1
|
Cabała S, Ożgo M, Herosimczyk A. The Kidney-Gut Axis as a Novel Target for Nutritional Intervention to Counteract Chronic Kidney Disease Progression. Metabolites 2024; 14:78. [PMID: 38276313 PMCID: PMC10819792 DOI: 10.3390/metabo14010078] [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: 12/20/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
A well-balanced diet is integral for overall health, aiding in managing key risk factors for kidney damage like hypertension while supplying necessary precursors for metabolite production. Dietary choices directly influence the composition and metabolic patterns of the gut microbiota, showing promise as therapeutic tools for addressing various health conditions, including chronic kidney diseases (CKD). CKD pathogenesis involves a decline in the glomerular filtration rate and the retention of nitrogen waste, fostering gut dysbiosis and the excessive production of bacterial metabolites. These metabolites act as uremic toxins, contributing to inflammation, oxidative stress, and tissue remodeling in the kidneys. Dietary interventions hold significance in reducing oxidative stress and inflammation, potentially slowing CKD progression. Functional ingredients, nutrients, and nephroprotective phytoconstituents could modulate inflammatory pathways or impact the gut mucosa. The "gut-kidney axis" underscores the impact of gut microbes and their metabolites on health and disease, with dysbiosis serving as a triggering event in several diseases, including CKD. This review provides a comprehensive overview, focusing on the gut-liver axis, and explores well-established bioactive substances as well as specific, less-known nutraceuticals showing promise in supporting kidney health and positively influencing CKD progression.
Collapse
Affiliation(s)
| | | | - Agnieszka Herosimczyk
- Department of Physiology, Cytobiology and Proteomics, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology Szczecin, Klemensa Janickiego 29, 71-270 Szczecin, Poland; (S.C.); (M.O.)
| |
Collapse
|
2
|
Nhan J, Sgambat K, Moudgil A. Plant-based diets: a fad or the future of medical nutrition therapy for children with chronic kidney disease? Pediatr Nephrol 2023; 38:3597-3609. [PMID: 36786858 DOI: 10.1007/s00467-023-05875-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 02/15/2023]
Abstract
Plant-based diets are growing in popularity worldwide due to the importance of reducing the population's ecological footprint as well as an emerging role in the prevention and treatment of chronic human diseases. In adults, plant-based diets have been shown to be beneficial for preventing and controlling conditions that are common in patients with chronic kidney disease (CKD), such as obesity, hypertension, type 2 diabetes, dyslipidemia, and metabolic acidosis. Emerging evidence suggests that the higher fiber content of plant-based diets may help to modulate production of uremic toxins through beneficial shifts in the gut microbiome. The effects of the plant-based diet on progression of CKD remain controversial, and there are no data to support this in children. However, knowledge that the bioavailability of potassium and phosphorus from plant-based foods is reduced has led to recent changes in international kidney-friendly diet recommendations for children with CKD. The new guidelines advise that high potassium fruits and vegetables should no longer be automatically excluded from the kidney-friendly diet. In fact, a plant-based diet can be safely implemented in children with CKD through building the diet around whole, high fiber foods, avoiding processed foods and using recommended cooking methods to control potassium. The health benefits of the plant-based diet compared to omnivorous diets in children with CKD need investigation.
Collapse
Affiliation(s)
- Jennifer Nhan
- Department of Nephrology, Children's National Hospital, 111 Michigan Avenue NW, Washington, DC, 20010, USA
| | - Kristen Sgambat
- Department of Nephrology, Children's National Hospital, 111 Michigan Avenue NW, Washington, DC, 20010, USA.
| | - Asha Moudgil
- Department of Nephrology, Children's National Hospital, 111 Michigan Avenue NW, Washington, DC, 20010, USA
| |
Collapse
|
3
|
Cooper TE, Khalid R, Chan S, Craig JC, Hawley CM, Howell M, Johnson DW, Jaure A, Teixeira-Pinto A, Wong G. Synbiotics, prebiotics and probiotics for people with chronic kidney disease. Cochrane Database Syst Rev 2023; 10:CD013631. [PMID: 37870148 PMCID: PMC10591284 DOI: 10.1002/14651858.cd013631.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
BACKGROUND Chronic kidney disease (CKD) is a major public health problem affecting 13% of the global population. Prior research has indicated that CKD is associated with gut dysbiosis. Gut dysbiosis may lead to the development and/or progression of CKD, which in turn may in turn lead to gut dysbiosis as a result of uraemic toxins, intestinal wall oedema, metabolic acidosis, prolonged intestinal transit times, polypharmacy (frequent antibiotic exposures) and dietary restrictions used to treat CKD. Interventions such as synbiotics, prebiotics, and probiotics may improve the balance of the gut flora by altering intestinal pH, improving gut microbiota balance and enhancing gut barrier function (i.e. reducing gut permeability). OBJECTIVES This review aimed to evaluate the benefits and harms of synbiotics, prebiotics, and probiotics for people with CKD. SEARCH METHODS We searched the Cochrane Kidney and Transplant Register of Studies up to 9 October 2023 through contact with the Information Specialist using search terms relevant to this review. Studies in the Register are identified through searches of CENTRAL, MEDLINE, and EMBASE, conference proceedings, the International Clinical Trials Registry Platform (ICTRP) Search Portal and ClinicalTrials.gov. SELECTION CRITERIA We included randomised controlled trials (RCTs) measuring and reporting the effects of synbiotics, prebiotics, or probiotics in any combination and any formulation given to people with CKD (CKD stages 1 to 5, including dialysis and kidney transplant). Two authors independently assessed the retrieved titles and abstracts and, where necessary, the full text to determine which satisfied the inclusion criteria. DATA COLLECTION AND ANALYSIS Data extraction was independently carried out by two authors using a standard data extraction form. Summary estimates of effect were obtained using a random-effects model, and results were expressed as risk ratios (RR) and their 95% confidence intervals (CI) for dichotomous outcomes, and mean difference (MD) or standardised mean difference (SMD) and 95% CI for continuous outcomes. The methodological quality of the included studies was assessed using the Cochrane risk of bias tool. Data entry was carried out by one author and cross-checked by another. Confidence in the evidence was assessed using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach. MAIN RESULTS Forty-five studies (2266 randomised participants) were included in this review. Study participants were adults (two studies in children) with CKD ranging from stages 1 to 5, with patients receiving and not receiving dialysis, of whom half also had diabetes and hypertension. No studies investigated the same synbiotic, prebiotic or probiotic of similar strains, doses, or frequencies. Most studies were judged to be low risk for selection bias, performance bias and reporting bias, unclear risk for detection bias and for control of confounding factors, and high risk for attrition and other biases. Compared to prebiotics, it is uncertain whether synbiotics improve estimated glomerular filtration rate (eGFR) at four weeks (1 study, 34 participants: MD -3.80 mL/min/1.73 m², 95% CI -17.98 to 10.38), indoxyl sulfate at four weeks (1 study, 42 participants: MD 128.30 ng/mL, 95% CI -242.77 to 499.37), change in gastrointestinal (GI) upset (borborymgi) at four weeks (1 study, 34 participants: RR 15.26, 95% CI 0.99 to 236.23), or change in GI upset (Gastrointestinal Symptom Rating Scale) at 12 months (1 study, 56 participants: MD 0.00, 95% CI -0.27 to 0.27), because the certainty of the evidence was very low. Compared to certain strains of prebiotics, it is uncertain whether a different strain of prebiotics improves eGFR at 12 weeks (1 study, 50 participants: MD 0.00 mL/min, 95% CI -1.73 to 1.73), indoxyl sulfate at six weeks (2 studies, 64 participants: MD -0.20 μg/mL, 95% CI -1.01 to 0.61; I² = 0%) or change in any GI upset, intolerance or microbiota composition, because the certainty of the evidence was very low. Compared to certain strains of probiotics, it is uncertain whether a different strain of probiotic improves eGFR at eight weeks (1 study, 30 participants: MD -0.64 mL/min, 95% CI -9.51 to 8.23; very low certainty evidence). Compared to placebo or no treatment, it is uncertain whether synbiotics improve eGFR at six or 12 weeks (2 studies, 98 participants: MD 1.42 mL/min, 95% CI 0.65 to 2.2) or change in any GI upset or intolerance at 12 weeks because the certainty of the evidence was very low. Compared to placebo or no treatment, it is uncertain whether prebiotics improves indoxyl sulfate at eight weeks (2 studies, 75 participants: SMD -0.14 mg/L, 95% CI -0.60 to 0.31; very low certainty evidence) or microbiota composition because the certainty of the evidence is very low. Compared to placebo or no treatment, it is uncertain whether probiotics improve eGFR at eight, 12 or 15 weeks (3 studies, 128 participants: MD 2.73 mL/min, 95% CI -2.28 to 7.75; I² = 78%), proteinuria at 12 or 24 weeks (1 study, 60 participants: MD -15.60 mg/dL, 95% CI -34.30 to 3.10), indoxyl sulfate at 12 or 24 weeks (2 studies, 83 participants: MD -4.42 mg/dL, 95% CI -9.83 to 1.35; I² = 0%), or any change in GI upset or intolerance because the certainty of the evidence was very low. Probiotics may have little or no effect on albuminuria at 12 or 24 weeks compared to placebo or no treatment (4 studies, 193 participants: MD 0.02 g/dL, 95% CI -0.08 to 0.13; I² = 0%; low certainty evidence). For all comparisons, adverse events were poorly reported and were minimal (flatulence, nausea, diarrhoea, abdominal pain) and non-serious, and withdrawals were not related to the study treatment. AUTHORS' CONCLUSIONS We found very few studies that adequately test biotic supplementation as alternative treatments for improving kidney function, GI symptoms, dialysis outcomes, allograft function, patient-reported outcomes, CVD, cancer, reducing uraemic toxins, and adverse effects. We are not certain whether synbiotics, prebiotics, or probiotics are more or less effective compared to one another, antibiotics, or standard care for improving patient outcomes in people with CKD. Adverse events were uncommon and mild.
Collapse
Affiliation(s)
- Tess E Cooper
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
| | - Rabia Khalid
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
| | - Samuel Chan
- Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
| | - Jonathan C Craig
- Cochrane Kidney and Transplant, Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
- College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Carmel M Hawley
- Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
| | - Martin Howell
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
| | - David W Johnson
- Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
| | - Allison Jaure
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
| | - Armando Teixeira-Pinto
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
| | - Germaine Wong
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
- Centre for Transplant and Renal Research, Westmead Hospital, Westmead, Australia
| |
Collapse
|
4
|
Martín-Del-Campo F, Avesani CM, Stenvinkel P, Lindholm B, Cueto-Manzano AM, Cortés-Sanabria L. Gut microbiota disturbances and protein-energy wasting in chronic kidney disease: a narrative review. J Nephrol 2023; 36:873-883. [PMID: 36689170 PMCID: PMC9869315 DOI: 10.1007/s40620-022-01560-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/18/2022] [Indexed: 01/24/2023]
Abstract
Protein-energy wasting (PEW) is common in patients with chronic kidney disease (CKD) and is associated with increased morbidity and mortality, and lower quality of life. It is a complex syndrome, in which inflammation and retention of uremic toxins are two main factors. Causes of inflammation and uremic toxin retention in CKD are multiple; however, gut dysbiosis plays an important role, serving as a link between those entities and PEW. Besides, there are several pathways by which microbiota may influence PEW, e.g., through effects on appetite mediated by microbiota-derived proteins and hormonal changes, or by impacting skeletal muscle via a gut-muscle axis. Hence, microbiota disturbances may influence PEW independently of its relationship with local and systemic inflammation. A better understanding of the complex interrelationships between microbiota and the host may help to explain how changes in the gut affect distant organs and systems of the body and could potentially lead to the development of new strategies targeting the microbiota to improve nutrition and clinical outcomes in CKD patients. In this review, we describe possible interactions of gut microbiota with nutrient metabolism, energy balance, hunger/satiety signals and muscle depletion, all of which are strongly related to PEW in CKD patients.
Collapse
Affiliation(s)
- Fabiola Martín-Del-Campo
- Unidad de Investigación Médica en Enfermedades Renales, Hospital de Especialidades, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico
| | - Carla Maria Avesani
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Technology and Intervention, Karolinska Institutet, M99 Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden
| | - Peter Stenvinkel
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Technology and Intervention, Karolinska Institutet, M99 Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden
| | - Bengt Lindholm
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Technology and Intervention, Karolinska Institutet, M99 Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden.
| | - Alfonso M Cueto-Manzano
- Unidad de Investigación Médica en Enfermedades Renales, Hospital de Especialidades, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico
| | - Laura Cortés-Sanabria
- Unidad de Investigación Médica en Enfermedades Renales, Hospital de Especialidades, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico
| |
Collapse
|
5
|
Wang H, Yin H, Zhong Y, Hu J, Xia S, Wang Z, Nie S, Xie M. Polysaccharides from fermented coix seed modulates circulating nitrogen and immune function by altering gut microbiota. Curr Res Food Sci 2022; 5:1994-2003. [PMID: 36324864 PMCID: PMC9619149 DOI: 10.1016/j.crfs.2022.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/24/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022] Open
Abstract
Coix lachryma-jobi L. seed is an important food item in Asia with culinary and medicinal values. The effects of non-fermented coix seed (NFC), fermented coix seed with Lactobacillus plantarum NCU137 (FC) and polysaccharides from NFC, FC (FCP) on mice circulating nitrogen and immune disorder induced by high relative humidity (RH, 90 ± 2%) exposure were compared. All the treatments reduced circulating nitrogen (BUN and ammonia) might via increasing excretion of fecal nitrogen induced by altering gut microbiota. In comparison, FC and FCP restored erythrocyte morphology by promoting erythrocyte Na+/K+ ATPase activity more effectively, and immune function was modulated by reducing plasma IgM and IFN-γ levels, up-regulating IL-4 and IL-6 levels. Herein, these results indicated that FCP, as the main active ingredient in FC, modulated circulating nitrogen through altering gut microbiota, and restored immune homeostasis by regulating Th1/Th2 cytokines in mice receiving high RH exposure.
Collapse
Affiliation(s)
- Hui Wang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China
| | - Hongmei Yin
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China,School of Health, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang, Jiangxi, 330022, China
| | - Yadong Zhong
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China
| | - Jielun Hu
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China
| | - Shengkun Xia
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China
| | - Zixuan Wang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China
| | - Shaoping Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China
| | - Mingyong Xie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China,Corresponding author.
| |
Collapse
|
6
|
Aguilera-Correa J, Gisbert-Garzarán M, Mediero A, Carias-Cálix R, Jiménez-Jiménez C, Esteban J, Vallet-Regí M. Arabic gum plus colistin coated moxifloxacin-loaded nanoparticles for the treatment of bone infection caused by Escherichia coli. Acta Biomater 2022; 137:218-237. [PMID: 34653694 DOI: 10.1016/j.actbio.2021.10.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/20/2021] [Accepted: 10/07/2021] [Indexed: 12/16/2022]
Abstract
Osteomyelitis is an inflammatory process of bone and bone marrow that may even lead to patient death. Even though this disease is mainly caused by Gram-positive organisms, the proportion of bone infections caused by Gram-negative bacteria, such as Escherichia coli, has significantly increased in recent years. In this work, mesoporous silica nanoparticles have been employed as platform to engineer a nanomedicine able to eradicate E. coli- related bone infections. For that purpose, the nanoparticles have been loaded with moxifloxacin and further functionalized with Arabic gum and colistin (AG+CO-coated MX-loaded MSNs). The nanosystem demonstrated high affinity toward E. coli biofilm matrix, thanks to AG coating, and marked antibacterial effect because of the bactericidal effect of moxifloxacin and the disaggregating effect of colistin. AG+CO-coated MX-loaded MSNs were able to eradicate the infection developed on a trabecular bone in vitro and showed pronounced antibacterial efficacy in vivo against an osteomyelitis provoked by E. coli. Furthermore, AG+CO-coated MX-loaded MSNs were shown to be essentially non-cytotoxic with only slight effect on cell proliferation and mild hepatotoxicity, which might be attributed to the nature of both antibiotics. In view of these results, these nanoparticles may be considered as a promising treatment for bone infections caused by enterobacteria, such as E. coli, and introduce a general strategy against bone infections based on the implementation of antibiotics with different but complementary activity into a single nanocarrier. STATEMENT OF SIGNIFICANCE: In this work, we propose a methodology to address E.coli bone infections by using moxifloxacin-loaded mesoporous silica nanoparticles coated with Arabic gum containing colistin (AG+CO-coated MX-loaded MSNs). The in vitro evaluation of this nanosystem demonstrated high affinity toward E. coli biofilm matrix thanks to the Arabic gum coating, a disaggregating and antibacterial effect of colistin, and a remarkable antibiofilm action because of the bactericidal ability of moxifloxacin and colistin. This anti-E. coli capacity of AG+CO-coated MX-loaded MSNs was brought out in an in vivo rabbit model of osteomyelitis where the nanosystem was able to eradicate more than 90% of the bacterial load within the infected bone.
Collapse
|
7
|
Liu J, Zhong J, Yang H, Wang D, Zhang Y, Yang Y, Xing G, Kon V. Biotic Supplements in Patients With Chronic Kidney Disease: Meta-Analysis of Randomized Controlled Trials. J Ren Nutr 2022; 32:10-21. [PMID: 34666930 PMCID: PMC9793596 DOI: 10.1053/j.jrn.2021.08.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 07/01/2021] [Accepted: 08/10/2021] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Gut flora imbalance characterizes patients with chronic kidney disease (CKD). Although biotic supplementation has been proposed to lessen inflammation and oxidative stress and, thus, reduce the risk of progressive kidney damage and cardiovascular disease, the effects remain controversial. We conducted a meta-analysis to assess the therapeutic benefits of biotics in CKD. METHODS PubMed, Embase, and Cochrane databases were systematically searched for randomized controlled trials that evaluated any biotic (prebiotic, probiotic, synbiotics) supplements in patients with CKD (CKD, stage 3-4 to end-stage renal disease). Primary endpoints included changes in renal function, markers of inflammation, and oxidative stress. Secondary endpoints included changes in levels of uremic toxins and variations in lipid metabolism. RESULTS Twenty-three eligible studies included 842 participants. In a pooled-analysis, biotics did not change estimated glomerular filtration rate (mean difference [MD] = 0.08, P = .92) or serum albumin (MD = -0.01, P = .86), although prebiotics reduced serum creatinine (standardized mean difference [SMD] = -0.23, P = .009) and blood urea nitrogen (MD = -6.05, P < .00001). Biotics improved total antioxidative capacity (SMD = 0.37, P = .007) and malondialdehyde (SMD = -0.96, P = .006) and reduced the inflammatory marker interleukin-6 (SMD = -0.30, P = .01) although not C-reactive protein (SMD = -0.22, P = .20). Biotic intervention reduced some uremic toxins, including p-cresol sulfate (SMD = -2.18, P < .0001) and indoxyl sulfate (MD = -5.14, P = .0009), which decreased in dialysis-dependent patients. Another toxin, indole-3-acetic acid (MD = -0.22, P = .63), did not change. Lipids were unaffected by biotic intervention (total cholesterol: SMD = -0.01, P = .89; high-density lipoprotein: SMD = -0.08, P = .76; low-density lipoprotein: MD = 3.54, P = .28; triglyceride: MD = -2.26, P = .58). CONCLUSION The results highlight the favorable influence of biotics on circulating markers of creatinine, oxidant stress (malondialdehyde, total antioxidative capacity), inflammation (interleukin-6), and uremic toxins (p-cresol sulfate) in patients with CKD. Biotics did not affect estimated glomerular filtration rate, albumin, indole-3-acetic acid, or lipids in either predialysis or dialysis patients.
Collapse
Affiliation(s)
- Jing Liu
- Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou University Institute of Nephrology, Zhengzhou, China.,Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - JianYong Zhong
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - HaiChun Yang
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - DongQin Wang
- Dongjing Town Health Service Center, Songjiang District, Shanghai, China
| | - Ying Zhang
- Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou University Institute of Nephrology, Zhengzhou, China
| | - YuMeng Yang
- Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou University Institute of Nephrology, Zhengzhou, China
| | - GuoLan Xing
- Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou University Institute of Nephrology, Zhengzhou, China
| | - Valentina Kon
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
8
|
Wang R, Li M, Strappe P, Zhou Z. Preparation, structural characteristics and physiological property of resistant starch. ADVANCES IN FOOD AND NUTRITION RESEARCH 2021; 95:1-40. [PMID: 33745510 DOI: 10.1016/bs.afnr.2020.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Starch is of the most important carbohydrates in human diets for maintaining normal body's energy metabolisms. However, due to the increased number of chronic diseases worldwide, the further study of the starch property in the dietary formula becomes essential for revealing its association with preventing or intervening the occurrence of such diseases as diabetes, obesity, intestinal diseases and even cardiovascular diseases. Considering that different starches demonstrate different digestion property based on their individual structural characteristics, in particular, the existence of resistant starch (RS) attracts much more interests recently because of its being a major producer of short-chain fatty acids followed by gut microbial fermentation. Furthermore, the understanding of the interaction between RS and microbiota in the gut and its substantial influence on the regulation of diabetes, kidney, disease hypertension and others is still being under investigated. Therefore, this chapter summarized the fine structure of starch, resistant starch structural characteristics, formation and preparation of resistant starches and their corresponding physiological property.
Collapse
Affiliation(s)
- Rui Wang
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, China
| | - Mei Li
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, China
| | - Padraig Strappe
- School of Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD, Australia
| | - Zhongkai Zhou
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, China; ARC Functional Grains Centre, Charles Sturt University, Wagga Wagga, NSW, Australia.
| |
Collapse
|
9
|
Paßlack N, Kohn B, Vahjen W, Zentek J. Effects of dietary cellobiose on the intestinal microbiota and excretion of nitrogen metabolites in healthy adult dogs. J Anim Physiol Anim Nutr (Berl) 2021; 105:569-578. [PMID: 33480132 DOI: 10.1111/jpn.13485] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 11/03/2020] [Indexed: 12/18/2022]
Abstract
In order to evaluate the potential prebiotic effects of cellobiose, 10 healthy adult research beagle dogs received a complete diet containing 0, 0.5 and 1 g cellobiose/kg bodyweight (BW)/day. At the end of each feeding period, faeces, urine and blood of the dogs were collected. The results demonstrated a significant increase of faecal lactate concentrations, indicating a bacterial fermentation of cellobiose in the canine intestine. Along with this, a dose-dependent linear increase of the relative abundance of Lactobacillaceae in the faeces of the dogs was observed (p = 0.014). In addition, a dose-dependent increase (p < 0.05) of Alloprevotella, Bacteroides and Prevotella, and a linear decrease for unidentified Lachnospiraceae (p = 0.011) was observed when cellobiose was added to the diet, although the relative abundance of these genera was low (<1%) among all groups. The faecal pH was not affected by dietary cellobiose. Cellobiose seemed to modulate the excretion of nitrogen metabolites, as lower concentrations of phenol (p = 0.034) and 4-ethylphenol (p = 0.002) in the plasma of the dogs were measured during the supplementation periods. Urinary phenols and indoles, however, were not affected by the dietary supplementation of cellobiose. In conclusion, cellobiose seems to be fermented by the intestinal microbiota of dogs. Although no effect on the faecal pH was detected, the observed increase of microbial lactate production might lower the pH in the large intestine and consecutively modulate the intestinal absorption of nitrogen metabolites. Also, the observed changes of some bacterial genera might have been mediated by increased intestinal lactate concentrations or a higher relative abundance of lactobacilli. Whether these results could be considered as a prebiotic effect and used as a dietetic strategy in diseased animals to improve gut function or hepatic and renal nitrogen metabolism should be evaluated in future studies.
Collapse
Affiliation(s)
- Nadine Paßlack
- Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany.,Small Animal Clinic, Faculty of Veterinary Medicine, Justus-Liebig-Universität Gießen, Gießen, Germany
| | - Barbara Kohn
- Small Animal Clinic, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Wilfried Vahjen
- Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Jürgen Zentek
- Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
10
|
Ikee R, Sasaki N, Yasuda T, Fukazawa S. Chronic Kidney Disease, Gut Dysbiosis, and Constipation: A Burdensome Triplet. Microorganisms 2020; 8:microorganisms8121862. [PMID: 33255763 PMCID: PMC7760012 DOI: 10.3390/microorganisms8121862] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022] Open
Abstract
Gut dysbiosis has been implicated in the progression of chronic kidney disease (CKD). Alterations in the gut environment induced by uremic toxins, the dietary restriction of fiber-rich foods, and multiple drugs may be involved in CKD-related gut dysbiosis. CKD-related gut dysbiosis is considered to be characterized by the expansion of bacterial species producing precursors of harmful uremic toxins, such as indoxyl sulfate and p-cresyl sulfate, and the contraction of species generating beneficial short-chain fatty acids, such as butyrate. Gut-derived uremic toxins cause oxidative stress and pro-inflammatory responses, whereas butyrate exerts anti-inflammatory effects and contributes to gut epithelial integrity. Gut dysbiosis is associated with the disruption of the gut epithelial barrier, which leads to the translocation of endotoxins. Research on CKD-related gut dysbiosis has mainly focused on chronic inflammation and consequent cardiovascular and renal damage. The pathogenic relationship between CKD-related gut dysbiosis and constipation has not yet been investigated in detail. Constipation is highly prevalent in CKD and affects the quality of life of these patients. Under the pathophysiological state of gut dysbiosis, altered bacterial fermentation products may play a prominent role in intestinal dysmotility. In this review, we outline the factors contributing to constipation, such as the gut microbiota and bacterial fermentation; introduce recent findings on the pathogenic link between CKD-related gut dysbiosis and constipation; and discuss potential interventions. This pathogenic link needs to be elucidated in more detail and may contribute to the development of novel treatment options not only for constipation, but also cardiovascular disease in CKD.
Collapse
Affiliation(s)
- Ryota Ikee
- Sapporo Nephrology Satellite Clinic, 9-2-15, Hassamu 6-jo, Nishi-ku, Sapporo 063-0826, Japan;
- Correspondence:
| | - Naomi Sasaki
- Sapporo Nephrology Clinic, 20-2-12, Nishimachikita, Nishi-ku, Sapporo 063-0061, Japan; (N.S.); (S.F.)
| | - Takuji Yasuda
- Sapporo Nephrology Satellite Clinic, 9-2-15, Hassamu 6-jo, Nishi-ku, Sapporo 063-0826, Japan;
| | - Sawako Fukazawa
- Sapporo Nephrology Clinic, 20-2-12, Nishimachikita, Nishi-ku, Sapporo 063-0061, Japan; (N.S.); (S.F.)
| |
Collapse
|
11
|
Ramos CI, Armani RG, Canziani MEF, Dalboni MA, Dolenga CJR, Nakao LS, Campbell KL, Cuppari L. Effect of prebiotic (fructooligosaccharide) on uremic toxins of chronic kidney disease patients: a randomized controlled trial. Nephrol Dial Transplant 2020; 34:1876-1884. [PMID: 29939302 DOI: 10.1093/ndt/gfy171] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/02/2018] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Microbial-derived uremic toxins, p-cresyl sulfate (PCS), indoxyl sulfate (IS) and indole 3-acetic acid (IAA), have been associated with the burden of chronic kidney disease (CKD). Prebiotics have emerged as an alternative to modulate the gut environment and to attenuate toxin production. This trial aims to investigate the effect of a prebiotic fructooligosaccharide (FOS) on uremic toxins of non-dialysis-dependent CKD (NDD-CKD) patients. METHODS A double-blind, placebo-controlled, randomized trial was conducted for 3 months. In all, 50 nondiabetic NDD-CKD patients [estimated glomerular filtration rate (eGFR) <45 mL/min/1.73 m2], aged 18-80 years, were allocated to prebiotic (FOS, 12 g/day) or placebo (maltodextrin, 12 g/day) groups. Primary outcomes were changes in serum (total and free) and urinary (total) PCS. Secondary outcomes included changes in IS, IAA, serum markers of intestinal permeability (zonulin), gut-trophic factors (epidermal growth factor and glucagon-like peptide-2), eGFR, inflammation (high sensitive c-reactive protein and interleukin-6), homeostatic model assessment-insulin resistance, lipid profile and gastrointestinal symptoms. RESULTS From 50 participants (54% men, 57.3 ± 14.6 years and eGFR 21.4 ± 7.6 mL/min/1.73 m2), 46 completed the follow-up. No changes in dietary intake or gastrointestinal symptoms were observed. There was a trend in the difference of serum total ΔPCS (treatment effect adjusted for baseline levels: -12.4 mg/L; 95% confidence interval (-5.6 to 0.9 mg/L; P = 0.07) and serum-free Δ%PCS [intervention -8.6 (-41.5 to 13.9%) versus placebo 3.5 (-28.8 to 85.5%); P = 0.07] between the groups. The trend in the difference of serum total ΔPCS was independent of eGFR and dietary protein:fiber ratio intake. No difference was found in urinary PCS. Aside from the decreased high-density lipoprotein cholesterol in the intervention, no differences were observed in the change of IS, IAA or other secondary outcome between the groups. CONCLUSIONS Our result suggests the potential of FOS in reducing serum total and free PCS in nondiabetic NDD-CKD patients.
Collapse
Affiliation(s)
- Christiane Ishikawa Ramos
- Nutrition Program, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.,Hospital do Rim - Fundação Oswaldo Ramos, São Paulo, Brazil.,Faculty of Health Sciences and Medicine, Bond University, Queensland, Australia
| | - Rachel Gatti Armani
- Hospital do Rim - Fundação Oswaldo Ramos, São Paulo, Brazil.,Division of Nephrology, UNIFESP, São Paulo, Brazil
| | | | - Maria Aparecida Dalboni
- Division of Nephrology, UNIFESP, São Paulo, Brazil.,Post-graduate Program in Medicine, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil
| | | | - Lia Sumie Nakao
- Department of Basic Pathology, Universidade Federal do Paraná (UFPR), Curitiba, Brazil
| | | | - Lilian Cuppari
- Nutrition Program, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.,Hospital do Rim - Fundação Oswaldo Ramos, São Paulo, Brazil.,Division of Nephrology, UNIFESP, São Paulo, Brazil
| |
Collapse
|
12
|
Meng Y, Bai H, Yu Q, Yan J, Zhao L, Wang S, Li Z, Wang Q, Chen L. High–Resistant Starch, Low-Protein Flour Intervention on Patients With Early Type 2 Diabetic Nephropathy: A Randomized Trial. J Ren Nutr 2019; 29:386-393. [DOI: 10.1053/j.jrn.2018.12.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 12/02/2018] [Accepted: 12/12/2018] [Indexed: 11/11/2022] Open
|
13
|
Cases A, Cigarrán-Guldrís S, Mas S, Gonzalez-Parra E. Vegetable-Based Diets for Chronic Kidney Disease? It Is Time to Reconsider. Nutrients 2019; 11:E1263. [PMID: 31167346 PMCID: PMC6627351 DOI: 10.3390/nu11061263] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/23/2019] [Accepted: 05/30/2019] [Indexed: 12/12/2022] Open
Abstract
Traditional dietary recommendations to renal patients limited the intake of fruits and vegetables because of their high potassium content. However, this paradigm is rapidly changing due to the multiple benefits derived from a fundamentally vegetarian diet such as, improvement in gut dysbiosis, reducing the number of pathobionts and protein-fermenting species leading to a decreased production of the most harmful uremic toxins, while the high fiber content of these diets enhances intestinal motility and short-chain fatty acid production. Metabolic acidosis in chronic kidney disease (CKD) is aggravated by the high consumption of meat and refined cereals, increasing the dietary acid load, while the intake of fruit and vegetables is able to neutralize the acidosis and its deleterious consequences. Phosphorus absorption and bioavailability is also lower in a vegetarian diet, reducing hyperphosphatemia, a known cause of cardiovascular mortality in CKD. The richness of multiple plants in magnesium and vitamin K avoids their deficiency, which is common in these patients. These beneficial effects, together with the reduction of inflammation and oxidative stress observed with these diets, may explain the reduction in renal patients' complications and mortality, and may slow CKD progression. Finally, although hyperkalemia is the main concern of these diets, the use of adequate cooking techniques can minimize the amount absorbed.
Collapse
Affiliation(s)
- Aleix Cases
- Medicine Department, Universitat de Barcelona, Institut d'Investigacions Biomèqiques August Pi i Sunyer, 08036 Barcelona, Spain.
| | | | - Sebastián Mas
- Servicio de Nefrología, Fundación Jiménez Díaz, 28040 Madrid, Spain.
- Centro de investigación en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain.
| | - Emilio Gonzalez-Parra
- Servicio de Nefrología, Fundación Jiménez Díaz, 28040 Madrid, Spain.
- Red de Investigación Renal (RedinRen), 28029 Madrid, Spain.
| |
Collapse
|
14
|
Noce A, Marrone G, Di Daniele F, Ottaviani E, Wilson Jones G, Bernini R, Romani A, Rovella V. Impact of Gut Microbiota Composition on Onset and Progression of Chronic Non-Communicable Diseases. Nutrients 2019; 11:nu11051073. [PMID: 31091761 PMCID: PMC6567014 DOI: 10.3390/nu11051073] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 02/07/2023] Open
Abstract
In recent years, mounting scientific evidence has emerged regarding the evaluation of the putative correlation between the gut microbiota composition and the presence of chronic non-communicable diseases (NCDs), such as diabetes mellitus, chronic kidney disease, and arterial hypertension. The aim of this narrative review is to examine the current literature with respect to the relationship between intestinal dysbiosis and the insurgence/progression of chronic NCDs, analyzing the physiopathological mechanisms that can induce microbiota modification in the course of these pathologies, and the possible effect induced by microbiota alteration upon disease onset. Therapy based on probiotics, prebiotics, synbiotics, postbiotics, and fecal microbiota transplant can represent a useful therapeutic tool, as has been highlighted on animal studies. To this moment, clinical studies that intended to demonstrate the beneficial effect induced by this kind of oral supplementation on the gut microbiota composition, and subsequent amelioration of signs and symptoms of chronic NCDs have been conducted on limited sample populations for a limited follow-up period. Therefore, to fully evaluate the therapeutic value of this kind of intervention, it would be ideal to design ample population; randomized clinical trials with a lengthy follow up period.
Collapse
Affiliation(s)
- Annalisa Noce
- UOC of Internal Medicine-Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome, via Montpellier 1, 00133 Rome, Italy.
| | - Giulia Marrone
- UOC of Internal Medicine-Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome, via Montpellier 1, 00133 Rome, Italy.
- PhD School of Applied Medical- Surgical Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy.
| | - Francesca Di Daniele
- UOC of Internal Medicine-Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome, via Montpellier 1, 00133 Rome, Italy.
| | - Eleonora Ottaviani
- UOC of Internal Medicine-Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome, via Montpellier 1, 00133 Rome, Italy.
| | - Georgia Wilson Jones
- UOC of Internal Medicine-Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome, via Montpellier 1, 00133 Rome, Italy.
| | - Roberta Bernini
- Department of Agriculture and Forest Sciences (DAFNE), University of Tuscia, 01100 Viterbo, Italy.
| | - Annalisa Romani
- PHYTOLAB-DISIA-Department of Informatics, Statistics and Applications G. Parenti, University of Florence, Viale Morgagni, 59-50134 Florence, Italy and QuMAP-PIN-Piazza Giovanni Ciardi, 25, 59100 Prato (PO), Italy.
| | - Valentina Rovella
- UOC of Internal Medicine-Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome, via Montpellier 1, 00133 Rome, Italy.
| |
Collapse
|
15
|
Snelson M, Kellow NJ, Coughlan MT. Modulation of the Gut Microbiota by Resistant Starch as a Treatment of Chronic Kidney Diseases: Evidence of Efficacy and Mechanistic Insights. Adv Nutr 2019; 10:303-320. [PMID: 30668615 PMCID: PMC6416045 DOI: 10.1093/advances/nmy068] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/17/2018] [Accepted: 08/12/2018] [Indexed: 12/14/2022] Open
Abstract
Chronic kidney disease (CKD) has been associated with changes in gut microbial ecology, or "dysbiosis," which may contribute to disease progression. Recent studies have focused on dietary approaches to favorably alter the composition of the gut microbial communities as a treatment method in CKD. Resistant starch (RS), a prebiotic that promotes proliferation of gut bacteria such as Bifidobacteria and Lactobacilli, increases the production of metabolites including short-chain fatty acids, which confer a number of health-promoting benefits. However, there is a lack of mechanistic insight into how these metabolites can positively influence renal health. Emerging evidence shows that microbiota-derived metabolites can regulate the incretin axis and mitigate inflammation via expansion of regulatory T cells. Studies from animal models and patients with CKD show that RS supplementation attenuates the concentrations of uremic retention solutes, including indoxyl sulfate and p-cresyl sulfate. Here, we present the current state of knowledge linking the microbiome to CKD, we explore the efficacy of RS in animal models of CKD and in humans with the condition, and we discuss how RS supplementation could be a promising dietary approach for slowing CKD progression.
Collapse
Affiliation(s)
- Matthew Snelson
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Nicole J Kellow
- Be Active Sleep & Eat (BASE) Facility, Department of Nutrition, Dietetics, and Food, Monash University, Notting Hill, Victoria, Australia
| | - Melinda T Coughlan
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Baker Heart Research Institute, Melbourne, Victoria, Australia
| |
Collapse
|
16
|
Mafra D, Borges N, Alvarenga L, Esgalhado M, Cardozo L, Lindholm B, Stenvinkel P. Dietary Components That May Influence the Disturbed Gut Microbiota in Chronic Kidney Disease. Nutrients 2019; 11:E496. [PMID: 30818761 PMCID: PMC6471287 DOI: 10.3390/nu11030496] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 02/20/2019] [Accepted: 02/22/2019] [Indexed: 12/11/2022] Open
Abstract
Gut microbiota imbalance is common in patients with chronic kidney disease (CKD) and associates with factors such as increased circulating levels of gut-derived uremic toxins, inflammation, and oxidative stress, which are linked to cardiovascular disease and increased morbimortality. Different nutritional strategies have been proposed to modulate gut microbiota, and could potentially be used to reduce dysbiosis in CKD. Nutrients like proteins, fibers, probiotics, and synbiotics are important determinants of the composition of gut microbiota and specific bioactive compounds such as polyphenols present in nuts, berries. and fruits, and curcumin, may also play a key role in this regard. However, so far, there are few studies on dietary components influencing the gut microbiota in CKD, and it is therefore not possible to conclude which nutrients should be prioritized in the diet of patients with CKD. In this review, we discuss some nutrients, diet patterns and bioactive compounds that may be involved in the modulation of gut microbiota in CKD and provide the background and rationale for studies exploring whether nutritional interventions with these dietary components could be used to alleviate the gut dysbiosis in patients with CKD.
Collapse
Affiliation(s)
- Denise Mafra
- Post Graduation Program in Medical Sciences, Federal Fluminense University (UFF), Niterói-Rio de Janeiro (RJ) 24220-900, Brazil.
- Post Graduation Program in Cardiovascular Sciences, Federal Fluminense University (UFF), Niterói-Rio de Janeiro (RJ) 24220-900, Brazil.
| | - Natália Borges
- Post Graduation Program in Cardiovascular Sciences, Federal Fluminense University (UFF), Niterói-Rio de Janeiro (RJ) 24220-900, Brazil.
| | - Livia Alvarenga
- Post Graduation Program in Medical Sciences, Federal Fluminense University (UFF), Niterói-Rio de Janeiro (RJ) 24220-900, Brazil.
| | - Marta Esgalhado
- Post Graduation Program in Cardiovascular Sciences, Federal Fluminense University (UFF), Niterói-Rio de Janeiro (RJ) 24220-900, Brazil.
| | - Ludmila Cardozo
- Post Graduation Program in Cardiovascular Sciences, Federal Fluminense University (UFF), Niterói-Rio de Janeiro (RJ) 24220-900, Brazil.
| | - Bengt Lindholm
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Technology and Intervention, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| | - Peter Stenvinkel
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Technology and Intervention, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
17
|
Jazani NH, Savoj J, Lustgarten M, Lau WL, Vaziri ND. Impact of Gut Dysbiosis on Neurohormonal Pathways in Chronic Kidney Disease. Diseases 2019; 7:diseases7010021. [PMID: 30781823 PMCID: PMC6473882 DOI: 10.3390/diseases7010021] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/29/2019] [Accepted: 02/08/2019] [Indexed: 02/06/2023] Open
Abstract
Chronic kidney disease (CKD) is a worldwide major health problem. Traditional risk factors for CKD are hypertension, obesity, and diabetes mellitus. Recent studies have identified gut dysbiosis as a novel risk factor for the progression CKD and its complications. Dysbiosis can worsen systemic inflammation, which plays an important role in the progression of CKD and its complications such as cardiovascular diseases. In this review, we discuss the beneficial effects of the normal gut microbiota, and then elaborate on how alterations in the biochemical environment of the gastrointestinal tract in CKD can affect gut microbiota. External factors such as dietary restrictions, medications, and dialysis further promote dysbiosis. We discuss the impact of an altered gut microbiota on neuroendocrine pathways such as the hypothalamus⁻pituitary⁻adrenal axis, the production of neurotransmitters and neuroactive compounds, tryptophan metabolism, and the cholinergic anti-inflammatory pathway. Finally, therapeutic strategies including diet modification, intestinal alpha-glucosidase inhibitors, prebiotics, probiotics and synbiotics are reviewed.
Collapse
Affiliation(s)
- Nima H Jazani
- Division of Nephrology, Department of Medicine, University of California-Irvine, Irvine, CA 92697, USA.
| | - Javad Savoj
- Department of Internal Medicine, Riverside Community Hospital, University of California-Riverside School of Medicine, Riverside, CA 92501, USA.
| | - Michael Lustgarten
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA.
| | - Wei Ling Lau
- Division of Nephrology, Department of Medicine, University of California-Irvine, Irvine, CA 92697, USA.
| | - Nosratola D Vaziri
- Division of Nephrology, Department of Medicine, University of California-Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
18
|
McFarlane C, Ramos CI, Johnson DW, Campbell KL. Prebiotic, Probiotic, and Synbiotic Supplementation in Chronic Kidney Disease: A Systematic Review and Meta-analysis. J Ren Nutr 2018; 29:209-220. [PMID: 30366767 DOI: 10.1053/j.jrn.2018.08.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/13/2018] [Accepted: 08/13/2018] [Indexed: 01/27/2023] Open
Abstract
OBJECTIVE Gut dysbiosis has been implicated in the pathogenesis of chronic kidney disease (CKD). Restoring gut microbiota with prebiotic, probiotic, and synbiotic supplementation has emerged as a potential therapeutic intervention but has not been systematically evaluated in the CKD population. DESIGN AND METHODS This is a systematic review. A structured search of MEDLINE, CINAHL, EMBASE, Cochrane Central Register of Controlled Trials, and the International Clinical Trials Register Search Portal was conducted for articles published since inception until July 2017. Included studies were randomized controlled trials investigating the effects of prebiotic, probiotic, and/or synbiotic supplementation (>1 week) on uremic toxins, microbiota profile, and clinical and patient-centered outcomes in adults and children with CKD. RESULTS Sixteen studies investigating 645 adults met the inclusion criteria; 5 investigated prebiotics, 6 probiotics, and 5 synbiotics. The quality of the studies (Grades of Recommendation, Assessment, Development and Evaluation) ranged from moderate to very low. Prebiotic, probiotic, and synbiotic supplementation may have led to little or no difference in serum urea (9 studies, 345 participants: mean difference [MD] -0.30 mmol/L, 95% confidence interval [CI] -2.20 to 1.61, P = .76, I2 = 53%), indoxyl sulfate (4 studies, 144 participants: MD -0.02 mg/dL, 95% CI -0.09 to 0.05, P = .61, I2 = 0%), and p-cresyl sulfate (4 studies, 144 participants: MD -0.13 mg/dL, 95% CI -0.41 to 0.15, P = .35, I2 = 0%). Prebiotic supplementation may have slightly reduced serum urea concentration (4 studies, 105 participants: MD -2.23 mmol/L, 95% CI -3.83 to -0.64, P = .006, I2 = 11). Of the 2 studies investigating microbiota changes, synbiotic interventions significantly increased Bifidobacterium. Supplement effects on clinical outcomes were uncertain. CONCLUSIONS There is limited evidence to support the use of prebiotics, probiotics, and/or synbiotics in CKD management.
Collapse
Affiliation(s)
- Catherine McFarlane
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, Queensland, Australia; School of Medicine, University of Queensland, Brisbane, Queensland, Australia; Renal Department, Sunshine Coast University Hospital, Birtinya, Queensland, Australia.
| | - Christiane I Ramos
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, Queensland, Australia; Nutrition Programme, Federal University of Sao Paulo, Sao Paulo, Brazil; Department of Nutrition and Dietetics, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - David W Johnson
- School of Medicine, University of Queensland, Brisbane, Queensland, Australia; Department of Nephrology, Princess Alexandra Hospital, Brisbane, Queensland, Australia; Translational Research Institute, Brisbane, QLD, Australia
| | - Katrina L Campbell
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, Queensland, Australia; Department of Nutrition and Dietetics, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
19
|
Biotic Supplements for Renal Patients: A Systematic Review and Meta-Analysis. Nutrients 2018; 10:nu10091224. [PMID: 30181461 PMCID: PMC6165363 DOI: 10.3390/nu10091224] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/28/2018] [Accepted: 08/28/2018] [Indexed: 12/11/2022] Open
Abstract
Intestinal dysbiosis is highly pervasive among chronic kidney disease (CKD) patients and may play a key role in disease progression and complications. We performed a systematic review and meta-analysis to evaluate effects of biotic supplements on a large series of outcomes in renal patients. Ovid-MEDLINE, PubMed and CENTRAL databases were searched for randomized controlled trials (RCTs) comparing any biotic (pre-, pro- or synbiotics) to standard therapy or placebo. Primary endpoints were change in renal function and cardiovascular events; secondary endpoints were change in proteinuria/albuminuria, inflammation, uremic toxins, quality of life and nutritional status. Seventeen eligible studies (701 participants) were reviewed. Biotics treatment did not modify estimated glomerular filtration rate (eGFR) (mean difference (MD) 0.34 mL/min/1.73 m2; 95% CI −0.19, 0.86), serum creatinine (MD −0.13 mg/dL; 95% confidence interval (CI) −0.32, 0.07), C-reactive protein (MD 0.75 mg/dL; 95% CI −1.54, 3.03) and urea (standardized MD (SMD) −0.02; 95% CI −0.25, 0.20) as compared to control. Outcome data on the other endpoints of interest were lacking, sparse or in an unsuitable format to be analyzed collectively. According to the currently available evidence, there is no conclusive rationale for recommending biotic supplements for improving outcomes in renal patients. Large-scale, well-designed and adequately powered studies focusing on hard rather than surrogate outcomes are still awaited.
Collapse
|
20
|
Koppe L, Fouque D, Soulage CO. The Role of Gut Microbiota and Diet on Uremic Retention Solutes Production in the Context of Chronic Kidney Disease. Toxins (Basel) 2018; 10:toxins10040155. [PMID: 29652797 PMCID: PMC5923321 DOI: 10.3390/toxins10040155] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 02/07/2023] Open
Abstract
Uremic retention solutes (URS) are associated with cardiovascular complications and poor survival in chronic kidney disease. The better understanding of the origin of a certain number of these toxins enabled the development of new strategies to reduce their production. URS can be classified according to their origins (i.e., host, microbial, or exogenous). The discovery of the fundamental role that the intestinal microbiota plays in the production of many URS has reinstated nutrition at the heart of therapeutics to prevent the accumulation of URS and their deleterious effects. The intestinal microbiota is personalized and is strongly influenced by dietary habits, such as the quantity and the quality of dietary protein and fibers. Herein, this review out lines the role of intestinal microbiota on URS production and the recent discoveries on the effect of diet composition on the microbial balance in the host with a focus on the effect on URS production.
Collapse
Affiliation(s)
- Laetitia Koppe
- Department Nephrology, Centre Hospitalier Lyon Sud, F-69495 Pierre-Benite, France.
- CarMeN Lab, INSA-Lyon, INSERM U1060, INRA, University Lyon 1, F-69621 Villeurbanne, France.
| | - Denis Fouque
- Department Nephrology, Centre Hospitalier Lyon Sud, F-69495 Pierre-Benite, France.
- CarMeN Lab, INSA-Lyon, INSERM U1060, INRA, University Lyon 1, F-69621 Villeurbanne, France.
| | - Christophe O Soulage
- CarMeN Lab, INSA-Lyon, INSERM U1060, INRA, University Lyon 1, F-69621 Villeurbanne, France.
| |
Collapse
|
21
|
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]
|
22
|
Sabatino A, Regolisti G, Gandolfini I, Delsante M, Fani F, Gregorini MC, Fiaccadori E. Diet and enteral nutrition in patients with chronic kidney disease not on dialysis: a review focusing on fat, fiber and protein intake. J Nephrol 2017; 30:743-754. [PMID: 28884267 DOI: 10.1007/s40620-017-0435-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 08/24/2017] [Indexed: 02/07/2023]
Abstract
The clinical data available on dietary requirements of patients with chronic kidney disease (CKD) not on dialysis are limited and largely inconclusive in terms of the renal, cardiovascular and nutritional outcomes achievable through dietary modifications. Restriction of protein intake during the early stages of CKD may in fact slow its progression, but at the same time this approach may also lead to protein-energy wasting, if energy intake is not adequate and properly monitored. Unfortunately, compliance to dietary recommendations is traditionally low in this patient population. A switch from saturated to mono- and polyunsaturated fats is generally recognized as advantageous for cardiac health; however, the benefits in term of renal function are largely unknown. Similarly, the association between dietary fiber intake and kidney disease is largely unknown. In fact, while there is evidence on the positive health effects of dietary fibers in the general population, nutritional guidelines for CKD lack formal recommendations concerning fiber intake. This paper reviews data and evidence from clinical trials and meta-analyses on renal and cardiovascular outcomes related to modifications in protein, fat and fiber intake. Suggestions for maintaining nutritional status through patient-oriented dietary patterns and enteral supplementation in CKD patients on conservative therapy are also presented.
Collapse
Affiliation(s)
- Alice Sabatino
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43100, Parma, Italy
| | - Giuseppe Regolisti
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43100, Parma, Italy
| | - Ilaria Gandolfini
- Postgraduate School of Nephrology, University of Parma, Parma, Italy
| | - Marco Delsante
- Postgraduate School of Nephrology, University of Parma, Parma, Italy
| | - Filippo Fani
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43100, Parma, Italy
| | | | - Enrico Fiaccadori
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43100, Parma, Italy. .,Postgraduate School of Nephrology, University of Parma, Parma, Italy.
| |
Collapse
|
23
|
Koh GY, Rowling MJ. Resistant starch as a novel dietary strategy to maintain kidney health in diabetes mellitus. Nutr Rev 2017; 75:350-360. [DOI: 10.1093/nutrit/nux006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
24
|
|
25
|
Kieffer DA, Piccolo BD, Vaziri ND, Liu S, Lau WL, Khazaeli M, Nazertehrani S, Moore ME, Marco ML, Martin RJ, Adams SH. Resistant starch alters gut microbiome and metabolomic profiles concurrent with amelioration of chronic kidney disease in rats. Am J Physiol Renal Physiol 2016; 310:F857-71. [PMID: 26841824 DOI: 10.1152/ajprenal.00513.2015] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 01/28/2016] [Indexed: 02/06/2023] Open
Abstract
Patients and animals with chronic kidney disease (CKD) exhibit profound alterations in the gut environment including shifts in microbial composition, increased fecal pH, and increased blood levels of gut microbe-derived metabolites (xenometabolites). The fermentable dietary fiber high amylose maize-resistant starch type 2 (HAMRS2) has been shown to alter the gut milieu and in CKD rat models leads to markedly improved kidney function. The aim of the present study was to identify specific cecal bacteria and cecal, blood, and urinary metabolites that associate with changes in kidney function to identify potential mechanisms involved with CKD amelioration in response to dietary resistant starch. Male Sprague-Dawley rats with adenine-induced CKD were fed a semipurified low-fiber diet or a high-fiber diet [59% (wt/wt) HAMRS2] for 3 wk (n = 9 rats/group). The cecal microbiome was characterized, and cecal contents, serum, and urine metabolites were analyzed. HAMRS2-fed rats displayed decreased cecal pH, decreased microbial diversity, and an increased Bacteroidetes-to-Firmicutes ratio. Several uremic retention solutes were altered in the cecal contents, serum, and urine, many of which had strong correlations with specific gut bacteria abundances, i.e., serum and urine indoxyl sulfate were reduced by 36% and 66%, respectively, in HAMRS2-fed rats and urine p-cresol was reduced by 47% in HAMRS2-fed rats. Outcomes from this study were coincident with improvements in kidney function indexes and amelioration of CKD outcomes previously reported for these rats, suggesting an important role for microbial-derived factors and gut microbe metabolism in regulating host kidney function.
Collapse
Affiliation(s)
- Dorothy A Kieffer
- Obesity and Metabolism Research Unit, United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California; Graduate Group in Nutritional Biology and Department of Nutrition, University of California, Davis, California
| | - Brian D Piccolo
- Arkansas Children's Nutrition Center and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | - Shuman Liu
- Division of Nephrology, University of California, Irvine, California
| | - Wei L Lau
- Division of Nephrology, University of California, Irvine, California
| | - Mahyar Khazaeli
- Division of Nephrology, University of California, Irvine, California
| | | | - Mary E Moore
- Department of Food Science and Technology, University of California, Davis, California; and
| | - Maria L Marco
- Department of Food Science and Technology, University of California, Davis, California; and
| | - Roy J Martin
- Obesity and Metabolism Research Unit, United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California
| | - Sean H Adams
- Graduate Group in Nutritional Biology and Department of Nutrition, University of California, Davis, California; Arkansas Children's Nutrition Center and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
26
|
Resistant starch for modulation of gut microbiota: Promising adjuvant therapy for chronic kidney disease patients? Eur J Nutr 2016; 55:1813-21. [PMID: 26830416 DOI: 10.1007/s00394-015-1138-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 12/11/2015] [Indexed: 02/07/2023]
Abstract
The gut microbiota has been extensively studied in all health science fields because its imbalance is linked to many disorders, such as inflammation and oxidative stress, thereby contributing to cardiovascular disease, obesity, diabetes and chronic kidney disease (CKD) complications. Novel therapeutic strategies that aim to reduce the complications caused by this imbalance have increased in recent years. Studies have shown that prebiotic supplementation can beneficially modulate the gut microbiota in CKD patients. Prebiotics consist of non-digestible dietary soluble fiber, which acts as a substrate for the gut microbiota. Resistant starch (RS) is a type of dietary fiber that can reach the large bowel and act as a substrate for microbial fermentation; for these reasons, it has been considered to be a prebiotic. Few studies have analyzed the effects of RS on the gut microbiota in CKD patients. This review discusses recent information about RS and the potential role of the gut microbiota, with a particular emphasis on CKD patients.
Collapse
|
27
|
Vanholder R, Glorieux G. The intestine and the kidneys: a bad marriage can be hazardous. Clin Kidney J 2015; 8:168-79. [PMID: 25815173 PMCID: PMC4370304 DOI: 10.1093/ckj/sfv004] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/13/2015] [Indexed: 12/20/2022] Open
Abstract
The concept that the intestine and chronic kidney disease influence each other, emerged only recently. The problem is multifaceted and bidirectional. On one hand, the composition of the intestinal microbiota impacts uraemic retention solute production, resulting in the generation of essentially protein-bound uraemic toxins with strong biological impact such as vascular damage and progression of kidney failure. On the other hand, the uraemic status affects the composition of intestinal microbiota, the generation of uraemic retention solutes and their precursors and causes disturbances in the protective epithelial barrier of the intestine and the translocation of intestinal microbiota into the body. All these elements together contribute to the disruption of the metabolic equilibrium and homeostasis typical to uraemia. Several measures with putative impact on intestinal status have recently been tested for their influence on the generation or concentration of uraemic toxins. These include dietary measures, prebiotics, probiotics, synbiotics and intestinal sorbents. Unfortunately, the quality and the evidence base of many of these studies are debatable, especially in uraemia, and often results within one study or among studies are contradictory. Nevertheless, intestinal uraemic metabolite generation remains an interesting target to obtain in the future as an alternative or additive to dialysis to decrease uraemic toxin generation. In the present review, we aim to summarize (i) the role of the intestine in uraemia by producing uraemic toxins and by generating pathophysiologically relevant changes, (ii) the role of uraemia in modifying intestinal physiology and (iii) the therapeutic options that could help to modify these effects and the studies that have assessed the impact of these therapies.
Collapse
Affiliation(s)
- Raymond Vanholder
- Nephrology Section, 0K12 , University Hospital , Ghent B9000 , Belgium
| | - Griet Glorieux
- Nephrology Section, 0K12 , University Hospital , Ghent B9000 , Belgium
| |
Collapse
|
28
|
Dietary fiber effects in chronic kidney disease: a systematic review and meta-analysis of controlled feeding trials. Eur J Clin Nutr 2014; 69:761-8. [DOI: 10.1038/ejcn.2014.237] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 09/11/2014] [Accepted: 09/21/2014] [Indexed: 11/08/2022]
|
29
|
Salmean YA, Segal MS, Palii SP, Dahl WJ. Fiber supplementation lowers plasma p-cresol in chronic kidney disease patients. J Ren Nutr 2014; 25:316-20. [PMID: 25446837 DOI: 10.1053/j.jrn.2014.09.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 09/01/2014] [Accepted: 09/16/2014] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVE To determine the effects of supplemental fiber on plasma p-cresol, stool frequency, and quality of life (QoL) in chronic kidney disease (CKD) patients. DESIGN AND SETTING In a 12-week single-blind study, participants were provided with control muffins and supplements (5.5 g sucrose/day) for 2 weeks, muffins containing 10 g/day pea hull fiber and control supplements for 4 weeks, and muffins with 10 g/day pea hull fiber and 15 g/day inulin as a supplement for 6 weeks. SUBJECTS Individuals with CKD (n = 13; 6 males, 7 females; aged 65 ± 3 years; estimated glomerular filtration rate <50 mL/minute/1.73(2)) completed the study. MAIN OUTCOME MEASURES Plasma p-cresol was determined by gas chromatography-mass spectrometry, stool frequency by 5-day journals, and QoL by the KDQOL-36™. RESULTS Plasma p-cresol decreased from 7.25 ± 1.74 mg/L during week 1 to 5.82 ± 1.72 mg/L during week 12 (P < .05), and in participants with high compliance (>70% inulin intake), from 6.71 ± 1.98 mg/L to 4.22 ± 1.16 mg/L (P < .05). Total fiber intake increased from 16.6 ± 1.7 g/day during control to 26.5 ± 2.4 g/day (P < .0001) with the added pea hull and to 34.5 ± 2.2 g/day with pea hull and inulin (P < .0001). Stool frequency increased from 1.4 ± 0.2 stools/day during control to 1.9 ± 0.3 stools/day during both fiber periods (P < .05). No change in overall QoL was observed. CONCLUSIONS Supplementing the diet of CKD patients with fiber may be a dietary therapy to reduce p-cresol and improve stool frequency.
Collapse
Affiliation(s)
- Younis A Salmean
- Food Science and Human Nutrition Department, University of Florida, Gainesville, Florida
| | - Mark S Segal
- North Florida/South Georgia Veterans Health System, Gainesville, Florida; Division of Nephrology, Hypertension and Renal Transplantation, University of Florida, Gainesville, Florida
| | - Sergiu P Palii
- Clinical and Translational Science Institute, College of Medicine, University of Florida, Gainesville, Florida
| | - Wendy J Dahl
- Food Science and Human Nutrition Department, University of Florida, Gainesville, Florida.
| |
Collapse
|
30
|
Abstract
Prior to the availability of hemodialysis, dietary protein restriction played a large part in the treatment of uremia. This therapy was based on observations that uremic symptoms increased with high protein intake. Early investigators thus presumed that "uremic toxins" were derived from the breakdown of dietary protein; its restriction improved uremic symptoms but caused malnutrition. After the availability of hemodialysis, protein restriction was no longer recommended. Studies in healthy subjects have shown that an intake of 0.6-0.8 g/kg/day is adequate to prevent protein malnutrition. Guidelines for hemodialysis patients, however, currently recommend higher protein intakes of 1.2 g/kg/day. A downside to higher intake may be increased production of protein-derived uremic solutes that caused the symptoms observed by early investigators. Some of these solutes are produced by colon microbes acting on protein which escapes digestion in the small intestine. Increasing dietary fiber may reduce the production of colon-derived solutes in hemodialysis patients without adverse effects of protein restriction. Fiber comprises carbohydrates and related substances that are resistant to digestion in the small intestine. Upon delivery to the colon, fiber is broken down to short chain fatty acids, providing energy to both the microbes and the host. With an increased energy supply, the microbes can incorporate dietary protein for growth rather than breaking them down to uremic solutes. Increasing fiber intake in hemodialysis patients has been shown to reduce the plasma levels of selected colon-derived solutes. Further studies are needed to test whether this provides clinical benefit.
Collapse
Affiliation(s)
- Tammy L Sirich
- Departments of Medicine, VA Palo Alto Health Care System and Stanford University, Palo Alto, California
| |
Collapse
|
31
|
Xu H, Huang X, Risérus U, Krishnamurthy VM, Cederholm T, Arnlöv J, Lindholm B, Sjögren P, Carrero JJ. Dietary fiber, kidney function, inflammation, and mortality risk. Clin J Am Soc Nephrol 2014; 9:2104-10. [PMID: 25280496 DOI: 10.2215/cjn.02260314] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND AND OBJECTIVES In the United States population, high dietary fiber intake has been associated with a lower risk of inflammation and mortality in individuals with kidney dysfunction. This study aimed to expand such findings to a Northern European population. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Dietary fiber intake was calculated from 7-day dietary records in 1110 participants aged 70-71 years from the Uppsala Longitudinal Study of Adult Men (examinations performed during 1991-1995). Dietary fiber was adjusted for total energy intake by the residual method. Renal function was estimated from the concentration of serum cystatin C, and deaths were registered prospectively during a median follow-up of 10.0 years. RESULTS Dietary fiber independently and directly associated with eGFR (adjusted difference, 2.6 ml/min per 1.73 m(2) per 10 g/d higher; 95% confidence interval [95% CI], 0.3 to 4.9). The odds of C-reactive protein >3 mg/L were lower (linear trend, P=0.002) with higher fiber quartiles. During follow-up, 300 participants died (incidence rate of 2.87 per 100 person-years at risk). Multiplicative interactions were observed between dietary fiber intake and kidney dysfunction in the prediction of mortality. Higher dietary fiber was associated with lower mortality in unadjusted analysis. These associations were stronger in participants with kidney dysfunction (eGFR<60 ml/min per 1.73 m(2)) (hazard ratio [HR], 0.58; 95% CI, 0.35 to 0.98) than in those without (HR, 1.30; 95% CI, 0.76 to 2.22; P value for interaction, P=0.04), and were mainly explained by a lower incidence of cancer-related deaths (0.25; 95% CI, 0.10 to 0.65) in individuals with kidney dysfunction versus individuals with an eGFR≥60 ml/min per 1.73 m(2) (1.61; 95% CI, 0.69 to 3.74; P value for interaction, P=0.01). CONCLUSIONS High dietary fiber was associated with better kidney function and lower inflammation in community-dwelling elderly men from Sweden. High dietary fiber was also associated with lower (cancer) mortality risk, especially in individuals with kidney dysfunction.
Collapse
Affiliation(s)
- Hong Xu
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention, and Technology, and Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoyan Huang
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention, and Technology, and Division of Nephrology, Peking University Shenzhen Hospital, Peking University, Shenzhen, China
| | - Ulf Risérus
- Sections of Clinical Nutrition and Metabolism and
| | - Vidya M Krishnamurthy
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention, and Technology, and
| | | | - Johan Arnlöv
- Geriatrics, Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden; and School of Health and Social Studies, Dalarna University, Falun, Sweden
| | - Bengt Lindholm
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention, and Technology, and
| | - Per Sjögren
- Sections of Clinical Nutrition and Metabolism and
| | - Juan Jesús Carrero
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention, and Technology, and Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden;
| |
Collapse
|
32
|
Fang CY, Lu JR, Chen BJ, Wu C, Chen YP, Chen MJ. Selection of uremic toxin-reducing probiotics in vitro and in vivo. J Funct Foods 2014. [DOI: 10.1016/j.jff.2014.01.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
33
|
Nonextracorporeal Methods for Decreasing Uremic Solute Concentration: A Future Way To Go? Semin Nephrol 2014; 34:228-43. [DOI: 10.1016/j.semnephrol.2014.02.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
34
|
Salmean YA, Zello GA, Dahl WJ. Foods with added fiber improve stool frequency in individuals with chronic kidney disease with no impact on appetite or overall quality of life. BMC Res Notes 2013; 6:510. [PMID: 24304924 PMCID: PMC4235217 DOI: 10.1186/1756-0500-6-510] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 11/29/2013] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Fiber intake may be low in individuals with chronic kidney disease (CKD) due to diet restriction and/or poor appetite associated with uremic symptoms, contributing to constipation and reduced quality of life. This report describes the effects of foods with added fiber on gastrointestinal function and symptoms, clinical markers, and quality of life in CKD patients. FINDINGS Adults with CKD (n = 15; 9 F, 6 M; 66 ± 15 y) were provided with cereal, cookies and snack bars without added fiber for 2 weeks, followed by similar foods providing 23 g/d of added fiber for 4 weeks, to incorporate into their usual diets. Participants completed the Kidney Disease Quality of Life (KDQOL-36) questionnaire, the Simplified Nutritional Appetite Questionnaire (SNAQ) and the Epworth Sleepiness Scale (ESS) bi-weekly, the Gastrointestinal Symptom Rating Scale (GSRS) weekly, and daily stool frequency and compliance. Control and intervention serum cholesterol and glucose were assessed. Providing 23 g/d of added fiber increased stool frequency (1.3 ± 0.2 to 1.6 ± 0.2 stools/d; P = 0.02), decreased total cholesterol (175 ± 12 to 167 ± 11 mg/dL; P = 0.02) and improved TC:HDL ratio (4.0 ± 0.3 to 3.7 ± 0.2; P = 0.02). GSRS and SNAQ scores did not change, but SNAQ scores suggested poor appetite in 7 participants with or without added fiber. KDQOL Mental Health Composite decreased from 53 ± 2 to 48 ± 2 (P = 0.01) while Physical Health Composite increased from 31 ± 2 to 35 ± 3 (P = 0.02), with no change in overall QOL. The ESS score decreased from 10 ± 1 to 8 ± 1 (P = 0.04). CONCLUSION Consuming foods with added fiber may be an effective means of increasing fiber intakes, improving stool frequency, and lipid profile in individuals with CKD. TRIAL REGISTRATION ClinicalTrials.gov, # NCT01842087.
Collapse
Affiliation(s)
- Younis A Salmean
- Food Science and Human Nutrition, University of Florida, 359 FSHN Building Newell Drive, Gainesville, FL 32611, USA
| | - Gordon A Zello
- College of Pharmacy and Nutrition, University of Saskatchewan, 110 Science Place, Saskatoon SK S7N 5C9, Canada
| | - Wendy J Dahl
- Food Science and Human Nutrition, University of Florida, 359 FSHN Building Newell Drive, Gainesville, FL 32611, USA
| |
Collapse
|
35
|
Choi HJ, Kim EJ, Shin YW, Park JH, Kim DH, Kim NJ. Protective Effect of Heat-processed Ginseng (Sun Ginseng) in the Adenine-induced Renal Failure Rats. J Ginseng Res 2013; 36:270-6. [PMID: 23717128 PMCID: PMC3659590 DOI: 10.5142/jgr.2012.36.3.270] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 05/03/2012] [Accepted: 05/03/2012] [Indexed: 11/18/2022] Open
Abstract
The effect of orally administered sun ginseng (SG), which is a ginseng processed by steaming, was examined in adenine-induced chronic renal failure rat. SG significantly decreased both blood urea nitrogen and serum creatinine levels, indicating an improvement of renal function. Also, SG significantly increased the urinary excretion of both urea and creatinine. Furthermore it lowered the blood pressure, and inhibited adenine-induced kidney hypertrophy and edema. Based on these findings, SG may ameliorate chronic renal failures.
Collapse
Affiliation(s)
- Hyuck Jae Choi
- East-West Medical Research Institute, Kyung Hee University Medical Center, Seoul 130-702, Korea
| | | | | | | | | | | |
Collapse
|
36
|
Poesen R, Meijers B, Evenepoel P. The colon: an overlooked site for therapeutics in dialysis patients. Semin Dial 2013; 26:323-32. [PMID: 23458264 DOI: 10.1111/sdi.12082] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Morbidity and mortality related to chronic kidney disease remain unacceptably high, despite tremendous progress in its prevention and treatment. In an ongoing quest to improve outcome in chronic kidney disease patients, the colon might be an appealing, but largely underexplored, therapeutic target. A clear bi-directional functional relationship exists between the colon and kidney, also referred as to the colo-renal axis. Uremia has an important impact on the colonic microbiome. The microbiome, in turn, is an important source of uremic toxins, with p-cresyl sulfate and indoxyl sulfate as important prototypes. These co-metabolites accumulate in the face of a falling kidney function, and may accelerate the progression of renal and cardiovascular disease. Several therapeutic interventions, including prebiotics and adsorbants, specifically target these colon-derived uremic toxins originating from bacterial metabolism. As kidney function declines, the colon also gains importance in the homeostasis and disposal of potassium and oxalate. Their colonic secretion may be increased by drugs increasing the expression of cAMP and by probiotics (e.g., Oxalobacter formigenes).
Collapse
Affiliation(s)
- Ruben Poesen
- Division of Nephrology, Department of Microbiology and Immunology, University Hospitals Leuven, Leuven, Belgium
| | | | | |
Collapse
|
37
|
Salmean YA, Segal MS, Langkamp-Henken B, Canales MT, Zello GA, Dahl WJ. Foods With Added Fiber Lower Serum Creatinine Levels in Patients With Chronic Kidney Disease. J Ren Nutr 2013; 23:e29-32. [DOI: 10.1053/j.jrn.2012.04.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 03/22/2012] [Accepted: 04/04/2012] [Indexed: 11/11/2022] Open
|
38
|
Røjen BA, Larsen M, Kristensen NB. Effect of abomasal infusion of oligofructose on portal-drained visceral ammonia and urea-nitrogen fluxes in lactating Holstein cows. J Dairy Sci 2012; 95:7248-60. [PMID: 22999277 DOI: 10.3168/jds.2012-5558] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 07/22/2012] [Indexed: 11/19/2022]
Abstract
The effects of abomasal infusion of oligofructose in lactating dairy cows on the relationship between hindgut fermentation and N metabolism, and its effects on NH(3) absorption and transfer of blood urea-N across the portal-drained viscera versus ruminal epithelia were investigated. Nine lactating Holstein cows fitted with ruminal cannulas and permanent indwelling catheters in major splanchnic blood vessels were used in an unbalanced crossover design with 14-d periods. Treatments were continuous abomasal infusion of water or 1,500 g/d of oligofructose. The same basal diet was fed with both treatments. Eight sample sets of arterial, portal, hepatic, and ruminal vein blood, ruminal fluid, and urine were obtained at 0.5h before the morning feeding and at 0.5, 1.5, 2.5, 3.5, 4.5, 5.5, and 6.5 h after feeding. It was hypothesized that an increased supply of fermentable substrate to the hindgut would increase the uptake of urea-N from blood to the hindgut at the expense of urea-N uptake to the forestomach. The study showed that abomasal oligofructose infusion decreased the total amount of urea-N transferred from the blood to the gut, NH(3) absorption, and arterial blood urea-N concentration. Subsequently, hepatic NH(3) uptake and urea-N production also decreased with oligofructose infusion. Additionally, urea-N concentration in milk and urinary N excretion decreased with oligofructose treatment. The oligofructose infusion did not affect ruminal NH(3) concentrations or any other ruminal variables, nor did it affect ruminal venous - arterial concentration differences for urea-N and NH(3). The oligofructose treatment did not affect milk yield, but did decrease apparent digestibility of OM, N, and starch. Nitrogen excreted in the feces was greater with the oligofructose infusion. In conclusion, the present data suggest that increased hindgut fermentation did not upregulate urea-N transfer to the hindgut at the expense of urea-N uptake by the rumen, and the observed reduction in arterial blood urea-N concentration appeared not to be due to increased urea-N transport, but rather could be explained by reduced NH(3) input to hepatic urea-N synthesis caused by increased sequestration of NH(3) in the hindgut and excretion in feces. Increasing the hindgut fermentation in lactating dairy cows by abomasal infusion of 1,500 g/d of oligofructose shifted some N excretion from the urine to feces and possibly reduced manure NH(3) volatilization without impairing rumen fermentation.
Collapse
Affiliation(s)
- B A Røjen
- Department of Animal Science, Aarhus University, Foulum, DK-8830 Tjele, Denmark.
| | | | | |
Collapse
|
39
|
Ali BH, Al-Salam S, Al Husseni I, Kayed RR, Al-Masroori N, Al-Harthi T, Al Zaabi M, Nemmar A. Effects of Gum Arabic in rats with adenine-induced chronic renal failure. Exp Biol Med (Maywood) 2010; 235:373-82. [PMID: 20404056 DOI: 10.1258/ebm.2009.009214] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Gum Arabic (GA [Acacia senegal]) is reputed, in Arabian medicinal practices, to be useful in treating patients with chronic renal failure (CRF), albeit without strong scientific evidence. We have previously shown that GA had no significant effect in rats with CRF induced by surgical nephrectomy. Here, we used another animal model of human CRF (feeding adenine at a concentration of 0.75%(w/w) for four weeks) to test the effect of GA on CRF. Renal morphology and measurements of plasma concentrations of urea and creatinine (Cr), and Cr clearance, in addition to urinary volume, osmolarity and protein concentrations, and N-acetylglucosamine and lactate dehydrogenase activities were performed. Interleukin-6 and the total antioxidant levels in urine, as well as the activity of superoxide dismutase in renal tissues, were estimated. Adenine feeding resulted in marked renal damage. GA (6%(w/v) and 12%(w/v) in drinking water for four consecutive weeks) significantly ameliorated the adverse biochemical alterations indicative of renal failure, abated the decrease in body weight and reduced the glomerular, tubular and interstitial lesions induced by adenine. Our study provides evidence that GA attenuated renal dysfunction in this model of CRF, suggesting a promising potential for it in protecting against renal failure progression. The mechanism(s) of this nephroprotection is uncertain but may involve anti-oxidant and/or anti-inflammatory actions.
Collapse
Affiliation(s)
- Badreldin H Ali
- Department of Pharmacology, Sultan Qaboos University, Al-Khod, Sultanate of Oman
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Evenepoel P, Meijers BKI, Bammens BRM, Verbeke K. Uremic toxins originating from colonic microbial metabolism. Kidney Int 2010:S12-9. [PMID: 19946322 DOI: 10.1038/ki.2009.402] [Citation(s) in RCA: 312] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Numerous molecules, which are either excreted or metabolized by the kidney, accumulate in patients with chronic kidney disease (CKD). These uremic retention molecules (URMs), contributing to the syndrome of uremia, may be classified according to their site of origin, that is, endogenous metabolism, microbial metabolism, or exogenous intake. It is increasingly recognized that bacterial metabolites, such as phenols, indoles, and amines, may contribute to uremic toxicity. In vitro studies have implicated bacterial URMs in CKD progression, cardiovascular disease, and bone and mineral disorders. Furthermore, several observational studies have demonstrated a link between serum levels of bacterial URMs and clinical outcomes. Bacterial metabolism may therefore be an important therapeutic target in CKD. There is evidence that besides reduced renal clearance, increased colonic generation and absorption explain the high levels of bacterial URMs in CKD. Factors promoting URM generation and absorption include an increased ratio of dietary protein to carbohydrate due to insufficient intake of fiber and/or reduced intestinal protein assimilation, as well as prolonged colonic transit time. Two main strategies exist to reduce bacterial URM levels: interventions that modulate intestinal bacterial growth (e.g., probiotics, prebiotics, dietary modification) and adsorbent therapies that bind bacterial URMs in the intestines to reduce their absorption (e.g., AST-120, sevelamer). The efficacy and clinical benefit of these strategies are currently an active area of interest.
Collapse
Affiliation(s)
- Pieter Evenepoel
- Department of Nephrology, University Hospitals Leuven, Leuven, Belgium.
| | | | | | | |
Collapse
|
41
|
Meijers BKI, De Preter V, Verbeke K, Vanrenterghem Y, Evenepoel P. p-Cresyl sulfate serum concentrations in haemodialysis patients are reduced by the prebiotic oligofructose-enriched inulin. Nephrol Dial Transplant 2009; 25:219-24. [PMID: 19692415 DOI: 10.1093/ndt/gfp414] [Citation(s) in RCA: 219] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
INTRODUCTION Protein-bound uraemic retention solutes, including p-cresyl sulfate and indoxyl sulfate, contribute substantially to the uraemic syndrome. These and several other uraemic retention solutes originate from intestinal bacterial protein fermentation. We investigated whether the prebiotic oligofructose-enriched inulin reduced serum concentration of p-cresyl sulfate and indoxyl sulfate, through interference with intestinal generation. METHODS We performed a single centre, non-randomized, open-label phase I/II study in maintenance HD patients with a 4-week, escalating dose regimen of oligofructose-enriched inulin (ORAFTI Synergy 1, Tienen, Belgium) (www.clinicaltrials.gov NCT00695513). Changes in p-cresyl sulfate and indoxyl sulfate serum concentrations as well as changes in p-cresyl sulfate and indoxyl sulfate generation rates were analysed. RESULTS Compliance with therapy was excellent. p-Cresyl sulfate serum concentrations at 4 weeks were significantly reduced by 20% (intention to treat, P = 0.01; per protocol, P = 0.03). Also p-cresyl sulfate generation rates were reduced (P = 0.007). In contrast, neither indoxyl sulfate generation rates (P = 0.9) nor serum concentrations (P = 0.4) were significantly changed. CONCLUSION The prebiotic oligofructose-inulin significantly reduced p-cresyl sulfate generation rates and serum concentrations in haemodialysis patients. Whether reduction of p-cresyl sulfate serum concentrations, an independent predictor of cardiovascular disease in HD patients, will result in improved cardiovascular outcomes remains to be proven.
Collapse
Affiliation(s)
- Björn K I Meijers
- Division of Nephrology, Department of Medicine, University Hospital Leuven, 3000 Leuven, Belgium
| | | | | | | | | |
Collapse
|
42
|
Abstract
BACKGROUND For more than fifty years, low protein diets have been proposed to patients with kidney failure. However, the effects of these diets in preventing severe kidney failure and the need for maintenance dialysis have not been resolved. OBJECTIVES To determine the efficacy of low protein diets in delaying the need to start maintenance dialysis. SEARCH STRATEGY Cochrane Renal Group studies register, the Cochrane Central Register of Controlled studies, MEDLINE, and EMBASE. Congress abstracts (American Society of Nephrology since 1990, European Dialysis Transplant Association since 1985, International Society of Nephrology since 1987). Direct contacts with investigators. SELECTION CRITERIA Randomised studies comparing two different levels of protein intake in adult patients suffering from moderate to severe kidney failure, followed for at least one year. DATA COLLECTION AND ANALYSIS Two authors independently selected studies and extracted data. Statistical analyses were performed using the random effects model and the results expressed as risk ratio (RR) for dichotomous outcomes with 95% confidence intervals (CI). Collection of the number of "renal deaths" defined as the need for starting dialysis, the death of a patient or a kidney transplant during the study. MAIN RESULTS Ten studies were identified from over 40 studies. A total of 2000 patients were analysed, 1002 had received reduced protein intake and 998 a higher protein intake. There were 281 renal deaths recorded, 113 in the low protein diet and 168 in the higher protein diet group (RR 0.68, 95% CI 0.55 to 0.84, P = 0.0002). To avoid one renal death, 2 to 56 patients need to be treated with a low protein diet during one year. AUTHORS' CONCLUSIONS Reducing protein intake in patients with chronic kidney disease reduces the occurrence of renal death by 32% as compared with higher or unrestricted protein intake. The optimal level of protein intake cannot be confirmed from these studies.
Collapse
Affiliation(s)
- Denis Fouque
- Département de Néphrologie, U870 INSERM-Université Claude Bernard Lyon 11, Hôpital Edouard Herriot, Lyon Cedex 03, France, 69437
| | | |
Collapse
|
43
|
Bajka BH, Clarke JM, Cobiac L, Topping DL. Butyrylated starch protects colonocyte DNA against dietary protein-induced damage in rats. Carcinogenesis 2008; 29:2169-74. [PMID: 18684730 DOI: 10.1093/carcin/bgn173] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dietary resistant starch (RS), as a high amylose maize starch (HAMS), prevents dietary protein-induced colonocyte genetic damage in rats, possibly through the short-chain fatty acid (SCFA) butyrate produced by large bowel bacterial RS fermentation. Increasing butyrate availability may improve colonic health and dietary high amylose maize butyrylated starch (HAMSB) is an effective method of achieving this goal. In this study, rats (n = 8 per group) were fed diets containing high levels (25%) of dietary protein as casein with 10 or 20% dietary HAMSB and HAMS. Colonocyte genetic damage was measured by the comet assay and was 2-fold higher in rats fed 25% protein than those fed 15% protein (P < 0.001). Concurrent feeding of 25% protein and either HAMS or HAMSB lowered genetic damage significantly relative to a low-RS high-protein control diet. The 20% HAMSB diet was twice as effective as 20% HAMS in opposing genetic damage. Large bowel digesta butyrate was significantly increased in rats fed 20% compared with 10% HAMS and in rats fed 20% compared with 10% HAMSB. The levels were significantly higher in the HAMSB groups relative to the HAMS groups. Hepatic portal venous SCFA were higher in rats fed HAMS and highest in those fed HAMSB. Caecal digesta ammonia was increased by HAMSB and correlated negatively with digesta pH. Ammonia is cytotoxic and lower digesta pH could lower its absorption, possibly contributing to lower genetic damage. Delivery of butyrate to the large bowel by HAMSB could reduce colorectal cancer risk by preventing diet-induced colonocyte genetic damage.
Collapse
Affiliation(s)
- Balazs H Bajka
- Preventative Health National Research Flagship, Adelaide, South Australia
| | | | | | | |
Collapse
|
44
|
Ali BH, Ziada A, Blunden G. Biological effects of gum arabic: a review of some recent research. Food Chem Toxicol 2008; 47:1-8. [PMID: 18672018 DOI: 10.1016/j.fct.2008.07.001] [Citation(s) in RCA: 208] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Revised: 06/10/2008] [Accepted: 07/03/2008] [Indexed: 02/06/2023]
Abstract
Gum arabic (GA) is a branched-chain, complex polysaccharide, either neutral or slightly acidic, found as a mixed calcium, magnesium and potassium salt of a polysaccharidic acid. The backbone is composed of 1,3-linked beta-D-galactopyranosyl units. The side chains are composed of two to five 1,3-linked beta-D-galactopyranosyl units, joined to the main chain by 1,6-linkages. Pharmacologically, GA has been claimed to act as an anti-oxidant, and to protect against experimental hepatic-, renal- and cardiac toxicities in rats. These reports could not be confirmed by others. GA has been claimed to alleviate the adverse effects of chronic renal failure in humans. This could not be corroborated experimentally in rats. Reports on the effects of GA on lipid metabolism in humans and rats are at variance, but mostly suggest that GA ingestion can reduce plasma cholesterol concentrations in rats. GA has proabsorptive properties and can be used in diarrhoea. It enhances dental remineralization, and has some antimicrobial activity, suggesting a possible use in dentistry. GA has been shown to have an adverse effect on electrolyte balance and vitamin D in mice, and to cause hypersensitivity in humans. More studies are needed before the pharmacological properties of GA can be utilized in therapy.
Collapse
Affiliation(s)
- Badreldin H Ali
- Department of Pharmacology, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Al Khod 123, Oman.
| | | | | |
Collapse
|