyl-2-furanpropionic acid (CMPF), free PCS, and free PCG (all P<0.05). Gender and body weight only showed differences for Crea and UA, while age, vintage, and diabetes mellitus only showed differences for one solute concentration (UA, UA, and free PCS, respectively). Multifactor analyses indicated a predominant association of concentration with protein intake and residual renal function. In conclusion, predialysis concentrations of uremic toxins seem to be dependent on protein equivalent of nitrogen appearance and residual renal function, and not on dialysis adequacy as assessed by Kt/Vurea. Efforts to control intestinal load of uremic toxin precursors by dietary or other interventions, and preserving RRF seem important approaches to decrease uremic solute concentration and by extension their toxicity.
Collapse
Affiliation(s)
- Sunny Eloot
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, Gent, Belgium
- * E-mail:
| | - Wim Van Biesen
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, Gent, Belgium
| | - Griet Glorieux
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, Gent, Belgium
| | - Nathalie Neirynck
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, Gent, Belgium
| | - Annemieke Dhondt
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, Gent, Belgium
| | - Raymond Vanholder
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, Gent, Belgium
| |
Collapse
|
38
|
Pre-, pro-, and synbiotics: do they have a role in reducing uremic toxins? A systematic review and meta-analysis. Int J Nephrol 2012; 2012:673631. [PMID: 23316359 PMCID: PMC3536316 DOI: 10.1155/2012/673631] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 08/29/2012] [Indexed: 12/25/2022] Open
Abstract
Objective. This paper assessed the effectiveness of pre-, pro-, and synbiotics on reducing two protein-bound uremic toxins, p-cresyl sulphate (PCS) and indoxyl sulphate (IS). Methods. English language studies reporting serum, urinary, or fecal PCS and/or IS (or their precursors) following pre-, pro-, or synbiotic interventions (>1 day) in human adults were included. Population estimates of differences in the outcomes between the pre- and the postintervention were estimated for subgroups of studies using four meta-analyses. Quality was determined using the GRADE approach. Results. 19 studies met the inclusion criteria, 14 in healthy adults and five in haemodialysis patients. Eight studies investigated prebiotics, six probiotics, one synbiotics, one both pre- and probiotics, and three studies trialled all three interventions. The quality of the studies ranged from moderate to very low. 12 studies were included in the meta-analyses with all four meta-analyses reporting statistically significant reductions in IS and PCS with pre- and probiotic therapy. Conclusion. There is a limited but supportive evidence for the effectiveness of pre- and probiotics on reducing PCS and IS in the chronic kidney disease population. Further studies are needed to provide more definitive findings before routine clinical use can be recommended.
Collapse
|
39
|
Furutani J, Segawa H, Aranami F, Kuwahara S, Sugano M, Bannai K, Yamato H, Ito M, Miyamoto KI. Dietary inorganic phosphorus regulates the intestinal peptide transporter PepT1. J Ren Nutr 2012; 23:e11-20. [PMID: 22677630 DOI: 10.1053/j.jrn.2012.02.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 02/18/2012] [Accepted: 02/21/2012] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Both organic and inorganic phosphorus (Pi) are present in regularly consumed foods, such as meats, eggs, and dairy products. Pi is often included in foods as an additive (as hidden phosphorus). The intestinal peptide transporter PepT1 mediates protein absorption, which is disturbed in renal insufficiency. Our aim was to determine the effects of dietary Pi content on the peptide transport activity and expression of PepT1. METHODS The following animal models were used: (1) 7-week-old male Wistar rats; and (2) rats that underwent 3/4 nephrectomy to induce chronic kidney disease (CKD). The rats were fed a normal-protein (20%) diet containing low (0.02%), normal (0.6%), or high (1.2%) Pi levels. They were also fed diets containing varying amounts of protein and either low or normal Pi levels as follows: (1) low Pi/normal protein, (2) low Pi/high (50%) protein, (3) normal Pi/normal protein, and (4) normal Pi/high protein. RESULTS Intestinal peptide transport activity and PepT1 expression levels were significantly higher in the CKD rats than in sham-operated control ones. Compared with the normal-protein diet, the high-protein diet increased PepT1 expression in the CKD rats. Intestinal dipeptide transport activity and PepT1 protein levels did not increase in the rats fed the low-Pi/high-protein diet. In contrast, intestinal dipeptide transport activity and PepT1 protein expression were markedly increased in the rats fed the normal-Pi/high-protein diet. CONCLUSION Dietary Pi levels regulate intestinal peptide transport activity through PepT1.
Collapse
Affiliation(s)
- Junya Furutani
- Department of Molecular Nutrition, Institution of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Slee AD. Exploring metabolic dysfunction in chronic kidney disease. Nutr Metab (Lond) 2012; 9:36. [PMID: 22537670 PMCID: PMC3407016 DOI: 10.1186/1743-7075-9-36] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 04/26/2012] [Indexed: 02/07/2023] Open
Abstract
Impaired kidney function and chronic kidney disease (CKD) leading to kidney failure and end-stage renal disease (ESRD) is a serious medical condition associated with increased morbidity, mortality, and in particular cardiovascular disease (CVD) risk. CKD is associated with multiple physiological and metabolic disturbances, including hypertension, dyslipidemia and the anorexia-cachexia syndrome which are linked to poor outcomes. Specific hormonal, inflammatory, and nutritional-metabolic factors may play key roles in CKD development and pathogenesis. These include raised proinflammatory cytokines, such as interleukin-1 and −6, tumor necrosis factor, altered hepatic acute phase proteins, including reduced albumin, increased C-reactive protein, and perturbations in normal anabolic hormone responses with reduced growth hormone-insulin-like growth factor-1 axis activity. Others include hyperactivation of the renin-angiotensin aldosterone system (RAAS), with angiotensin II and aldosterone implicated in hypertension and the promotion of insulin resistance, and subsequent pharmacological blockade shown to improve blood pressure, metabolic control and offer reno-protective effects. Abnormal adipocytokine levels including leptin and adiponectin may further promote the insulin resistant, and proinflammatory state in CKD. Ghrelin may be also implicated and controversial studies suggest activities may be reduced in human CKD, and may provide a rationale for administration of acyl-ghrelin. Poor vitamin D status has also been associated with patient outcome and CVD risk and may indicate a role for supplementation. Glucocorticoid activities traditionally known for their involvement in the pathogenesis of a number of disease states are increased and may be implicated in CKD-associated hypertension, insulin resistance, diabetes risk and cachexia, both directly and indirectly through effects on other systems including activation of the mineralcorticoid receptor. Insight into the multiple factors altered in CKD may provide useful information on disease pathogenesis, clinical assessment and treatment rationale such as potential pharmacological, nutritional and exercise therapies.
Collapse
Affiliation(s)
- Adrian D Slee
- School of Life Sciences, Brayford Pool Campus, University of Lincoln, Lincoln, UK.
| |
Collapse
|
41
|
Vanholder R, Eloot S, Schepers E, Neirynck N, Glorieux G, Massy Z. an Obituary for GFR as the main marker for kidney function? Semin Dial 2011; 25:9-14. [PMID: 22141430 DOI: 10.1111/j.1525-139x.2011.01003.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This publication comments on the recently published findings of a study by Eloot et al. (cJASN, 6: 1266-1273, 2011) that evaluated the correlation between several formulae for calculating estimated GFR (eGFR) and different low molecular weight uremic toxins; eGFRs were based on serum creatinine (SCrea), cystatin C (Cys C), or a combination of both. Unexpectedly, the correlations for the different solutes were highly inconsistent, irrespective of the eGFR formula. On the other hand, the different eGFR formulae gave consistent results per solute. Correlation coefficients for some solutes were low (hippuric acid, p-cresylsulfate, indole acetic acid, uric acid, asymmetric dimethylarginine) to nonsignificant (carboxy-methyl-propyl-furanpropionic acid). These data point to the fact that eGFR is a deceiving predictor of uremic solute concentration and their biological action; this inconsistency is very likely the result of the impact of other factors affecting concentration, such as tubular secretion, generation by intestinal flora and metabolism.
Collapse
|
42
|
Abstract
The transport of urea and other solutes across the peritoneal membrane follows the general dialytic principles of diffusion and convection. Nevertheless, the unique anatomic configuration of the peritoneum suggests that specific modeling is needed to explain solute kinetics in peritoneal dialysis. The historical two-pore model of membrane transport had to be adjusted by adding a third pore, and the distributed model has further complemented this analysis. Recent findings with regard to the endothelial glycocalyx add an additional level of complexity. Unfortunately, most studies to date use only urea, glucose, and creatinine kinetics to describe transport during peritoneal dialysis and adequacy of solute removal. Studies of transport of other classes of uremic solutes are scanty but allow us to gain insight into how peritoneal dialysis works.
Collapse
Affiliation(s)
- Bert Bammens
- Department of Nephrology, Dialysis and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
43
|
Civilibal M, Caliskan S, Kurugoglu S, Candan C, Canpolat N, Sever L, Kasapcopur O, Arisoy N. Progression of coronary calcification in pediatric chronic kidney disease stage 5. Pediatr Nephrol 2009; 24:555-63. [PMID: 18982357 DOI: 10.1007/s00467-008-1038-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2008] [Revised: 09/30/2008] [Accepted: 10/06/2008] [Indexed: 02/06/2023]
Abstract
Coronary artery calcification (CAC) is common in adults with chronic kidney disease (CKD) and progresses with time. However, data are limited for younger patients. We have previously reported CAC in eight of 53 children with CKD. After 2 years, CAC evaluation was repeated in 48 patients. The median CAC score (CACS) increased from 101.3 (1473.6 +/- 1978.6, range 8.5-4332) to 1759.2 (2236.4 +/- 2463.3, range 0-5858) Agatston units (AU). When the individual changes in CACS were evaluated one by one, we showed a mild decrease in two patients on hemodialysis (HD) and in one transplant (Tx) recipient, a moderate increase in one patient on HD, one on peritoneal dialysis (PD) and one Tx recipient, and a large increase in one HD patient. Also, CAC disappeared in one HD patient. All patients with no calcification at baseline remained calcification-free at follow-up. To obtain the individual cumulative exposure, we calculated time-averaged mean values, using the laboratory values from the beginning of dialysis to the first and second multidetector spiral computed tomography (MDCT) scans (baseline and final values, respectively). Final CACS was positively related to final calcium-phosphorus (CaxP) product, while CAC progression was inversely associated with final serum albumin level. This report is the first study with the largest number and the youngest cohort to document the natural history of coronary calcification.
Collapse
Affiliation(s)
- Mahmut Civilibal
- Department of Pediatric Nephrology, Istanbul University Cerrahpasa Medical Faculty, Istanbul, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Pham NM, Recht NS, Hostetter TH, Meyer TW. Removal of the protein-bound solutes indican and p-cresol sulfate by peritoneal dialysis. Clin J Am Soc Nephrol 2007; 3:85-90. [PMID: 18045861 DOI: 10.2215/cjn.02570607] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND OBJECTIVES Protein-bound solutes are poorly cleared by peritoneal dialysis. We examined the hypothesis that plasma concentrations of bound solutes would therefore rise as residual renal function is lost. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Clearances of urea indican and p-cresol sulfate were measured in peritoneal dialysis patients with and without residual function. RESULTS In patients with residual function, protein binding restricted the peritoneal indican and p-cresol sulfate clearances to 0.3 +/- 0.1 ml/min, as compared to the peritoneal urea clearance of 5.5 +/- 1.1 ml/min. The urinary indican and p-cresol sulfate clearances of 2.7 +/- 2.5 and 1.3 +/- 1.0 ml/min were closer to the urinary urea clearance of 3.9 +/- 2.2 ml/min, reflecting the superior ability of native kidney function to clear bound solutes. Urinary clearance thus provided the majority of the total indican and p-cresol sulfate clearances of 3.0 +/- 2.5 and 1.6 +/- 1.0 ml/min in patients with residual function but the minority of total urea clearance of 9.4 +/- 2.2 ml/min. Loss of residual function lowered the total clearances for indican and p-cresol sulfate to 0.5 +/- 0.2 and 0.4 +/- 0.2 ml/min, whereas the urea clearance fell only slightly. However there was only a modest increase in the plasma indican level and no increase in the plasma p-cresol sulfate level in patients with no residual function because reduction in the daily removal of these solutes accompanied the reduction in their total clearance rates. CONCLUSIONS Reduction in the removal of indican and p-cresol sulfate kept plasma levels from rising markedly when residual function was lost.
Collapse
Affiliation(s)
- Nhat M Pham
- Department of Medicine, VA Palo Alto Health Care System, and Stanford University, Palo Alto, California, USA
| | | | | | | |
Collapse
|
45
|
Braden B, Lembcke B, Kuker W, Caspary WF. 13C-breath tests: current state of the art and future directions. Dig Liver Dis 2007; 39:795-805. [PMID: 17652042 DOI: 10.1016/j.dld.2007.06.012] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2007] [Revised: 06/14/2007] [Accepted: 06/28/2007] [Indexed: 12/11/2022]
Abstract
13C-breath tests provide a non-invasive diagnostic method with high patient acceptance. In vivo, human and also bacterial enzyme activities, organ functions and transport processes can be assessed semiquantitatively using breath tests. As the samples can directly be analysed using non-dispersive isotope selective infrared spectrometers or sent to analytical centres by normal mail breath tests can be easily performed also in primary care settings. The 13C-urea breath test which detects a Helicobacter pylori infection of the stomach is the most prominent application of stable isotopes. Determination of gastric emptying using test meals labelled with 13C-octanoic or 13C-acetic acid provide reliable results compared to scintigraphy. The clinical use of 13C-breath tests for the diagnosis of exocrine pancreatic insufficiency is still limited due to expensive substrates and long test periods with many samples. However, the quantification of liver function using hepatically metabolised 13C-substrates is clinically helpful in special indications. The stable isotope technique presents an elegant, non-invasive diagnostic tool promising further options of clinical applications. This review is aimed at providing an overview on the relevant clinical applications of 13C-breath tests.
Collapse
Affiliation(s)
- B Braden
- John Radcliffe Hospital, Headley Way, OX3 9DU Oxford, UK.
| | | | | | | |
Collapse
|
46
|
Evenepoel P, Bammens B, Verbeke K, Vanrenterghem Y. Superior dialytic clearance of β2-microglobulin and p-cresol by high-flux hemodialysis as compared to peritoneal dialysis. Kidney Int 2006; 70:794-9. [PMID: 16820785 DOI: 10.1038/sj.ki.5001640] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Both residual renal and dialytic clearance confer to the total solute clearance in dialysis patients. Dialytic clearances of the middle molecule beta-microglobulin (beta(2)M) and the protein-bound solute p-cresol (pcr) are generally believed to be higher with peritoneal dialysis (PD) as compared to hemodialysis (HD). Supportive data, however, are lacking. We performed a single-center cross-sectional observational study including 70 unselected patients treated with either high-flux HD (n=20) or PD (n=50). Mid-day serum levels (PD) and time-averaged concentrations (HD) of the water-soluble solutes urea nitrogen, creatinine and phosphate, the middle molecule beta(2)M, and the protein-bound solute pcr were determined. Dialytic solute clearances (l/week/1.73 m(2)) were calculated from total dialysate collection during the mid-week session in HD and 24 h dialysate collection in PD. Renal clearances were calculated for each of the respective solutes from a timed urine collection. Total clearances were obtained by summation. HD delivered significantly higher clearances of all retention solutes studied. This superiority was especially pronounced for pcr (30.9+/-62.7 vs 4.4+/-2.3, HD vs PD, P<0.0001) and beta(2)M (28.6+/-6.6 vs 5.8+/-3.1, HD vs PD, P<0.0001). Renal clearances, conversely, were significantly higher in patients on PD. Serum levels of all solutes but pcr were significantly lower in HD than in PD. Both a higher residual renal function and a lower generation rate contribute to the lower pcr levels in PD. In conclusion, superior dialytic clearance of both water-soluble solutes, beta(2)M, and pcr is achieved by high-flux HD as compared to PD.
Collapse
Affiliation(s)
- P Evenepoel
- Department of Medicine, Division of Nephrology, University Hospital Leuven, Leuven, Belgium.
| | | | | | | |
Collapse
|
47
|
Bammens B, Evenepoel P, Keuleers H, Verbeke K, Vanrenterghem Y. Free serum concentrations of the protein-bound retention solute p-cresol predict mortality in hemodialysis patients. Kidney Int 2006; 69:1081-7. [PMID: 16421516 DOI: 10.1038/sj.ki.5000115] [Citation(s) in RCA: 279] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Based on in vitro data, protein-bound uremic retention solutes have increasingly been recognized to play a pathophysiological role in the uremic syndrome. p-Cresol, a representative of this group of molecules, has been shown to be implicated in uremic immunodeficiency and endothelial dysfunction, potentially linking its serum levels to mortality. Thus far, however, no clinical information on this issue is available. To determine the relationship between p-cresol and all-cause mortality, 175 prevalent hemodialysis (HD) patients were enrolled in a prospective study. At baseline, serum levels of the water-soluble solutes urea, creatinine, and phosphate, the middle molecule beta2-microglobulin, total and free concentrations of the protein-bound solute p-cresol, and several risk factors for mortality were evaluated. During a median follow-up of 34 months, 60 patients died. Baseline comorbidity (Davies score) (hazard ratio (HR), 1.49; 95% confidence interval (95% CI), 1.19-1.86), impaired nutritional status (HR, 4.22; 95% CI, 2.15-8.29), time since initiation of dialysis (HR, 0.98; 95% CI, 0.97-1.00), and higher free concentrations of the protein-bound solute p-cresol (HR, 2.28; 95% CI, 1.12-4.64) were independently associated with mortality (multivariate Cox proportional hazards analysis). Our data suggest that free serum levels of p-cresol, a representative of the protein-bound uremic retention solutes, are associated with mortality in HD patients. These findings may encourage nephrologists to widen their field of interest beyond the scope of small water-soluble uremic solutes and middle molecules.
Collapse
Affiliation(s)
- B Bammens
- Department of Medicine, Division of Nephrology, University Hospital Gasthuisberg, Leuven, Belgium
| | | | | | | | | |
Collapse
|
48
|
Evenepoel P, Bammens B, Verbeke K, Vanrenterghem Y. Acarbose treatment lowers generation and serum concentrations of the protein-bound solute p-cresol: a pilot study. Kidney Int 2006; 70:192-8. [PMID: 16688114 DOI: 10.1038/sj.ki.5001523] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Several protein-bound uremic retention solutes (including p-cresol) originate from colonic bacterial fermentation of protein. Higher colonic availability of carbohydrates drives this process towards lower production of toxic metabolites. Small intestinal alpha-glucosidase inhibitors like Acarbose (Glucobay) enhance the amount of undigested carbohydrates reaching the colon. We studied the effect of Acarbose on generation and serum concentrations of p-cresol. Nine healthy volunteers (age 25 (22-36) years) with a creatinine clearance of 89.6 ml/min/1.73 m(2) (85.5-116.4) were treated with Acarbose for 3 weeks. Dose was gradually increased to reach 300 mg/day after 1 week. Blood sampling, 24-h urine and stool collections on 3 consecutive days were performed before and during the last days of the treatment period. p-Cresol generation was estimated from mean 24-h urinary elimination. Gastrointestinal side effects, if present, were mild to moderate. Serum concentrations of p-cresol declined significantly after Acarbose treatment (before: 1.14 mg/l (0.93-3.03); after: 1.11 mg/l (0.31-1.82); P=0.047). Urinary excretion of p-cresol, reflecting its colonic generation rate, was significantly lower after treatment (before: 29.93 mg/day (6.79-75.19); after: 10.54 mg/day (1.08-30.85); P=0.031). The fecal excretion of nitrogen increased after treatment (before: 1.04 g/day (0.47-2.29); after: 1.99 g/day (0.76-3.08); P=0.047). This pilot study suggests that Acarbose treatment lowers generation and serum concentrations of the protein-bound uremic solute p-cresol. Although further confirmation is warranted, the data may point to a novel treatment option for chronic kidney disease patients in view of the potential toxic effects of p-cresol and related substances.
Collapse
Affiliation(s)
- P Evenepoel
- Department of Medicine, Division of Nephrology, University Hospital Gasthuisberg, Leuven, Belgium.
| | | | | | | |
Collapse
|
49
|
Suliman ME, Qureshi AR, Stenvinkel P, Pecoits-Filho R, Bárány P, Heimbürger O, Anderstam B, Rodríguez Ayala E, Divino Filho JC, Alvestrand A, Lindholm B. Inflammation contributes to low plasma amino acid concentrations in patients with chronic kidney disease. Am J Clin Nutr 2005. [DOI: 10.1093/ajcn/82.2.342] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Mohammed E Suliman
- From the Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - A Rashid Qureshi
- From the Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Peter Stenvinkel
- From the Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Roberto Pecoits-Filho
- From the Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Peter Bárány
- From the Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Olof Heimbürger
- From the Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Björn Anderstam
- From the Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Ernesto Rodríguez Ayala
- From the Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - José C Divino Filho
- From the Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Anders Alvestrand
- From the Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Bengt Lindholm
- From the Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
50
|
Suliman ME, Qureshi AR, Stenvinkel P, Pecoits-Filho R, Bárány P, Heimbürger O, Anderstam B, Rodríguez Ayala E, Divino Filho JC, Alvestrand A, Lindholm B. Inflammation contributes to low plasma amino acid concentrations in patients with chronic kidney disease. Am J Clin Nutr 2005; 82:342-9. [PMID: 16087977 DOI: 10.1093/ajcn.82.2.342] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Inflammation and malnutrition are common in chronic kidney disease (CKD) patients, and plasma concentrations of free amino acids (AAs) in these patients are often abnormal. Malnutrition contributes to alterations in AA concentrations. OBJECTIVE The objective was to study the effects of inflammation on plasma AA concentrations. DESIGN Concentrations of plasma AAs, serum albumin, and several inflammatory markers were analyzed in 200 fasting, nondiabetic CKD patients who were close to the start of renal replacement therapy. The nutritional status of these patients was assessed by a subjective global assessment. RESULTS The patients with inflammation [C-reactive protein (CRP) concentrations >10 mg/L] or malnutrition had lower AA concentrations than did the patients with no inflammation or malnutrition. The presence of both inflammation and malnutrition was associated with more marked reductions in AA concentrations than was malnutrition alone. Significant inverse correlations were observed between the plasma concentrations of most of the essential and nonessential AAs and inflammatory markers, whereas serum albumin concentrations were positively correlated with several AA concentrations. A stepwise multivariate regression analysis showed that serum CRP concentrations were independently associated with low concentrations of the sums of both nonessential AAs and all AAs. An analysis of all-cause mortality with a Kaplan-Meier test showed that the patients with higher AA concentrations had significantly better survival than did the patients with lower AA concentrations. CONCLUSIONS Plasma AA concentrations are low in CKD patients with inflammation and are inversely correlated with concentrations of inflammatory markers. Although inflammation and malnutrition are closely related, CRP concentrations were independently associated with low concentrations of the sums of both nonessential AAs and all AAs, which suggests an independent role of inflammation as a cause of low plasma AA concentrations in CKD patients.
Collapse
Affiliation(s)
- Mohammed E Suliman
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|