Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Nutrients ; 16(11)2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38892721

ABSTRACT

The gut microbiota performs several crucial roles in a holobiont with its host, including immune regulation, nutrient absorption, synthesis, and defense against external pathogens, significantly influencing host physiology. Disruption of the gut microbiota has been linked to various chronic conditions, including cardiovascular, kidney, liver, respiratory, and intestinal diseases. Studying how animals adapt their gut microbiota across their life course at different life stages and under the dynamics of extreme environmental conditions can provide valuable insights from the natural world into how the microbiota modulates host biology, with a view to translating these into treatments or preventative measures for human diseases. By modulating the gut microbiota, opportunities to address many complications associated with chronic diseases appear. Such a biomimetic approach holds promise for exploring new strategies in healthcare and disease management.


Subject(s)
Gastrointestinal Microbiome , Animals , Gastrointestinal Microbiome/physiology , Humans , Life Style , Nutritional Status , Chronic Disease
2.
J Ren Nutr ; 33(6S): S110-S117, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37676185

ABSTRACT

Chronic kidney disease is a significant risk factor for cardiovascular disease. In addition to traditional risk factors, such as hypertension, dyslipidemia, diabetes and smoking, patients with chronic kidney disease have a uremic phenotype marked by premature aging, mitochondrial dysfunction, persistent low-grade inflammation, gut dysbiosis and oxidative stress. These complications contribute to abnormal vascular and myocardial remodeling processes, resulting in accelerated vascular calcification, cellular and organ senescence and a high risk of cardiovascular disease. Nonpharmacological strategies, such as increasing physical activity and a healthy diet, may slow the progression of kidney disease and consequently protect the heart. Thus, a deep promotion and advocacy of nutritional guidance based on scientific data is needed. This narrative review discusses how nutritional interventions may delay progressive organ damage in the kidney-heart axis.


Subject(s)
Cardiovascular Diseases , Hypertension , Renal Insufficiency, Chronic , Humans , Cardiovascular Diseases/complications , Kidney , Renal Insufficiency, Chronic/complications , Risk Factors
3.
J Ren Nutr ; 33(6S): S118-S127, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37632513

ABSTRACT

Some chronic diseases, including chronic kidney disease (CKD), may be associated with poor outcomes, including a high rate of hospitalization and death after COVID-19 infection. In addition to the vaccination program, diet intervention is essential for boosting immunity and preventing complications. A healthy diet containing bioactive compounds may help mitigate inflammatory responses and oxidative stress caused by COVID-19. In this review, we discuss dietary interventions for mitigating COVID-19 complications, including in persons with CKD, which can worsen COVID-19 symptoms and its clinical outcomes, while diet may help patients with CKD to resist the ravages of COVID-19 by improving the immune system, modulating gut dysbiosis, mitigating COVID-19 complications, and reducing hospitalization and mortality. The concept of food as medicine, also known as culinary medicine, for patients with CKD can be extrapolated to COVID-19 infection because healthy foods and nutraceuticals have the potential to exert an important antiviral, anti-inflammatory, and antioxidant role.


Subject(s)
COVID-19 , Renal Insufficiency, Chronic , Humans , COVID-19/complications , Renal Insufficiency, Chronic/complications , Diet , Dietary Supplements , Antioxidants/therapeutic use
4.
Toxins (Basel) ; 15(8)2023 08 11.
Article in English | MEDLINE | ID: mdl-37624256

ABSTRACT

Residual kidney function for patients with chronic kidney disease (CKD) is associated with better quality of life and outcome; thus, strategies should be implemented to preserve kidney function. Among the multiple causes that promote kidney damage, gut dysbiosis due to increased uremic toxin production and endotoxemia need attention. Several strategies have been proposed to modulate the gut microbiota in these patients, and diet has gained increasing attention in recent years since it is the primary driver of gut dysbiosis. In addition, medications and faecal transplantation may be valid strategies. Modifying gut microbiota composition may mitigate chronic kidney damage and preserve residual kidney function. Although various studies have shown the influential role of diet in modulating gut microbiota composition, the effects of this modulation on residual kidney function remain limited. This review discusses the role of gut microbiota metabolism on residual kidney function and vice versa and how we could preserve the residual kidney function by modulating the gut microbiota balance.


Subject(s)
Gastrointestinal Microbiome , Humans , Dysbiosis , Quality of Life , Fecal Microbiota Transplantation , Kidney
5.
Mol Nutr Food Res ; 67(9): e2200859, 2023 05.
Article in English | MEDLINE | ID: mdl-36861422

ABSTRACT

Fish is an excellent source of ω-3 polyunsaturated fatty acids (PUFAs), amino acids, collagen, vitamins, and iodine and its intake is associated with health benefits, mainly reduces risk of cardiovascular mortality. However, recent studies have shown that fish is also an important source of trimethylamine N-oxide (TMAO), a uremic toxin produced by the gut microbiota that promotes an increased risk of cardiovascular diseases. In patients with chronic kidney disease (CKD), TMAO levels are markedly increased due to gut dysbiosis and reduced kidney function. No study has yet evaluated the effects of a fish-rich diet on TMAO plasma levels and cardiovascular outcomes. This review discusses the pros and cons of a fish-rich diet in patients with CKD - a matter of depth.


Subject(s)
Gastrointestinal Microbiome , Renal Insufficiency, Chronic , Renal Insufficiency , Animals , Methylamines
6.
Complement Ther Clin Pract ; 49: 101669, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36152526

ABSTRACT

BACKGROUND: Chronic kidney disease patients on hemodialysis commonly have a worse quality of life (QoL) due to complications of the disease and dialysis procedure. Physical exercise has emerged as a strategy to improve this scenario. The objective of this study was to evaluate the effect of an intradialytic aerobic exercise program on QoL and aerobic fitness in hemodialysis patients. MATERIAL AND METHODS: These are a secondary analysis of clinical trial data previously published in which hemodialysis patients were randomized into "bike group" (using an adapted exercise bicycle) or "control group" (usual care). The exercise sessions lasted 45 min (5 min of warm-up, 35 min of moderate-intensity and 5 min of cool-down) three times/week for three months. The QoL domains were assessed using the SF-36 QoL questionnaire. Aerobic fitness was evaluated using the 6-min walk test (6MWT). Circulating cytokines, biochemical parameters and Kt/V were also assessed. RESULTS: Nine patients completed three months of exercise (5 men, 44 ± 11 years), and nine were in the control group (6 men, 44 ± 14 years). In the bike group, there was a trend to improve the physical role domain (p = 0.06) regarding QoL, an improvement in the 6MWT (p = 0.02), and in the Kt/V (p = 0.03) after three months. There was a positive correlation between the general health domain and Kt/V (r = 0.691; p = 0.003) and an inverse correlation between the physical functioning domain and plasma TNF-α levels (r = -0.514; p = 0.04). CONCLUSIONS: 12 weeks of intradialytic aerobic exercise was enough to benefit hemodialysis patients' quality of life, aerobic fitness, and quality of dialysis. CLINICALTRIALS: gov id: NCT04375553.


Subject(s)
Kidney Failure, Chronic , Renal Dialysis , Male , Humans , Quality of Life , Pilot Projects , Kidney Failure, Chronic/therapy , Kidney Failure, Chronic/complications , Bicycling , Exercise , Exercise Therapy/methods
7.
Crit Rev Food Sci Nutr ; : 1-14, 2022 Aug 12.
Article in English | MEDLINE | ID: mdl-35959705

ABSTRACT

Inflammageing is a persistent low-level inflammatory burden that accompanies age-related dysregulation of the immune system during normative aging and within the diseasome of aging. A healthy diet containing a balanced amount of macronutrients, vitamins and minerals, adequate in calories and rich in poly(phenols), has an essential role in mitigating the effects of inflammageing and extending healthspan through modulation of the activity of a range of factors. These include transcription factors, such as nuclear factor erythroid-derived 2 related factor 2 (Nrf2) and nuclear factor-κB (NF-kB), the inflammasome and the activities of the gut microbiota. The aim of this narrative review is to discuss the potential of food to ameliorate the effects of the diseasome of aging.

8.
Nutr Clin Pract ; 37(1): 81-93, 2022 Feb.
Article in English | MEDLINE | ID: mdl-33979013

ABSTRACT

Patients with chronic kidney disease (CKD) have several pathophysiological alterations, including anemia, one of the first changes in CKD patients. More recently, researchers have observed that the intestinal microbiota alterations are also another complication in these patients. The most common treatment for anemia is oral (mainly ferrous sulfate) or intravenous iron supplementation. Despite being a necessary treatment, recent studies have reported that supplementation with oral iron may increase its availability in the intestine, leading to disturbance in the gut microbiota and also to oxidative stress in the enterocytes, which may change the permeability and the microbiota profile. Although it is a therapy routinely used in patients with CKD, supplementation with oral iron on the gut microbiota has been rarely studied in these patients. Thus, this review will discuss the relationship between iron and the gut microbiota and the possible effects of oral iron supplementation on gut microbiota in patients with CKD.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Renal Insufficiency, Chronic , Dietary Supplements , Humans , Iron , Renal Insufficiency, Chronic/complications
9.
Nutr Rev ; 80(5): 1041-1061, 2022 04 08.
Article in English | MEDLINE | ID: mdl-34613396

ABSTRACT

Chronic kidney disease (CKD) manifests with systemic inflammation, oxidative stress, and gut dysbiosis, resulting in metabolic disorders and elevated rates of cardiovascular disease-associated death. These all correlate with a high economic cost to healthcare systems. Growing evidence indicates that diet is an indispensable ally in the prevention and management of CKD and its complications. In this context, the root vegetable beetroot (Beta vulgaris rubra) deserves special attention because it is a source of several bioactive compounds, such as nitrate, betaine, and betalain, and has shown beneficial effects in CKD, including reduction of blood pressure, anti-inflammatory effects, and antioxidant actions by scavenging radical oxidative species, as observed in preclinical studies. Beetroot consumption as a possible therapeutic strategy to improve the clinical treatment of patients with CKD and future directions for clinical studies are addressed in this narrative review.


Subject(s)
Beta vulgaris , Renal Insufficiency, Chronic , Antioxidants/therapeutic use , Dysbiosis , Humans , Renal Insufficiency, Chronic/drug therapy , Vegetables
10.
Nutrition ; 83: 111094, 2021 03.
Article in English | MEDLINE | ID: mdl-33418489

ABSTRACT

Propolis is a polyphenolic plant resin collected by bees to protect hives against pathogens and temperature drop. It exhibits antibacterial, antioxidant, and antiinflammatory properties. Propolis has been reported to possess antidiabetic properties and display beneficial effects against cardiovascular disease, gut dysbiosis, and chronic kidney disease. It has an excellent clinical safety profile, with no known toxic effects described so far. In this review, we discuss the salutogenic effects of propolis, with particular reference to modulating notable features of chronic kidney disease, notably those involving cardiovascular risks.


Subject(s)
Anti-Infective Agents , Propolis , Animals , Bees , Dysbiosis , Life Style , Plant Extracts
11.
Nat Rev Nephrol ; 17(3): 153-171, 2021 03.
Article in English | MEDLINE | ID: mdl-32963366

ABSTRACT

The observation that unhealthy diets (those that are low in whole grains, fruits and vegetables, and high in sugar, salt, saturated fat and ultra-processed foods) are a major risk factor for poor health outcomes has boosted interest in the concept of 'food as medicine'. This concept is especially relevant to metabolic diseases, such as chronic kidney disease (CKD), in which dietary approaches are already used to ameliorate metabolic and nutritional complications. Increased awareness that toxic uraemic metabolites originate not only from intermediary metabolism but also from gut microbial metabolism, which is directly influenced by diet, has fuelled interest in the potential of 'food as medicine' approaches in CKD beyond the current strategies of protein, sodium and phosphate restriction. Bioactive nutrients can alter the composition and metabolism of the microbiota, act as modulators of transcription factors involved in inflammation and oxidative stress, mitigate mitochondrial dysfunction, act as senolytics and impact the epigenome by altering one-carbon metabolism. As gut dysbiosis, inflammation, oxidative stress, mitochondrial dysfunction, premature ageing and epigenetic changes are common features of CKD, these findings suggest that tailored, healthy diets that include bioactive nutrients as part of the foodome could potentially be used to prevent and treat CKD and its complications.


Subject(s)
Diet, Healthy/methods , Renal Insufficiency, Chronic/therapy , Uremia/prevention & control , Disease Progression , Humans , Phenotype , Renal Insufficiency, Chronic/complications , Uremia/etiology
12.
J. bras. nefrol ; 42(3): 307-314, July-Sept. 2020. tab, graf
Article in English, Portuguese | LILACS | ID: biblio-1134843

ABSTRACT

ABSTRACT Background: Patients with chronic kidney disease (CKD) present an imbalance of the gut microbiota composition, leading to increased production of uremic toxins like p-cresyl sulfate (PCS), product from bacterial fermentation of the amino acids tyrosine (Tyr) and phenylalanine (Phe) from the diet. Thus, diet may be a determinant in the uremic toxins levels produced by the gut microbiota. The aim of this study was to evaluate the possible relationship between Tyr and Phe intake and PCS plasma levels in non-dialysis CKD patients. Methods: Twenty-seven non-dialysis CKD patients (stages 3 and 4) without previous nutritional intervention were evaluated. The dietary intake was evaluated using a 24-hour recall, 3-day food record and protein intake was also estimated by Protein Nitrogen Appearance (PNA). The plasma levels of PCS were measured using reverse phase high performance liquid chromatography. Results: The evaluated patients (GRF, 34.8 ± 12.4 mL/min, 54.2 ± 14.3 years, BMI, 29.3 ± 6.1 kg/m2) presented mean protein intake of 1.1 ± 0.5 g/kg/day), Tyr of 4.5 ± 2.4 g/day and Phe of 4.6 ± 2.5 g/day. PCS plasma levels (20.4 ± 15.5 mg/L) were elevated and positively associated with both, Tyr (r = 0.58, p = 0.002) and Phe intake (r = 0.53, p = 0.005), even after adjustments for eGFR and age. Conclusion: This study suggests that the diet is an important modulator of the uremic toxins plasma levels produced by the gut microbiota, in non-dialysis CKD patients.


RESUMO Introdução: Pacientes com doença renal crônica (DRC) apresentam desequilíbrio na composição da microbiota intestinal, gerando toxinas urêmicas, como o p-cresil sulfato (PCS), pela fermentação bacteriana dos aminoácidos tirosina (Tyr) e fenilalanina (Phe) da dieta. Assim, a dieta pode ser determinante nos níveis de toxinas urêmicas produzidos pela microbiota intestinal. O objetivo deste estudo foi avaliar a possível relação entre a ingestão de Tyr e Phe e os níveis plasmáticos de PCS em pacientes com DRC não dialisados. Métodos: Foram avaliados 27 pacientes com DRC em tratamento conservador (estágios 3 e 4), sem intervenção nutricional prévia. A ingestão alimentar foi avaliada pelo recordatório alimentar de 24h (R-24h) de 3 dias, e a ingestão proteica também foi verificada através do Protein Nitrogen Appearance (PNA). Os níveis plasmáticos de PCS foram determinados por cromatografia líquida de fase reversa. Resultados: Os pacientes avaliados (TFG, 34,8 ± 12,4 mL/min, 54,2 ± 14,3 anos, IMC 29,3 ± 6,1 kg/m2) apresentaram ingestão média de proteína de 1,1 ± 0,5 g/kg/dia, Tyr de 4,5 ± 2,4 g/dia e Phe de 4,6 ± 2,5 g/dia. Os níveis plasmáticos de PCS (20,4 ± 15,5 mg/L) foram elevados e positivamente associados à ingestão de Tyr (r = 0,58, p = 0,002) e Phe (r = 0,53, p = 0,005), mesmo após ajustes pela TFG e idade. Conclusão: Este estudo sugere que a dieta é um importante modulador dos níveis plasmáticos de toxinas urêmicas produzidas pela microbiota intestinal em pacientes com DRC não dialisados.


Subject(s)
Humans , Phenylalanine , Tyrosine , Diet , Renal Insufficiency, Chronic , Indican , Sulfates , Sulfuric Acid Esters , Cresols , Eating
13.
Food Res Int ; 136: 109306, 2020 10.
Article in English | MEDLINE | ID: mdl-32846516

ABSTRACT

Inflammatory and innate immune responses triggered by pathogen-associated and other danger-associated signals emerging during infections, results in the activation of cytosolic inflammasomes. The nod-like receptor pyrin domain containing 3 (NLRP3) is one of the inflammasomes mediating such responses through the activation of caspase-1, which increases the production and release of pro-inflammatory cytokines, such as IL-1ß and IL-18 and induces programmed cell death through pyroptosis. NLRP3 is thought to play a crucial role in the underlying inflammatory responses in many lifestyles related chronic diseases. Consequently, research on the NLRP3 inflammasome has expanded dramatically in recent years. Although several studies have investigated the role of NLRP3 activation in chronic kidney disease (CKD), few studies have evaluated strategies to modulate its activation by means of interventions using non-pharmacological strategies. This review discusses some nutritional strategies (bioactive compounds, probiotics and caloric restriction) that have been shown to influence NLRP3 in experimental models of renal disease, and in CKD. It discusses how nutritional interventions could potentially dampen NLRP3 associated inflammatory burden, as part of nutritional strategies to prevent and treat CKD and its complications.


Subject(s)
Inflammasomes/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Nutrition Therapy/methods , Renal Insufficiency, Chronic/diet therapy , Renal Insufficiency, Chronic/physiopathology , Animals , Caloric Restriction , Fatty Acids, Unsaturated/administration & dosage , Humans , Immunity, Innate , Inflammation/physiopathology , Phytochemicals/administration & dosage , Probiotics , Renal Insufficiency, Chronic/prevention & control
14.
J Bras Nefrol ; 42(3): 307-314, 2020.
Article in English, Portuguese | MEDLINE | ID: mdl-32459280

ABSTRACT

BACKGROUND: Patients with chronic kidney disease (CKD) present an imbalance of the gut microbiota composition, leading to increased production of uremic toxins like p-cresyl sulfate (PCS), product from bacterial fermentation of the amino acids tyrosine (Tyr) and phenylalanine (Phe) from the diet. Thus, diet may be a determinant in the uremic toxins levels produced by the gut microbiota. The aim of this study was to evaluate the possible relationship between Tyr and Phe intake and PCS plasma levels in non-dialysis CKD patients. METHODS: Twenty-seven non-dialysis CKD patients (stages 3 and 4) without previous nutritional intervention were evaluated. The dietary intake was evaluated using a 24-hour recall, 3-day food record and protein intake was also estimated by Protein Nitrogen Appearance (PNA). The plasma levels of PCS were measured using reverse phase high performance liquid chromatography. RESULTS: The evaluated patients (GRF, 34.8 ± 12.4 mL/min, 54.2 ± 14.3 years, BMI, 29.3 ± 6.1 kg/m2) presented mean protein intake of 1.1 ± 0.5 g/kg/day), Tyr of 4.5 ± 2.4 g/day and Phe of 4.6 ± 2.5 g/day. PCS plasma levels (20.4 ± 15.5 mg/L) were elevated and positively associated with both, Tyr (r = 0.58, p = 0.002) and Phe intake (r = 0.53, p = 0.005), even after adjustments for eGFR and age. CONCLUSION: This study suggests that the diet is an important modulator of the uremic toxins plasma levels produced by the gut microbiota, in non-dialysis CKD patients.


Subject(s)
Diet , Indican , Phenylalanine , Renal Insufficiency, Chronic , Tyrosine , Cresols , Eating , Humans , Sulfates , Sulfuric Acid Esters
15.
J Nutr ; 149(3): 372-380, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30796783

ABSTRACT

Epigenetic alterations, such as those linked to DNA methylation, may potentially provide molecular explanations for complications associated with altered gene expression in illnesses, such as chronic kidney disease (CKD). Although both DNA hypo- and hypermethylation have been observed in the uremic milieu, this remains only a single aspect of the epigenetic landscape and, thus, of any biochemical dysregulation associated with CKD. Nevertheless, the role of uremia-promoting alterations on the epigenetic landscape regulating gene expression is still a novel and scarcely studied field. Although few studies have actually reported alterations of DNA methylation via methyl donor nutrient intake, emerging evidence indicates that nutritional modification of the microbiome can affect one-carbon metabolism and the capacity to methylate the genome in CKD. In this review, we discuss the nutritional modifications that may affect one-carbon metabolism and the possible impact of methyl donor nutrients on the microbiome, CKD, and its phenotype.


Subject(s)
DNA Methylation , Epigenesis, Genetic/physiology , Gene Expression Regulation/physiology , Renal Insufficiency, Chronic/metabolism , Aging , Humans , Nutritional Status
16.
Probiotics Antimicrob Proteins ; 11(2): 648-654, 2019 06.
Article in English | MEDLINE | ID: mdl-29651635

ABSTRACT

Components present in the diet, L-carnitine, choline, and betaine are metabolized by gut microbiota to produce metabolites such as trimethylamine-N-oxide (TMAO) that appear to promote cardiovascular disease in chronic kidney disease (CKD) patients. The objective of this pilot study was to evaluate the effects of probiotic supplementation for 3 months on plasma TMAO levels in CKD patients on hemodialysis (HD). A randomized, double-blind trial was performed in 21 patients [54.8 ± 10.4 years, nine men, BMI 26.1 ± 4.8 kg/m2, dialysis vintage 68.5 (34.2-120.7) months]. Ten patients were randomly allocated to the placebo group and 11 to the probiotic group [three capsules, totaling 9 × 1013 colony-forming units per day of Streptococcus thermophilus (KB19), Lactobacillus acidophilus (KB27), and Bifidobacteria longum (KB31). Plasma TMAO, choline, and betaine levels were measured by LC-MS/MS at baseline and after 3 months. While TMAO did not change after probiotic supplementation, there was a significant increase in betaine plasma levels. In contrast, the placebo group showed a significant decrease in plasma choline levels. Short-term probiotic supplementation does not appear to influence plasma TMAO levels in HD patients. Long-term studies are needed to determine whether probiotics may affect TMAO production in CKD patients.


Subject(s)
Methylamines/blood , Probiotics/administration & dosage , Renal Dialysis , Adult , Aged , Bifidobacterium longum , Dietary Supplements , Double-Blind Method , Female , Humans , Lactobacillus acidophilus , Male , Methylamines/metabolism , Middle Aged , Pilot Projects , Renal Insufficiency, Chronic/metabolism , Streptococcus thermophilus
17.
Food Funct ; 9(12): 6508-6516, 2018 Dec 13.
Article in English | MEDLINE | ID: mdl-30468238

ABSTRACT

An imbalance of gut microbiota is considered a new cardiovascular risk factor for chronic kidney disease (CKD) patients, since it is directly associated with increased uremic toxin production, inflammation and oxidative stress. Strategies such as prebiotic supplementation have been suggested to mitigate these complications. We hypothesized that prebiotic-resistant starch could ameliorate uremic toxins levels, oxidative stress, and inflammatory states in hemodialysis (HD) patients. This pilot study evaluated 31 HD patients assigned to either resistant starch (16 g of resistant starch Hi-Maize® 260) or placebo (manioc flour) supplementation, which they received for 4 weeks on alternate days through cookies on dialysis days and powder in a sachet on non-dialysis days. Levels of interleukin (IL)-6, high-sensitive C-reactive protein, thiobarbituric acid reactive substances plasma (TBARS), protein carbonylation, indoxyl sulfate (IS) and p-cresyl sulfate were measured. Anthropometric and biochemical parameters, as well as, food intake were also evaluated. As expected, resistant starch group increased fiber intake (p > 0.01), in addition the prebiotic supplementation reduced IL-6 (p = 0.01), TBARS (p > 0.01), and IS (p > 0.01) plasma levels. No significant differences were evident in the placebo group. Prebiotic-resistant starch supplementation seems to be a promising nutritional strategy to improve inflammation, oxidative stress and to reduce IS plasma levels in CKD patients on HD.


Subject(s)
Cresols/urine , Indican/urine , Oxidative Stress/drug effects , Prebiotics/administration & dosage , Renal Insufficiency, Chronic/diet therapy , Starch/metabolism , Sulfuric Acid Esters/urine , Adult , Anthropometry , Biomarkers/blood , Biomarkers/urine , C-Reactive Protein/metabolism , Female , Humans , Interleukin-6/blood , Male , Middle Aged , Pilot Projects , Renal Dialysis , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/therapy , Renal Insufficiency, Chronic/urine , Thiobarbituric Acid Reactive Substances/metabolism , Urine/chemistry , Zea mays/chemistry , Zea mays/metabolism
18.
Int Urol Nephrol ; 50(12): 2207-2220, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29915880

ABSTRACT

Açai (Euterpe oleracea Mart.) fruit from the Amazon region in Brazil contains bioactive compounds such as α-tocopherol, anthocyanins (cyanidin 3-glycoside and cyanidin 3-rutinoside), and other flavonoids with antioxidant and anti-inflammatory properties. Moreover, the prebiotic activity of anthocyanins in modulating the composition of gut microbiota has emerged as an additional mechanism by which anthocyanins exert health-promoting effects. Açai consumption may be a nutritional therapeutic strategy for chronic kidney disease (CKD) patients since these patients present with oxidative stress, inflammation, and dysbiosis. However, the ability of açai to modulate these conditions has not been studied in CKD, and this review presents recent information about açai and its possible therapeutic effects in CKD.


Subject(s)
Anthocyanins , Euterpe , Inflammation/drug therapy , Oxidative Stress/drug effects , Renal Insufficiency, Chronic , Anthocyanins/chemistry , Anthocyanins/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , Humans , Phytotherapy/methods , Plants, Medicinal , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/therapy , Treatment Outcome
19.
Nutrition ; 46: 26-32, 2018 02.
Article in English | MEDLINE | ID: mdl-29290351

ABSTRACT

Red meat is an important dietary source of high biological value protein and micronutrients such as vitamins, iron, and zinc that exert many beneficial functions. However, high consumption of animal protein sources, especially red meat, results in an increased intake of saturated fat, cholesterol, iron, and salt, as well as an excessive acid load. Red meat intake may lead to an elevated production of uremic toxins by the gut microbiota, such as trimethylamine n-oxide (TMAO), indoxyl sulfate, and p-cresyl sulfate. These uremic toxins are associated with increased risk for cardiovascular (CV) mortality. Limiting the intake of red meat in patients with chronic kidney disease (CKD) thus may be a good strategy to reduce CV risk, and may slow the progression of kidney disease. In the present review, we discuss the role of red meat in the diet of patients with CKD. Additionally, we report on a pilot study that focused on the effect of a low-protein diet on TMAO plasma levels in nondialysis CKD patients.


Subject(s)
Red Meat/adverse effects , Red Meat/analysis , Renal Insufficiency, Chronic/diet therapy , Cardiovascular Diseases/etiology , Diet, Protein-Restricted , Dietary Fats/administration & dosage , Dietary Proteins/administration & dosage , Gastrointestinal Microbiome/physiology , Humans , Iron, Dietary/administration & dosage , Methylamines/blood , Methylamines/metabolism , Micronutrients/administration & dosage , Risk Factors , Toxins, Biological/metabolism , Vitamins/administration & dosage , Zinc/administration & dosage
20.
J Ren Nutr ; 28(1): 28-36, 2018 01.
Article in English | MEDLINE | ID: mdl-28888762

ABSTRACT

OBJECTIVE: The objective of the study was to evaluate the effects of probiotic supplementation on the gut microbiota profile and inflammatory markers in chronic kidney disease patients undergoing maintenance hemodialysis (HD). DESIGN AND METHODS: This was a randomized, double-blind, placebo-controlled study. Forty-six HD patients were assigned to receive 1 of 2 treatments: probiotic (n = 23; Streptococcus thermophilus, Lactobacillus acidophilus e Bifidobacterialongum, 90 billion colony-forming units per day) or placebo (n = 23) daily for 3 months. Blood and feces were collected at baseline and after intervention. The inflammatory markers (C-reactive protein and interleukin-6) were analyzed by immunoenzymatic assay (enzyme-linked immunosorbent assay). Uremic toxins plasma levels (indoxyl sulfate, p-cresyl sulfate, and indole-3-acetic acid) were obtained by Reversed-Phase High-Performance Liquid Chromatography. Routine laboratory parameters were measured by standard techniques. Fecal pH was measured by the colorimetric method, and the gut microbiota profile was assessed by Denaturing Gradient Gel Electrophoresis analysis. RESULTS: Sixteen patients remained in the probiotic group (11 men, 53.6 ± 11.0 year old, 25.3 ± 4.6 kg/m2) and 17 in the placebo group (10 men, 50.3 ± 8.5 year old, 25.2 ± 5.7 kg/m2). After probiotic supplementation there was a significant increase in serum urea (from 149.6 ± 34.2 mg/dL to 172.6 ± 45.0 mg/dL, P = .02), potassium (from 4.4 ± 0.4 mmol/L to 4.8 ± 0.4 mmol/L, P = .02), and indoxyl sulfate (from 31.2 ± 15.9 to 36.5 ± 15.0 mg/dL, P = .02). The fecal pH was reduced from 7.2 ± 0.8 to 6.5 ± 0.5 (P = .01). These parameters did not change significantly in placebo group. Changes in the percentage delta (Δ) between groups were exhibited with no statistical differences observed. The inflammatory markers and gut profile were not altered by supplementation. CONCLUSIONS: Aprobiotic supplementation failed to reduce uremic toxins and inflammatory markers. Therefore, probiotic therapy should be chosen with caution in HD patients. Further studies addressing probiotic therapy in chronic kidney disease patients are needed.


Subject(s)
Probiotics/administration & dosage , Renal Insufficiency, Chronic/therapy , Adult , Bifidobacterium , Biomarkers/blood , Blood Glucose/metabolism , Body Mass Index , C-Reactive Protein/metabolism , Cresols/blood , Double-Blind Method , Feces/chemistry , Feces/microbiology , Female , Gastrointestinal Microbiome , Humans , Hydrogen-Ion Concentration , Indican/blood , Indoleacetic Acids/blood , Interleukin-6/blood , Lactobacillus acidophilus , Male , Middle Aged , Nutrition Assessment , Renal Insufficiency, Chronic/microbiology , Streptococcus thermophilus , Sulfuric Acid Esters/blood , Waist Circumference
SELECTION OF CITATIONS
SEARCH DETAIL
...