Skip to main content
Erschienen in: Journal of Nephrology 3/2023

Open Access 23.01.2023 | Review

Gut microbiota disturbances and protein-energy wasting in chronic kidney disease: a narrative review

verfasst von: Fabiola Martín-del-Campo, Carla Maria Avesani, Peter Stenvinkel, Bengt Lindholm, Alfonso M. Cueto-Manzano, Laura Cortés-Sanabria

Erschienen in: Journal of Nephrology | Ausgabe 3/2023

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.

Graphical abstract

Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

The gut has recently received increased attention as a potential target for preventive and therapeutic interventions aimed at decreasing the production, release and systemic absorption of gut-microbiota-derived uremic toxins in patients with advanced chronic kidney disease (CKD). The hypothesis is that these therapeutic interventions could potentially contribute to the prevention of metabolic and cardiovascular complications in this vulnerable population [1].
Microbiota disturbances have been linked to occurrence, progression and development of a range of complications in many chronic diseases, such as inflammatory bowel diseases [2], diabetes [3], neurological disorders [4] and cancer [5], and to acute infections not only in the gut but also in remote organs like the lungs during COVID-19 infection [6]. Trillions of commensal bacteria dynamically interact with the host through the intestinal epithelial cells, modulating local and systemic immunological functions; these interactions are related to nutrient digestion, absorption, metabolism and clearance of waste products, consequently affecting essentially all nutrition processes in the body [7]. Understanding the pathways and mechanisms by which nutritional balance could be modified and likely regulated by the gut microbiota is therefore important in identifying new potential therapeutic targets to prevent and treat complex nutritional disturbances.
It is now well recognized that protein-energy wasting (PEW) is a main complication in patients with advanced CKD that is associated with higher morbidity and mortality and lower quality of life [8, 8]. PEW is a complex syndrome characterized by loss of body muscle and fat stores, associated with inflammation and with the patient’s uremic conditions [10]. 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 closely related to PEW in CKD patients.

The uremic gut

The gut is an organ with multiple functions including absorption of nutrients, while at the same time preventing access of inflammatory and antigenic compounds into the body through mechanical, immunological and ecological external defense barriers. In addition, the gut is the host of a complex microbiota that is a source of numerous essential as well as potentially harmful metabolites. The human gut contains most of the microbial biomass in the body, with trillions of commensal bacteria, from more than 1000 different operational taxonomic units, accounting for 10 times more cells and a 150 times larger gene pool than the host [11]. The composition of gut microbiota varies according to age, geography, health conditions, lifestyle, and genetics, among many other factors; however, it is relatively stable in healthy individuals, and exhibits certain characteristics associated with health especially when it is mainly dominated by Firmicutes, Bacteroidetes, Actinobacteria and Proteobacteria bacteria phyla [12]. On the other hand, when the fine tuned balance between these microbes is altered, leading to gut dysbiosis, it may contribute to the development and progression of several diseases affecting not only the gut but also distant organs and systems.
It is now well established that CKD patients exhibit alterations in gut microbiota, with an increase in bacterial total counts of predominantly proteolytic bacteria. Compared to healthy individuals, patients have a lower α-diversity (species richness), as well as a decreased relative abundance of bacteria of the phyla Firmicutes and Actinobacteria, while Proteobacteria is increased. Enterococcus and Clostridium families are enriched in CKD, whereas the Prevotella, Coprococcus, Megamonas, Sutterella, Enterobacter, Acidaminococcus, Dorea, and Roseburia families are more abundant in healthy individuals [13]. When bacterial families are classified according to their enzyme characteristics, CKD patients have greater abundance in urease, uricase, tryptophanase (indole-forming enzyme) and hydroxyphenylacetate decarboxylase (p-cresol-forming enzyme) families, as well as a decrease in those with phosphotransbutyrylase and butyrate kinase (butyrate-forming enzymes) compared to healthy controls [14]. However, the latter studies did not always consider factors such as sex, age, comorbidities, dietary intake, environmental conditions and genetic aspects that may also influence gut microbiota in the CKD patient; this is supported by studies showing that gut microbiota in dialysis patients did not differ from that of healthy household contacts with similar dietary characteristics [15, 15].
Gut microbiota of CKD patients may not only differ from healthy people, but also according to the type of renal replacement therapy. A metagenomic analysis of microbiota in pediatric peritoneal dialysis (PD) patients showed lower relative abundance of Firmicutes and Actinobacteria phyla, and greater abundance of Bacteroidetes phyla, Proteobacteria phyla, and Enterobacteriaceae family than in hemodialysis (HD) patients. Children receiving PD and renal transplantation were reported to have a lower relative abundance of Bifidobacteriacea as well as lower α-diversity compared to healthy controls, however, the mean concentrations of uremic toxins did not differ among modalities [17]. Differences in gut microbiota have also been observed between adult non-dialysis, PD and HD patients, although whether PD or HD patients have better or worse microbiota is controversial, and may be associated with differences in metabolic and clinical variables [18, 18]. The microbiota in pediatric patients seems to show fewer differences in metabolic and clinical variables compared to adult populations, perhaps due to the different etiology of CKD in children [17].
Possible factors associated with these differences include characteristics of the renal replacement therapy itself, such as the hemodynamic changes in HD, the intestinal absorption of glucose from the dialysate during the PD procedure, or the use of immunosuppressive drugs in renal transplantation. Further research is necessary to confirm differences between these treatment modalities, to identify mechanisms by which a specific renal replacement therapy may affect the gut microbiota, and to analyze the potential clinical consequences of such changes.
In uremia, increased concentrations of nitrogen waste products, decreased intake of fiber, presence of malnutrition/malabsorption and constipation, and medication use, are some of the most common factors that were reported to be associated with bacterial overgrowth and dysbiosis in the gut [20]. Of importance, CKD patients have a high pill burden due to multiple comorbidities [21], and chronic use of some of the commonly used medications in CKD may alter the gut microbiota: (a) phosphate binders affect the intestinal environment, increasing gastrointestinal symptoms, and may bind to harmful molecules—other than phosphate—such as p-cresol, endotoxin, advanced glycation end products, bile acids, and oxalates, but also to beneficial molecules as vitamin K, folic acid, and short chain fatty acids (SCFAs), hence modifying nutrient absorption and gut microbiota [22]; (b) antibiotics lead to a dramatic impairment of the gut microbiota with a decrease in bacterial diversity and loss of different taxa, and the inappropriate use or overuse of antibiotics increases the risk of antibiotic-resistant infections, epithelial barrier disruption, and development of metabolic and immune diseases [23]; (c) oral iron medication promotes dysbiosis, thereby decreasing SCFA-forming bacteria and increasing proteolytic bacteria, and moreover, a high iron load promotes replication and virulence of pathobionts such as Salmonella, Shigella, Campylobacter or Citrobacter. In addition, luminal iron load induces the generation of reactive oxygen species, causing oxidative stress and intestinal epithelial damage [24]. All the above-mentioned mechanisms favor increased uremic toxin production, translocation of bacterial compounds including lipopolysaccharide, (LPS), and decreased SCFA synthesis [20].
Bacterial overgrowth, along with an increase in sympathetic activity, accumulation of nitrogen waste compounds, and the presence of stress and intestinal inflammatory diseases, all favor the loss of intestinal wall integrity by damaging the apical binding complex and decreasing survival of epithelial cells [25]. Apical binding disruptions occurring by several mechanisms may increase intestinal permeability, leading to a “leaky gut” and thereby enabling bacterial translocation. When urea reaches the intestinal lumen, it is metabolized by urease-containing bacteria into ammonium that is subsequently hydrolyzed into caustic ammonium hydroxide, which erodes the epithelial wall. The latter stimulates influx of inflammatory leukocytes, triggering retraction and endocytosis of apical binding transcellular proteins (occludin and claudin) as a consequence of increased local production of cytokines [26]. On the other hand, bacterial tryptophanase decreases host tryptophan bioavailability, and promotes an enteric rise of the levels of serotonin, a tryptophan metabolite, that may further worsen gut barrier dysfunction [27].
Bacterial translocation is recognized as a cause of local and systemic inflammation [28]. Once in the circulation, bacterial endotoxins join the LPS protein binding sites, forming a complex protein, which interacts with myeloid differentiation factor-2in the toll-like receptor 4 anchored to the CD14, thereby stimulating the transcription of nuclear factor kappa B (NF-κB), which in turn leads to increasing production and release of inflammatory cytokines as tumor necrosis factor-α (TNFα) and IL-1β [29] [25]. Endotoxin also mediates endothelial damage by boosting monocyte recruitment, transforming macrophages into foam cells and activating coagulant activity [1]. Additionally, pathogenic bacteria stimulate dendritic cells, activating T-cell response and increasing the production of TNFα and interferon-γ (INF-γ) [29]even more. A summary of findings related to the uremic gut is presented in Fig. 1.

Gut microbiota in food deprivation diseases

As far as we know, there are few studies focusing on links between PEW in CKD and microbiota characteristics. However, recent studies in other malnutrition syndromes suggest that microbiota could play a role in its development, and modification of the microbiota could be an opportunity to positively change the course of processes leading to PEW and thereby improve clinical outcomes.
Children with Kwashiorkor present low bacterial diversity in the gut, as well as an increase in Proteobacteria phyla and Streptococcus, and a decrease in Prevotella and Bacillus genus [30]. Malnourished Iranian children showed similar changes, with an increase in Proteobacteria phyla (Escherichia, Streptococcus, Veillonella, Shigella), and a decrease in Roseburia, Faecalibacterium and Butyrivibrio genus, along with increased severity of malnutrition [31]. Moreover, analysis of genome-scale metabolic models of gut microbiota shows that malnourished children have reduced levels of essential amino acids and lower metabolic capability to produce several non-essential amino acids, compared to healthy children [32].
Patients with anorexia nervosa present lower microbiota diversity, lower counts of anaerobic butyrate-forming bacteria, and lower SCFA concentrations, as well as increased counts of proteolytic and mucin-degrading bacteria, compared to healthy subjects. Weight gain in anorectic patients is associated with favorable changes in gut microbiota, with an increase in α-diversity, Firmicutes and Actinobacteria phyla, and Bifidobacteria and Ruminococcus spp [33]. It is possible that modifications in gut microbiota in food deprivation states could be the result of an adaptive response to optimize food transformation in low calorie diets. However, it should be noted that the etiology of PEW in CKD has significant differences compared to the aforementioned conditions, which are affected by factors such as extremely low dietary intake, poor quality of diet and poor hygiene.
Although CKD patients are exposed to many conditions other than low food intake that could lead to malnutrition (endocrine disorders, comorbidities, dialysis-related factors, metabolic acidosis, inflammation, and oxidative stress), it is possible that dysbiosis caused by inadequate dietary intake with lack of fibers may play a major role for the development of PEW. However, studies in this regard are scarce and implications of gut microbiota alterations on PEW development should be interpreted cautiously, particularly given the lack of longitudinal analyses. In a case–control study in HD patients, similar findings were seen, as patients with PEW presented lower α-diversity, Prevotella and Faecalibacterium genus abundance, as well as an increase in Enterococcus, Scardovia, Bifidobacterium, Anaerovorax, Collimonas y Akkermansia genus abundance compared to patients with normal nutrition by subjective global assessment [34]. In dialysis patients, muscle indicators such as mid-arm muscle area, and handgrip strength were positively correlated with Roseburia, Phascolarctobacterium and Coprococcus genus and negatively with Escherichia genus [35].

Nutrient metabolism in the gut: changes induced by uremia

The main determinant of bacterial metabolism is nutrient availability, particularly the disposable carbohydrate/protein ratio in the colon, which regulates the proteolytic and saccharolytic fermentation in the gut. Fermentable carbohydrates are the leading energy source, whereas proteins and amino acids are used essentially to increase biomass, however, protein fermentation (putrefaction) increases when carbohydrate availability is low [7, 36]. In CKD patients, the high concentrations of urea and nitrogen waste products reaching the gut through the intestinal fluids, together with greater bioavailability of amino acids and peptides related to malabsorption and/or intestinal edema, stimulate overgrowth of proteolytic bacteria, which in turn, increases production of uremic toxins. Additionally, low intake of fermentable carbohydrates is common in CKD patients, as foods rich in potassium and phosphorus such as fruits, vegetables and legumes that have a high fiber content are frequently restricted, thus decreasing the fermentation by saccharolytic bacteria and consequently the production of SCFAs [20, 36].

Bacterial uremic toxins and their role in PEW: impact of the gut-muscle axis in CKD

There is growing evidence of a special communication network between gut microbiota and skeletal muscle (size, composition and functionality), particularly in aging and cancer cachexia settings, the so-called gut-muscle axis [37]. In CKD patients, intestinal bacterial metabolism generates uremic toxins, such as p-cresyl sulphate (PCS), indoxyl sulphate (IS), phenylacetic acid, indole-3-acetic acid (IAA), hippuric acid (HA), and trimethylamine that promote local and systemic pro-inflammatory activity and associate with progression of cardiovascular disease and CKD [1]. Gut-derived uremic toxins and bacterial endotoxins may enhance PEW development, regardless of their well-known inflammatory effect, particularly because of their effect on skeletal muscle. Nonetheless, this effect of gut dysbiosis has been poorly explored (Table 1).
Table 1
Gut derived compounds related to dysbiosis and their possible effect on PEW development in CKD
Compounds
Association with CKD
Effect on PEW
References
Inflammatory cytokines
Anorexia
Muscle and fat wasting
Increased resting energy expenditure
[7, 61]
p-Cresyl sulphate
Lipolytic
Muscle fat infiltration
[32]
Indoxyl sulphate
Muscle catabolism and atrophy
[34, 35]
Glycine-conjugated compounds
Decreased muscle function and physical performance
[3638]
Acrolein
Muscle wasting
[44]
Endotoxins
Muscle wasting and atrophy
Increased resting energy expenditure
Anorexia
[45, 46]
SCFAs
Inflammation
Energy imbalance
Protein catabolism
[6, 48, 49]
[50]
Appetite regulation peptides
↑↓
Anorexia
[51, 53, 54, 56]
SCFAs short chain fatty acids
In rats, administration of PCS originating from bacterial degradation of tyrosine and phenylalanine promoted insulin resistance, and loss and redistribution of fat; of note, body composition changes included a decrease in white fat, while there was an increase in liver ectopic lipid content and muscle fat infiltration [38], which is also associated with mobility impairment [39]. In the same study, PCS decreased lipogenesis and increased lipolysis in human adipocytes, which suggests a catabolic effect affecting fat mass [38].
IS is an aromatic compound that derives from bacterial tryptophan metabolism. In CKD murine models, IS accumulates in skeletal muscle and promotes muscle loss and atrophy by inhibiting protein synthesis, cell proliferation and viability, increasing oxidative stress and inflammation, impairing mitochondrial function, and accelerating amino acid degradation [40]. In addition, as reported in the same paper, serum concentrations of IS in PD patients were found to be associated with decreased muscle mass, showing further subsequent reductions following 2 years of dialysis [40]. In an in vitro study, myoblast cells were treated with different gut-derived uremic toxins: IS, and to a lesser extent IAA, significantly inhibited cell proliferation, but only IS increased oxidative stress in a dose-dependent manner, and IS also increased inflammation, as well as myostatin and atrogin-1, which are negative regulators of muscle growth involved in muscular atrophy [41].
Gut bacterial metabolites have also been implicated as factors affecting muscle function and physical performance in both young and older healthy adults; hydrocinnamate, cinnamoylglycine, indolepropionate and HA were negatively associated with lower extremity muscle quality, an indicator of muscle function [42], and physical performance [43]. HA is a toxic solute derived from bacterial degradation of aromatic compounds such as aromatic amino acids, preservatives with benzoic acid, and contaminants as toluene or polyphenols, which inhibit glucose utilization in muscle cells, thus potentially increasing muscular weakness in CKD patients [44]. It has controversial effects, since high levels of HA derived from dietary antioxidant polyphenols, as catechin (green and black tea) or chlorogenic acid (coffee) have positive effects on muscle metabolism and performance [45] by promoting myogenic differentiation and myofiber regeneration [46]. It is possible that diverse dietary sources, gut dysbiosis and interactions with the uremic host environment could lead to the observed differences [47].
Polyamines, for their part, are metabolites from protein fermentation in the gut, which are required for normal cell proliferation and differentiation; in muscle cells, regulation of polyamines is associated with hypertrophy and restoring muscle after injury, and a shift in polyamine equilibrium can lead to dysregulation in muscle metabolism and growth [48]. In CKD patients, putrescine levels are higher, while spermidine and spermine levels are lower compared to subjects with normal kidney function. Additionally, there was an increase in spermidine and spermine degradation into acrolein, a toxic compound [49], which has inhibitory effects on myogenic differentiation and induces muscle wasting in animal models [50].
Finally, endotoxin or LPS, a phospholipid of the bacterial outer membrane, increases systemic inflammation, which induces muscle and fat catabolism and metabolic derangements (insulin resistance). Peripheral and central nervous system inflammatory effects also mediate LPS-induced muscle catabolism. Skeletal muscle has receptors for pathogen-associated molecules, catabolic hormones and proinflammatory cytokines, so that LPS may act by different action mechanisms to promote muscle wasting and atrophy; the most recognized mechanisms involve activation of the ubiquitin–proteasome and autophagy-lysosome pathways and down-regulation of insulin-like growth factor-1 [51]. Moreover, LPS exerts inflammatory effects in the central nervous system by stimulation of the melanocortin system that increases resting energy expenditure and weight loss, as well as by activation of hypothalamus NF-κB that increases anorexia and muscle wasting [52].
Recently, it has been shown that intestinal infections may activate the NLRP3 inflammasome stimulating the innate and adaptive immune responses, which in turn may increase muscle atrophy and wasting via caspase-1 activation [53]. Moreover, inflammasome signaling may be associated with hyperfiltration in some kidney diseases [54].

SCFA saves energy in the gut and has anti-inflammatory effects

Dietary fibers do not per se provide energy directly to humans, as we are unable to digest and absorb them, however, colonic bacteria can metabolize resistant starch and fermentable fibers to butyrate, propionate and acetate SCFAs, as well as to lactate and various gases. Butyrate is the main energy source of epithelial cells and promotes integrity of the intestinal wall; propionate is mostly metabolized in the liver, as a precursor in gluconeogenesis and lipogenesis; a proportion of the acetate is metabolized to glutamine (main energy source of the small intestine), while muscles can metabolize another part to obtain energy. SCFAs and other compounds derived from the metabolism of fibers may provide substantial amounts of energy, 1.5–2 kcal/g of fiber, contributing to preserve the “lost” energy from the small intestine, thus enhancing energy homeostasis. It has been proposed that it could be a healthy opportunity to conserve energy from the diet in poor communities or in individuals with compromised diets due to restrictions such as those recommended in CKD patients [7]. It has also been observed that obese subjects have higher relative abundance of Firmicutes and a lower amount of Bacteroidetes compared to lean subjects, and an increase in Bacteroidetes was associated with greater weight loss during the diet period [55]. In adolescents with obesity, the Firmicutes/Bacteroidetes ratio, the relative abundance of Bacteroidetes and Actinobacteria phylum as well as seven different genus bacteria (Actinomyces, Odoribacter, Oscilospira, Bifidobacterium, Streptococcus, Bacteroides, Faecalibacterium) were positively associated to total, visceral, subcutaneous and hepatic fat content, and to SCFA concentrations, suggesting that their gut microbiota profile has a better ability to extract energy from carbohydrates [56]. Although SCFAs are associated with efficient energy harvest in the gut and weight gain, it seems, on the basis of rodent models, that these effects can be linked to changes in energy utilization and expenditure, as SCFAs inhibit fat accumulation, enhance adaptive thermogenesis and fat oxidation (increase energy expenditure), and decrease markers of protein catabolism [57]; these effects could be associated with improvements in body composition.
On the other hand, SCFA modulates the inflammatory response, suppressing synthesis and release of cytokines in neutrophils, macrophages and other cells, and suppresses adhesion molecule expression in endothelial cells. Butyrate has the greatest anti-inflammatory effect, as it suppresses LPS-stimulated production of TNFα, IL-6 and nitric oxide, and increases IL-10 release. The main anti-inflammatory mechanism of action is the attenuation of histone deacetylase enzyme activity, which stimulates the increase in histone and nonhistone protein acetylation, such as that of NF-κb, modulating gene expression of cytokines [58].
The causes of anorexia, which further worsens PEW in CKD patients, include a range of factors such as retention of uremic solutes due to loss of residual renal function, inflammation, effects of the dialysis procedure, dietary restrictions, taste abnormalities, depression, gastrointestinal symptoms, comorbidities, pill burden, as well as alterations in hunger-satiety hormonal regulation [59].
In addition, the gut microbiota may have a significant role in satiety control. In healthy rats undergoing different food access and exercise interventions, Lactobacillus and Bifidobacterium genera were associated positively to leptin and negatively to ghrelin levels, suggesting that regulatory mechanisms of gut microbiota may influence food energy harvesting and body weight [60]. However, in disease states, gut microbiota and satiety mediators as ghrelin have different effects; in patients with polycystic ovary syndrome, ghrelin levels were negatively correlated with chronic inflammation-related gram-negative bacteria as Bacteroides, Escherichia/Shigella and Blautia species, and positively correlated with Akkermansia species, which are involved in maintaining intestinal integrity [61]. In children with acute lung injury, the administration of oral Lactobacillus acidophilus over 10 days increased ghrelin levels and decreased inflammation markers, suggesting a regulatory effect of gut microbiota on ghrelin levels [62]. Total ghrelin levels are increased in CKD patients, as ghrelin is mainly degraded by kidneys; however, this increase associates with an increase in obestatin and desacyl forms of ghrelin, which may induce effects that are opposite to those of the active orexigenic form of ghrelin, i.e., acyl ghrelin, thus decreasing appetite. On the other hand, serum leptin, the counteracting hormone of ghrelin, increases with the decrease in renal function, exacerbating the anorexigenic effect [63]. Nonetheless, there is a lack of studies on the impact of gut microbiota alterations on the levels of leptin and different forms of ghrelin, and on appetite in CKD patients.
Additionally, it has been suggested that microbiota-derived proteins may have a direct influence on satiety control. Administration of an Escherichia coli derived protein, caseinolytic protease, resulted in decreased food intake in mice, through neuronal central effects by activating the melanocortin-4 receptor and stimulating release of satietogenic gut hormones as glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) [64]. CKD patients, including those undergoing PD, show an increase in cholecystokinin and peptide YY, leading to early satiety and decrease in nutrient intake [65, 66]. Consequently, it is possible that the observed abundance of gram-negative bacteria (Proteobacteria) in patients on PD [17] might contribute to increased appetite abnormalities.
Amino acid imbalances, characterized by low levels of branched chain amino acids, are related to an anorexic pattern in uremia; serotonin production is increased along with higher availability of tryptophan in cerebrospinal fluid, thus suppressing appetite; this phenomenon is enhanced by inflammation and metabolic acidosis [59]. Serotonin balance may be altered by the availability of tryptophan and interactions among host-bacteria (diet, drugs); gut microbiota may affect serotonin availability by increasing tryptophan degradation (tryptophanase), by synthesizing tryptophan (tryptophan synthase) or by producing de novo serotonin direct from tryptophan. A wide range of bacterial families are involved in serotonin biosynthesis, including Bifidobacterium, Streptococcus, Escherichia, Enterococcus, Lactococcus, Lactobacillus and Clostridium [27, 67].
On the other hand, SCFAs have an anorectic effect by stimulating intestinal GLP-1 and PYY, and decreasing ghrelin release in the context of obesity, however, it seems that the appetite suppression effect occurs only with a high and acute dose of fiber or SCFAs [57]. In CKD, inflammation and uremic toxin retention are well recognized causes of PEW by inducing anorexia, decreasing nutrient intake, and increasing resting energy expenditure as well as muscle and fat catabolism [10, 68]. It is possible that the anti-inflammatory effect of SCFAs could be more important than the appetite suppressant and energy expenditure effect in preventing PEW, however, this warrants further research.
Current studies mainly focus on the relationship between gut microbiota, appetite and weight regulation in obesity. The results of these studies may suggest possible new therapeutic targets also in patients with under-nutrition conditions, such as PEW.

Gut microbiome manipulation and PEW in CKD

Modification of gut microbiota through nutritional and lifestyle interventions seems promising in CKD [69]. Studies in cancer cachexia, obesity, and aging settings show that modifications of gut microbiota using exercise or dietary means—including nutraceutical supplements with prebiotics, probiotics and synbiotics, or postbiotics (i.e., specific products from live bacteria such as butyrate)—may improve markers of muscle wasting; however, these studies have been carried out mostly in animal models and to date, there are few clinical trials [37].
To the best of our knowledge, no clinical trials involving CKD patients have attempted to specifically evaluate the effect of gut microbiota modification on PEW outcomes. Probiotics, prebiotics and symbiotics have been studied in CKD patients as a means to decrease inflammation and uremic toxins; however, there is limited evidence and results are controversial [70]. Nevertheless, a few studies analyzed nutritional status parameters related to the PEW syndrome. In a clinical trial to evaluate the effect of probiotics on glycemic control and oxidative stress in diabetic HD patients, the administration of a mixture of Lactobacillus acidophilus, Lactobacillus casei and Bifidobacterium bifidum over 12 weeks resulted in significant improvements in nutritional status as evaluated by means of subjective global assessment [71]. Intake of a mixture of 40 g of fermentable carbohydrates (inulin and potato starch) for 5 weeks, significantly increased calorie and protein intake, as well as body weight in CKD patients [72]. In HD patients, administration of resistant starch (16 g/day) increased calorie and fat intake compared to the placebo group [73]. In PD patients, the administration of a probiotic mixture (Bifidobacterium longum, Lactobacillus bulgaricus, and Streptococcus thermophilus) for two months increased adiposity parameters such as mid-arm circumference and triceps skinfold, as well as serum albumin [74]. Finally, Viramontes-Hörner et al. [75] found a significant reduction in frequency and severity of gastrointestinal symptoms in HD patients after 2 months of administration of a symbiotic product (inulin + Lactobacillus acidophilus and Bifidobacterium lactis), as well as a trend towards improvement of PEW (as assessed by subjective global assessment) in spite of a lower calorie intake. Several limitations of the previous interventional studies must be emphasized: nutritional parameters were evaluated as secondary outcomes, PEW was not specifically diagnosed, interventions were not homogeneous (either prebiotics or probiotics), and it was not clearly stated whether these interventions were able to beneficially modify gut microbiota, and subsequently improve nutritional parameters. Future research is warranted to clarify these issues.

Conclusions

Gut microbiota disturbances may contribute to PEW in CKD. Interactions between gut dysbiosis and PEW in patients with CKD are complex and involve many interlinked factors such as inflammation, retention of uremic solutes and hormonal abnormalities. Available evidence suggests bidirectional communication between gut microbiota and key factors of PEW, such as energy balance, appetite, nutrient intake, and muscle metabolism. There are many gut-derived compounds related to dysbiosis that may influence PEW development in CKD. Altogether these findings suggest that interventions to manipulate the intestinal microbiome could be an efficient strategy to improve nutritional status and thereby clinical outcomes in CKD patients. Further studies are warranted in this as yet largely unexplored field.

Acknowledgements

Baxter Novum is the result of a grant from Baxter Healthcare to Karolinska Institutet.

Declarations

Conflict of interest

Bengt Lindholm has been affiliated with Baxter Healthcare and is employed by Karolinska Institutet at Baxter Novum. None of the other authors declare any conflicts of interest.

Ethical statement

Ethical statement is not applicable for this manuscript.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Urologie

Kombi-Abonnement

Mit e.Med Urologie erhalten Sie Zugang zu den urologischen CME-Fortbildungen und Premium-Inhalten der urologischen Fachzeitschriften.

Literatur
1.
Zurück zum Zitat Ramezani A, Massy ZA, Meijers B, Evenepoel P, Vanholder R, Raj DS (2016) Role of the gut microbiome in uremia: a potential therapeutic target. Am J Kidney Dis 67(3):483–498PubMedCrossRef Ramezani A, Massy ZA, Meijers B, Evenepoel P, Vanholder R, Raj DS (2016) Role of the gut microbiome in uremia: a potential therapeutic target. Am J Kidney Dis 67(3):483–498PubMedCrossRef
2.
Zurück zum Zitat Rapozo DCM, Bernardazzi C, de Souza HSP (2017) Diet and microbiota in inflammatory bowel disease: the gut in disharmony. World J Gastroenterol 23(12):2124–2140PubMedPubMedCentralCrossRef Rapozo DCM, Bernardazzi C, de Souza HSP (2017) Diet and microbiota in inflammatory bowel disease: the gut in disharmony. World J Gastroenterol 23(12):2124–2140PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Blandino G, Inturri R, Lazzara F, di Rosa M, Malaguarnera L (2016) Impact of gut microbiota on diabetes mellitus. Diabetes Metab 42(5):303–315PubMedCrossRef Blandino G, Inturri R, Lazzara F, di Rosa M, Malaguarnera L (2016) Impact of gut microbiota on diabetes mellitus. Diabetes Metab 42(5):303–315PubMedCrossRef
4.
Zurück zum Zitat Dinan TG, Cryan JF (2017) The microbiome-gut-brain axis in health and disease. Gastroenterol Clin N Am 46(1):77–89CrossRef Dinan TG, Cryan JF (2017) The microbiome-gut-brain axis in health and disease. Gastroenterol Clin N Am 46(1):77–89CrossRef
5.
Zurück zum Zitat Pope JL, Tomkovich S, Yang Y, Jobin C (2017) Microbiota as a mediator of cancer progression and therapy. Transl Res 179:139–154PubMedCrossRef Pope JL, Tomkovich S, Yang Y, Jobin C (2017) Microbiota as a mediator of cancer progression and therapy. Transl Res 179:139–154PubMedCrossRef
7.
Zurück zum Zitat Ramakrishna BS (2013) Role of the gut microbiota in human nutrition and metabolism. J Gastroenterol Hepatol (Australia) 28(S4):9–17CrossRef Ramakrishna BS (2013) Role of the gut microbiota in human nutrition and metabolism. J Gastroenterol Hepatol (Australia) 28(S4):9–17CrossRef
8.
Zurück zum Zitat Moreau-Gaudry X, Jean G, Genet L, Lataillade D, Legrand E, Kuentz F et al (2014) A simple protein-energy wasting score predicts survival in maintenance hemodialysis patients. J Ren Nutr 24(6):395–400PubMedCrossRef Moreau-Gaudry X, Jean G, Genet L, Lataillade D, Legrand E, Kuentz F et al (2014) A simple protein-energy wasting score predicts survival in maintenance hemodialysis patients. J Ren Nutr 24(6):395–400PubMedCrossRef
9.
Zurück zum Zitat de Roij van Zuijdewijn CLM, Grooteman MPC, Bots ML, Blankestijn PJ, van den Dorpel MA, Nubé MJ et al (2016) Comparing tests assessing protein-energy wasting: relation with quality of life. J Ren Nutr 26(2):111–117PubMedCrossRef de Roij van Zuijdewijn CLM, Grooteman MPC, Bots ML, Blankestijn PJ, van den Dorpel MA, Nubé MJ et al (2016) Comparing tests assessing protein-energy wasting: relation with quality of life. J Ren Nutr 26(2):111–117PubMedCrossRef
10.
Zurück zum Zitat Carrero JJ, Stenvinkel P, Cuppari L, Ikizler TA, Kalantar-Zadeh K, Kaysen G et al (2013) Etiology of the protein-energy wasting syndrome in chronic kidney disease: a consensus statement from the International Society of Renal Nutrition and Metabolism (ISRNM). J Ren Nutr 23(2):77–90PubMedCrossRef Carrero JJ, Stenvinkel P, Cuppari L, Ikizler TA, Kalantar-Zadeh K, Kaysen G et al (2013) Etiology of the protein-energy wasting syndrome in chronic kidney disease: a consensus statement from the International Society of Renal Nutrition and Metabolism (ISRNM). J Ren Nutr 23(2):77–90PubMedCrossRef
11.
Zurück zum Zitat Rajilić-Stojanović M, de Vos WM (2014) The first 1000 cultured species of the human gastrointestinal microbiota. FEMS Microbiol Rev 38(5):996–1047PubMedCrossRef Rajilić-Stojanović M, de Vos WM (2014) The first 1000 cultured species of the human gastrointestinal microbiota. FEMS Microbiol Rev 38(5):996–1047PubMedCrossRef
12.
Zurück zum Zitat Greenhalgh K, Meyer KM, Aagaard KM, Wilmes P (2016) The human gut microbiome in health: establishment and resilience of microbiota over a lifetime. Environ Microbiol 18(7):2103–2116PubMedPubMedCentralCrossRef Greenhalgh K, Meyer KM, Aagaard KM, Wilmes P (2016) The human gut microbiome in health: establishment and resilience of microbiota over a lifetime. Environ Microbiol 18(7):2103–2116PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Xu KY, Xia GH, Lu JQ, Chen MX, Zhen X, Wang S et al (2017) Impaired renal function and dysbiosis of gut microbiota contribute to increased trimethylamine-N-oxide in chronic kidney disease patients. Sci Rep 7(1):1445PubMedPubMedCentralCrossRef Xu KY, Xia GH, Lu JQ, Chen MX, Zhen X, Wang S et al (2017) Impaired renal function and dysbiosis of gut microbiota contribute to increased trimethylamine-N-oxide in chronic kidney disease patients. Sci Rep 7(1):1445PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Wong J, Piceno YM, DeSantis TZ, Pahl M, Andersen GL, Vaziri ND (2014) Expansion of urease- and uricase-containing, indole- and p-cresol-forming and contraction of short-chain fatty acid-producing intestinal microbiota in ESRD. Am J Nephrol 39(3):230–237PubMedCrossRef Wong J, Piceno YM, DeSantis TZ, Pahl M, Andersen GL, Vaziri ND (2014) Expansion of urease- and uricase-containing, indole- and p-cresol-forming and contraction of short-chain fatty acid-producing intestinal microbiota in ESRD. Am J Nephrol 39(3):230–237PubMedCrossRef
15.
Zurück zum Zitat Poesen R, Windey K, Neven E, Kuypers D, de Preter V, Augustijns P et al (2016) The influence of CKD on colonic microbial metabolism. J Am Soc Nephrol 27(5):1389–1399PubMedCrossRef Poesen R, Windey K, Neven E, Kuypers D, de Preter V, Augustijns P et al (2016) The influence of CKD on colonic microbial metabolism. J Am Soc Nephrol 27(5):1389–1399PubMedCrossRef
16.
Zurück zum Zitat Teixeira RR, de Andrade LS, Pereira NBF, Montenegro H, Hoffmann C, Cuppari L (2022) Gut microbiota profile of patients on peritoneal dialysis: comparison with household contacts. Eur J Clin Nutr [Internet] (Available from: Epub ahead of print. PMID: 35906334). Teixeira RR, de Andrade LS, Pereira NBF, Montenegro H, Hoffmann C, Cuppari L (2022) Gut microbiota profile of patients on peritoneal dialysis: comparison with household contacts. Eur J Clin Nutr [Internet] (Available from: Epub ahead of print. PMID: 35906334).
17.
Zurück zum Zitat Crespo-Salgado J, Vehaskari VM, Stewart T, Ferris M, Zhang Q, Wang G et al (2016) Intestinal microbiota in pediatric patients with end stage renal disease: a Midwest Pediatric Nephrology Consortium study. Microbiome 4(1):50PubMedPubMedCentralCrossRef Crespo-Salgado J, Vehaskari VM, Stewart T, Ferris M, Zhang Q, Wang G et al (2016) Intestinal microbiota in pediatric patients with end stage renal disease: a Midwest Pediatric Nephrology Consortium study. Microbiome 4(1):50PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Luo D, Zhao W, Lin Z, Wu J, Lin H, Li Y et al (2021) the effects of hemodialysis and peritoneal dialysis on the gut microbiota of end-stage renal disease patients, and the relationship between gut microbiota and patient prognoses. Front Cell Infect Microbiol 11:579386PubMedPubMedCentralCrossRef Luo D, Zhao W, Lin Z, Wu J, Lin H, Li Y et al (2021) the effects of hemodialysis and peritoneal dialysis on the gut microbiota of end-stage renal disease patients, and the relationship between gut microbiota and patient prognoses. Front Cell Infect Microbiol 11:579386PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Hu J, Zhong X, Yan J, Zhou D, Qin D, Xiao X et al (2020) High-throughput sequencing analysis of intestinal flora changes in ESRD and CKD patients. BMC Nephrol 21(1):12PubMedPubMedCentralCrossRef Hu J, Zhong X, Yan J, Zhou D, Qin D, Xiao X et al (2020) High-throughput sequencing analysis of intestinal flora changes in ESRD and CKD patients. BMC Nephrol 21(1):12PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Vaziri ND, Zhao YY, Pahl Mv (2016) Altered intestinal microbial flora and impaired epithelial barrier structure and function in CKD: the nature, mechanisms, consequences and potential treatment. Nephrol Dial Transplant 31(5):737–746PubMedCrossRef Vaziri ND, Zhao YY, Pahl Mv (2016) Altered intestinal microbial flora and impaired epithelial barrier structure and function in CKD: the nature, mechanisms, consequences and potential treatment. Nephrol Dial Transplant 31(5):737–746PubMedCrossRef
21.
Zurück zum Zitat Manley HJ, Garvin CG, Drayer DK, Reid GM, Bender WL, Neufeld TK et al (2004) Medication prescribing patterns in ambulatory haemodialysis patients: comparisons of USRDS to a large not-for-profit dialysis provider. Nephrol Dial Transplant 19(7):1842–1848PubMedCrossRef Manley HJ, Garvin CG, Drayer DK, Reid GM, Bender WL, Neufeld TK et al (2004) Medication prescribing patterns in ambulatory haemodialysis patients: comparisons of USRDS to a large not-for-profit dialysis provider. Nephrol Dial Transplant 19(7):1842–1848PubMedCrossRef
22.
Zurück zum Zitat Biruete A, Hill Gallant KM, Lindemann SR, Wiese GN, Chen NX, Moe SM (2019) Phosphate binders and nonphosphate effects in the gastrointestinal tract. J Ren Nutr 30(1):4–10PubMedPubMedCentralCrossRef Biruete A, Hill Gallant KM, Lindemann SR, Wiese GN, Chen NX, Moe SM (2019) Phosphate binders and nonphosphate effects in the gastrointestinal tract. J Ren Nutr 30(1):4–10PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Lange K, Buerger M, Stallmach A, Bruns T (2016) Effects of antibiotics on gut microbiota. Dig Dis 34(3):260–268PubMedCrossRef Lange K, Buerger M, Stallmach A, Bruns T (2016) Effects of antibiotics on gut microbiota. Dig Dis 34(3):260–268PubMedCrossRef
25.
Zurück zum Zitat Koppe L, Mafra D, Fouque D (2015) Probiotics and chronic kidney disease. Kidney Int 88(5):958–966PubMedCrossRef Koppe L, Mafra D, Fouque D (2015) Probiotics and chronic kidney disease. Kidney Int 88(5):958–966PubMedCrossRef
26.
Zurück zum Zitat Lau WL, Kalantar-Zadeh K, Vaziri ND (2015) The gut as a source of inflammation in chronic kidney disease. Nephron 130(2):92–98PubMedCrossRef Lau WL, Kalantar-Zadeh K, Vaziri ND (2015) The gut as a source of inflammation in chronic kidney disease. Nephron 130(2):92–98PubMedCrossRef
27.
Zurück zum Zitat Jazani N, Savoj J, Lustgarten M, Lau W, Vaziri N (2019) Impact of gut dysbiosis on neurohormonal pathways in chronic kidney disease. Diseases 7(1):21PubMedPubMedCentralCrossRef Jazani N, Savoj J, Lustgarten M, Lau W, Vaziri N (2019) Impact of gut dysbiosis on neurohormonal pathways in chronic kidney disease. Diseases 7(1):21PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Wang F, Jiang H, Shi K, Ren Y, Zhang P, Cheng S (2012) Gut bacterial translocation is associated with microinflammation in end-stage renal disease patients. Nephrology 17(8):733–738PubMedCrossRef Wang F, Jiang H, Shi K, Ren Y, Zhang P, Cheng S (2012) Gut bacterial translocation is associated with microinflammation in end-stage renal disease patients. Nephrology 17(8):733–738PubMedCrossRef
29.
Zurück zum Zitat Hauser AB, Stinghen AEM, Gonçalves SM, Bucharles S, Pecoits-Filho R (2011) A gut feeling on endotoxemia: causes and consequences in chronic kidney disease. Nephron Clin Pract 118(2):165–172CrossRef Hauser AB, Stinghen AEM, Gonçalves SM, Bucharles S, Pecoits-Filho R (2011) A gut feeling on endotoxemia: causes and consequences in chronic kidney disease. Nephron Clin Pract 118(2):165–172CrossRef
30.
Zurück zum Zitat Tidjani Alou M, Million M, Traore SI, Mouelhi D, Khelaifia S, Bachar D et al (2017) Gut bacteria missing in severe acute malnutrition, can we identify potential probiotics by culturomics? Front Microbiol 8:899PubMedPubMedCentralCrossRef Tidjani Alou M, Million M, Traore SI, Mouelhi D, Khelaifia S, Bachar D et al (2017) Gut bacteria missing in severe acute malnutrition, can we identify potential probiotics by culturomics? Front Microbiol 8:899PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Ghosh TS, sen GS, Bhattacharya T, Yadav D, Barik A, Chowdhury A et al (2014) Gut microbiomes of Indian children of varying nutritional status. PLoS ONE 9(4):e95547PubMedPubMedCentralCrossRef Ghosh TS, sen GS, Bhattacharya T, Yadav D, Barik A, Chowdhury A et al (2014) Gut microbiomes of Indian children of varying nutritional status. PLoS ONE 9(4):e95547PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Kumar M, Ji B, Babaei P, Das P, Lappa D, Ramakrishnan G et al (2018) Gut microbiota dysbiosis is associated with malnutrition and reduced plasma amino acid levels: Lessons from genome-scale metabolic modeling. Metab Eng 49:128–142PubMedPubMedCentralCrossRef Kumar M, Ji B, Babaei P, Das P, Lappa D, Ramakrishnan G et al (2018) Gut microbiota dysbiosis is associated with malnutrition and reduced plasma amino acid levels: Lessons from genome-scale metabolic modeling. Metab Eng 49:128–142PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Mack I, Penders J, Cook J, Dugmore J, Mazurak N, Enck P (2018) Is the impact of starvation on the gut microbiota specific or unspecific to anorexia nervosa? A narrative review based on a systematic literature search. Curr Neuropharmacol 16(8):1131–1149PubMedPubMedCentralCrossRef Mack I, Penders J, Cook J, Dugmore J, Mazurak N, Enck P (2018) Is the impact of starvation on the gut microbiota specific or unspecific to anorexia nervosa? A narrative review based on a systematic literature search. Curr Neuropharmacol 16(8):1131–1149PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Lin TY, Hung SC (2021) Association of subjective global assessment of nutritional status with gut microbiota in hemodialysis patients: a case-control study. Nephrol Dial Transplant 36(6):1104–1111PubMedCrossRef Lin TY, Hung SC (2021) Association of subjective global assessment of nutritional status with gut microbiota in hemodialysis patients: a case-control study. Nephrol Dial Transplant 36(6):1104–1111PubMedCrossRef
35.
Zurück zum Zitat Hu J, Zhong X, Liu Y, Yan J, Zhou D, Qin D et al (2022) Correlation between intestinal flora disruption and protein–energy wasting in patients with end-stage renal disease. BMC Nephrol 23(1):1–12CrossRef Hu J, Zhong X, Liu Y, Yan J, Zhou D, Qin D et al (2022) Correlation between intestinal flora disruption and protein–energy wasting in patients with end-stage renal disease. BMC Nephrol 23(1):1–12CrossRef
36.
Zurück zum Zitat Evenepoel P, Meijers BKI, Bammens BRM, Verbeke K (2009) Uremic toxins originating from colonic microbial metabolism. Kidney Int Suppl 76(114):S12–S19CrossRef Evenepoel P, Meijers BKI, Bammens BRM, Verbeke K (2009) Uremic toxins originating from colonic microbial metabolism. Kidney Int Suppl 76(114):S12–S19CrossRef
37.
Zurück zum Zitat Grosicki GJ, Fielding RA, Lustgarten MS (2018) Gut microbiota contribute to age-related changes in skeletal muscle size, composition, and function: biological basis for a gut-muscle axis. Calcif Tissue Int 102(4):433–442PubMedCrossRef Grosicki GJ, Fielding RA, Lustgarten MS (2018) Gut microbiota contribute to age-related changes in skeletal muscle size, composition, and function: biological basis for a gut-muscle axis. Calcif Tissue Int 102(4):433–442PubMedCrossRef
38.
Zurück zum Zitat Koppe L, Pillon NJ, Vella RE, Croze ML, Pelletier CC, Chambert S et al (2013) p-Cresyl sulfate promotes insulin resistance associated with CKD. J Am Soc Nephrol 24(1):88–99PubMedCrossRef Koppe L, Pillon NJ, Vella RE, Croze ML, Pelletier CC, Chambert S et al (2013) p-Cresyl sulfate promotes insulin resistance associated with CKD. J Am Soc Nephrol 24(1):88–99PubMedCrossRef
39.
Zurück zum Zitat Robles PG, Sussman MS, Naraghi A, Brooks D, Goldstein RS, White LM et al (2015) Intramuscular fat infiltration contributes to impaired muscle function in COPD. Med Sci Sports Exerc 47(7):1334–1341PubMedCrossRef Robles PG, Sussman MS, Naraghi A, Brooks D, Goldstein RS, White LM et al (2015) Intramuscular fat infiltration contributes to impaired muscle function in COPD. Med Sci Sports Exerc 47(7):1334–1341PubMedCrossRef
40.
Zurück zum Zitat Sato E, Mori T, Mishima E, Suzuki A, Sugawara S, Kurasawa N et al (2016) Metabolic alterations by indoxyl sulfate in skeletal muscle induce uremic sarcopenia in chronic kidney disease. Sci Rep 6:36618PubMedPubMedCentralCrossRef Sato E, Mori T, Mishima E, Suzuki A, Sugawara S, Kurasawa N et al (2016) Metabolic alterations by indoxyl sulfate in skeletal muscle induce uremic sarcopenia in chronic kidney disease. Sci Rep 6:36618PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Enoki Y, Watanabe H, Arake R, Sugimoto R, Imafuku T, Tominaga Y et al (2016) Indoxyl sulfate potentiates skeletal muscle atrophy by inducing the oxidative stress-mediated expression of myostatin and atrogin-1. Sci Rep 6:32084PubMedPubMedCentralCrossRef Enoki Y, Watanabe H, Arake R, Sugimoto R, Imafuku T, Tominaga Y et al (2016) Indoxyl sulfate potentiates skeletal muscle atrophy by inducing the oxidative stress-mediated expression of myostatin and atrogin-1. Sci Rep 6:32084PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Lustgarten MS, Price LL, Fielding RA (2015) Analytes and metabolites associated with muscle quality in young, healthy adults. Med Sci Sports Exerc 47(8):1659–1664PubMedPubMedCentralCrossRef Lustgarten MS, Price LL, Fielding RA (2015) Analytes and metabolites associated with muscle quality in young, healthy adults. Med Sci Sports Exerc 47(8):1659–1664PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Lustgarten MS, Price LL, Chalé A, Fielding RA (2014) Metabolites related to gut bacterial metabolism, peroxisome proliferator-activated receptor-alpha activation, and insulin sensitivity are associated with physical function in functionally-limited older adults. Aging Cell 13(5):918–925PubMedPubMedCentralCrossRef Lustgarten MS, Price LL, Chalé A, Fielding RA (2014) Metabolites related to gut bacterial metabolism, peroxisome proliferator-activated receptor-alpha activation, and insulin sensitivity are associated with physical function in functionally-limited older adults. Aging Cell 13(5):918–925PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Spustová V, Dzúrik R, Geryková M (1987) Hippurate participation in the inhibition of glucose utilization in renal failure. Czech Med 10(2):79–89PubMed Spustová V, Dzúrik R, Geryková M (1987) Hippurate participation in the inhibition of glucose utilization in renal failure. Czech Med 10(2):79–89PubMed
45.
Zurück zum Zitat Kim H, Suzuki T, Saito K, Yoshida H, Kojima N, Kim M et al (2013) Effects of exercise and tea catechins on muscle mass, strength and walking ability in community-dwelling elderly Japanese sarcopenic women: a randomized controlled trial. Geriatr Gerontol Int 13(2):458–465PubMedCrossRef Kim H, Suzuki T, Saito K, Yoshida H, Kojima N, Kim M et al (2013) Effects of exercise and tea catechins on muscle mass, strength and walking ability in community-dwelling elderly Japanese sarcopenic women: a randomized controlled trial. Geriatr Gerontol Int 13(2):458–465PubMedCrossRef
46.
Zurück zum Zitat Kim AR, Kim KM, Byun MR, Hwang JH, il PJ, Oh HT et al (2017) Catechins activate muscle stem cells by Myf5 induction and stimulate muscle regeneration. Biochem Biophys Res Commun 489(2):142–148PubMedCrossRef Kim AR, Kim KM, Byun MR, Hwang JH, il PJ, Oh HT et al (2017) Catechins activate muscle stem cells by Myf5 induction and stimulate muscle regeneration. Biochem Biophys Res Commun 489(2):142–148PubMedCrossRef
47.
Zurück zum Zitat Lees HJ, Swann JR, Wilson ID, Nicholson JK, Holmes E (2013) Hippurate: The natural history of a mammalian-microbial cometabolite. J Proteome Res 12(4):1527–1546PubMedCrossRef Lees HJ, Swann JR, Wilson ID, Nicholson JK, Holmes E (2013) Hippurate: The natural history of a mammalian-microbial cometabolite. J Proteome Res 12(4):1527–1546PubMedCrossRef
48.
Zurück zum Zitat Lee NKL, Maclean HE (2011) Polyamines, androgens, and skeletal muscle hypertrophy. J Cell Physiol 226(6):1453–1460PubMedCrossRef Lee NKL, Maclean HE (2011) Polyamines, androgens, and skeletal muscle hypertrophy. J Cell Physiol 226(6):1453–1460PubMedCrossRef
49.
Zurück zum Zitat Igarashi K, Ueda S, Yoshida K, Kashiwagi K (2006) Polyamines in renal failure. Amino Acids 31(4):477–483PubMedCrossRef Igarashi K, Ueda S, Yoshida K, Kashiwagi K (2006) Polyamines in renal failure. Amino Acids 31(4):477–483PubMedCrossRef
50.
Zurück zum Zitat Chen HJ, Wang CC, Chan DC, Chiu CY, sen YR, Liu SH (2019) Adverse effects of acrolein, a ubiquitous environmental toxicant, on muscle regeneration and mass. J Cachexia Sarcopenia Muscle 10(1):165–176PubMedCrossRef Chen HJ, Wang CC, Chan DC, Chiu CY, sen YR, Liu SH (2019) Adverse effects of acrolein, a ubiquitous environmental toxicant, on muscle regeneration and mass. J Cachexia Sarcopenia Muscle 10(1):165–176PubMedCrossRef
52.
Zurück zum Zitat Duan K, Chen Q, Cheng M, Zhao C, Lin Z, Tan S et al (2016) Hypothalamic activation is essential for endotoxemia-induced acute muscle wasting. Sci Rep 6:38544PubMedPubMedCentralCrossRef Duan K, Chen Q, Cheng M, Zhao C, Lin Z, Tan S et al (2016) Hypothalamic activation is essential for endotoxemia-induced acute muscle wasting. Sci Rep 6:38544PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Liu Y, Wang D, Li T, Yang F, Li Z, Bai X et al (2022) The role of NLRP3 inflammasome in inflammation-related skeletal muscle atrophy. Front Immunol 13:1035709PubMedPubMedCentralCrossRef Liu Y, Wang D, Li T, Yang F, Li Z, Bai X et al (2022) The role of NLRP3 inflammasome in inflammation-related skeletal muscle atrophy. Front Immunol 13:1035709PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Moriconi D, Antonioli L, Masi S, Bellini R, Pellegrini C, Rebelos E et al (2022) Glomerular hyperfiltration in morbid obesity: role of the inflammasome signalling. Nephrology 27(8):673–680PubMedCrossRef Moriconi D, Antonioli L, Masi S, Bellini R, Pellegrini C, Rebelos E et al (2022) Glomerular hyperfiltration in morbid obesity: role of the inflammasome signalling. Nephrology 27(8):673–680PubMedCrossRef
55.
Zurück zum Zitat Ley RE, Turnbaugh PJ, Klein S, Gordon JI (2006) Microbial ecology: human gut microbes associated with obesity. Nature 444(7122):1022–1023PubMedCrossRef Ley RE, Turnbaugh PJ, Klein S, Gordon JI (2006) Microbial ecology: human gut microbes associated with obesity. Nature 444(7122):1022–1023PubMedCrossRef
56.
Zurück zum Zitat Goffredo M, Mass K, Parks EJ, Wagner DA, McClure EA, Graf J et al (2016) Role of gut microbiota and short chain fatty acids in modulating energy harvest and fat partitioning in youth. J Clin Endocrinol Metab 101(11):4367–4376PubMedPubMedCentralCrossRef Goffredo M, Mass K, Parks EJ, Wagner DA, McClure EA, Graf J et al (2016) Role of gut microbiota and short chain fatty acids in modulating energy harvest and fat partitioning in youth. J Clin Endocrinol Metab 101(11):4367–4376PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Byrne CS, Chambers ES, Morrison DJ, Frost G (2015) The role of short chain fatty acids in appetite regulation and energy homeostasis. Int J Obes 39(9):1331–1338CrossRef Byrne CS, Chambers ES, Morrison DJ, Frost G (2015) The role of short chain fatty acids in appetite regulation and energy homeostasis. Int J Obes 39(9):1331–1338CrossRef
59.
Zurück zum Zitat Carrero JJ (2011) Mechanisms of altered regulation of food intake in chronic kidney disease. J Ren Nutr 21(1):7–11PubMedCrossRef Carrero JJ (2011) Mechanisms of altered regulation of food intake in chronic kidney disease. J Ren Nutr 21(1):7–11PubMedCrossRef
60.
Zurück zum Zitat Queipo-Ortuño MI, Seoane LM, Murri M, Pardo M, Gomez-Zumaquero JM, Cardona F et al (2013) Gut microbiota composition in male rat models under different nutritional status and physical activity and its association with serum leptin and ghrelin levels. PLoS ONE 8(5):e65465PubMedPubMedCentralCrossRef Queipo-Ortuño MI, Seoane LM, Murri M, Pardo M, Gomez-Zumaquero JM, Cardona F et al (2013) Gut microbiota composition in male rat models under different nutritional status and physical activity and its association with serum leptin and ghrelin levels. PLoS ONE 8(5):e65465PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Liu R, Zhang C, Shi Y, Zhang F, Li L, Wang X et al (2017) Dysbiosis of gut microbiota associated with clinical parameters in polycystic ovary syndrome. Front Microbiol 8:324PubMedPubMedCentral Liu R, Zhang C, Shi Y, Zhang F, Li L, Wang X et al (2017) Dysbiosis of gut microbiota associated with clinical parameters in polycystic ovary syndrome. Front Microbiol 8:324PubMedPubMedCentral
62.
Zurück zum Zitat Wang Y, Gao L, Yang Z, Chen F, Zhang Y (2018) Effects of probiotics on ghrelin and lungs in children with acute lung injury: a double-blind randomized, controlled trial. Pediatr Pulmonol 53(2):197–203PubMedCrossRef Wang Y, Gao L, Yang Z, Chen F, Zhang Y (2018) Effects of probiotics on ghrelin and lungs in children with acute lung injury: a double-blind randomized, controlled trial. Pediatr Pulmonol 53(2):197–203PubMedCrossRef
63.
Zurück zum Zitat Gunta SS, Mak RH (2013) Ghrelin and leptin pathophysiology in chronic kidney disease. Pediatr Nephrol 28(4):611–616PubMedCrossRef Gunta SS, Mak RH (2013) Ghrelin and leptin pathophysiology in chronic kidney disease. Pediatr Nephrol 28(4):611–616PubMedCrossRef
64.
Zurück zum Zitat Breton J, Tennoune N, Lucas N, Francois M, Legrand R, Jacquemot J et al (2016) Gut commensal E. coli proteins activate host satiety pathways following nutrient-induced bacterial growth. Cell Metab 23(2):324–334PubMedCrossRef Breton J, Tennoune N, Lucas N, Francois M, Legrand R, Jacquemot J et al (2016) Gut commensal E. coli proteins activate host satiety pathways following nutrient-induced bacterial growth. Cell Metab 23(2):324–334PubMedCrossRef
65.
Zurück zum Zitat Wright M, Woodrow G, O’Brien S, Armstrong E, King N, Dye L et al (2004) Cholecystokinin and leptin: their influence upon the eating behaviour and nutrient intake of dialysis patients. Nephrol Dial Transplant 19(1):133–140PubMedCrossRef Wright M, Woodrow G, O’Brien S, Armstrong E, King N, Dye L et al (2004) Cholecystokinin and leptin: their influence upon the eating behaviour and nutrient intake of dialysis patients. Nephrol Dial Transplant 19(1):133–140PubMedCrossRef
66.
Zurück zum Zitat Pérez-Fontán M, Cordido F, Rodríguez-Carmona A, Penín M, Díaz-Cambre H, López-Muñiz A et al (2008) Short-term regulation of peptide YY secretion by a mixed meal or peritoneal glucose-based dialysate in patients with chronic renal failure. Nephrol Dial Transplant 23(11):3696–3703PubMedCrossRef Pérez-Fontán M, Cordido F, Rodríguez-Carmona A, Penín M, Díaz-Cambre H, López-Muñiz A et al (2008) Short-term regulation of peptide YY secretion by a mixed meal or peritoneal glucose-based dialysate in patients with chronic renal failure. Nephrol Dial Transplant 23(11):3696–3703PubMedCrossRef
67.
Zurück zum Zitat Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P, Ma L et al (2015) Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 161(2):264–276PubMedPubMedCentralCrossRef Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P, Ma L et al (2015) Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 161(2):264–276PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Jankowska M, Cobo G, Lindholm B, Stenvinkel P (2017) Inflammation and protein-energy wasting in the uremic milieu. Contrib Nephrol 191:58–71PubMedCrossRef Jankowska M, Cobo G, Lindholm B, Stenvinkel P (2017) Inflammation and protein-energy wasting in the uremic milieu. Contrib Nephrol 191:58–71PubMedCrossRef
69.
Zurück zum Zitat Mafra D, Borges N, Alvarenga L, Esgalhado M, Cardozo L, Lindholm B et al (2019) Dietary components that may influence the disturbed gut microbiota in chronic kidney disease. Nutrients 11(3):496PubMedPubMedCentralCrossRef Mafra D, Borges N, Alvarenga L, Esgalhado M, Cardozo L, Lindholm B et al (2019) Dietary components that may influence the disturbed gut microbiota in chronic kidney disease. Nutrients 11(3):496PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat McFarlane C, Ramos CI, Johnson DW, Campbell KL (2019) Prebiotic, probiotic, and synbiotic supplementation in chronic kidney disease: a systematic review and meta-analysis. J Ren Nutr 29(3):209–220PubMedCrossRef McFarlane C, Ramos CI, Johnson DW, Campbell KL (2019) Prebiotic, probiotic, and synbiotic supplementation in chronic kidney disease: a systematic review and meta-analysis. J Ren Nutr 29(3):209–220PubMedCrossRef
71.
Zurück zum Zitat Soleimani A, Zarrati Mojarrad M, Bahmani F, Taghizadeh M, Ramezani M, Tajabadi-Ebrahimi M et al (2017) Probiotic supplementation in diabetic hemodialysis patients has beneficial metabolic effects. Kidney Int 91(2):435–442PubMedCrossRef Soleimani A, Zarrati Mojarrad M, Bahmani F, Taghizadeh M, Ramezani M, Tajabadi-Ebrahimi M et al (2017) Probiotic supplementation in diabetic hemodialysis patients has beneficial metabolic effects. Kidney Int 91(2):435–442PubMedCrossRef
72.
Zurück zum Zitat Younes H, Egret N, Hadj-Abdelkader M, Rémésy C, Demigné C, Gueret C et al (2006) Fermentable carbohydrate supplementation alters nitrogen excretion in chronic renal failure. J Ren Nutr 16(1):67–74PubMedCrossRef Younes H, Egret N, Hadj-Abdelkader M, Rémésy C, Demigné C, Gueret C et al (2006) Fermentable carbohydrate supplementation alters nitrogen excretion in chronic renal failure. J Ren Nutr 16(1):67–74PubMedCrossRef
73.
Zurück zum Zitat Esgalhado M, Kemp JA, Azevedo R, Paiva BR, Stockler-Pinto MB, Dolenga CJ et al (2018) Could resistant starch supplementation improve inflammatory and oxidative stress biomarkers and uremic toxins levels in hemodialysis patients? A pilot randomized controlled trial. Food Funct 9(12):6508–6516PubMedCrossRef Esgalhado M, Kemp JA, Azevedo R, Paiva BR, Stockler-Pinto MB, Dolenga CJ et al (2018) Could resistant starch supplementation improve inflammatory and oxidative stress biomarkers and uremic toxins levels in hemodialysis patients? A pilot randomized controlled trial. Food Funct 9(12):6508–6516PubMedCrossRef
74.
Zurück zum Zitat Pan Y, Yang L, Dai B, Lin B, Lin S, Lin E (2021) Effects of probiotics on malnutrition and health-related quality of life in patients undergoing peritoneal dialysis: a randomized controlled trial. J Ren Nutr 31(2):199–205PubMedCrossRef Pan Y, Yang L, Dai B, Lin B, Lin S, Lin E (2021) Effects of probiotics on malnutrition and health-related quality of life in patients undergoing peritoneal dialysis: a randomized controlled trial. J Ren Nutr 31(2):199–205PubMedCrossRef
75.
Zurück zum Zitat Viramontes-Hörner D, Márquez-Sandoval F, Martín-del-Campo F, Vizmanos-Lamotte B, Sandoval-Rodríguez A, Armendáriz-Borunda J et al (2015) Effect of a symbiotic gel (Lactobacillus acidophilus + Bifidobacterium lactis + Inulin) on presence and severity of gastrointestinal symptoms in hemodialysis patients. J Ren Nutr 25(3):284–291PubMedCrossRef Viramontes-Hörner D, Márquez-Sandoval F, Martín-del-Campo F, Vizmanos-Lamotte B, Sandoval-Rodríguez A, Armendáriz-Borunda J et al (2015) Effect of a symbiotic gel (Lactobacillus acidophilus + Bifidobacterium lactis + Inulin) on presence and severity of gastrointestinal symptoms in hemodialysis patients. J Ren Nutr 25(3):284–291PubMedCrossRef
Metadaten
Titel
Gut microbiota disturbances and protein-energy wasting in chronic kidney disease: a narrative review
verfasst von
Fabiola Martín-del-Campo
Carla Maria Avesani
Peter Stenvinkel
Bengt Lindholm
Alfonso M. Cueto-Manzano
Laura Cortés-Sanabria
Publikationsdatum
23.01.2023
Verlag
Springer International Publishing
Erschienen in
Journal of Nephrology / Ausgabe 3/2023
Print ISSN: 1121-8428
Elektronische ISSN: 1724-6059
DOI
https://doi.org/10.1007/s40620-022-01560-1

Weitere Artikel der Ausgabe 3/2023

Journal of Nephrology 3/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

„Jeder Fall von plötzlichem Tod muss obduziert werden!“

17.05.2024 Plötzlicher Herztod Nachrichten

Ein signifikanter Anteil der Fälle von plötzlichem Herztod ist genetisch bedingt. Um ihre Verwandten vor diesem Schicksal zu bewahren, sollten jüngere Personen, die plötzlich unerwartet versterben, ausnahmslos einer Autopsie unterzogen werden.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Schlechtere Vorhofflimmern-Prognose bei kleinem linken Ventrikel

17.05.2024 Vorhofflimmern Nachrichten

Nicht nur ein vergrößerter, sondern auch ein kleiner linker Ventrikel ist bei Vorhofflimmern mit einer erhöhten Komplikationsrate assoziiert. Der Zusammenhang besteht nach Daten aus China unabhängig von anderen Risikofaktoren.

Semaglutid bei Herzinsuffizienz: Wie erklärt sich die Wirksamkeit?

17.05.2024 Herzinsuffizienz Nachrichten

Bei adipösen Patienten mit Herzinsuffizienz des HFpEF-Phänotyps ist Semaglutid von symptomatischem Nutzen. Resultiert dieser Benefit allein aus der Gewichtsreduktion oder auch aus spezifischen Effekten auf die Herzinsuffizienz-Pathogenese? Eine neue Analyse gibt Aufschluss.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.