Chronic kidney disease, uremic toxins and microbiota

in Microbiota and Host
Authors:
Zeliha Rumanli University of Health Sciences, Gulhane Faculty of Pharmacy, Department of Pharmacology, Ankar a, Türkiye

Search for other papers by Zeliha Rumanli in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0003-0550-6281
,
Ismail Mert Vural University of Health Sciences, Gulhane Faculty of Pharmacy, Department of Pharmacology, Ankar a, Türkiye

Search for other papers by Ismail Mert Vural in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0002-0011-9023
, and
Gulcin Alp Avci University of Health Sciences, Gulhane Faculty of Dentistry, Department of Basic Medical Sciences, Ankara, Türkiye

Search for other papers by Gulcin Alp Avci in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0003-4049-0756

Correspondence should be addressed to Z Rumanli: zeliha.rumanli@sbu.edu.tr
Open access
Sign up for journal news

Chronic kidney disease (CKD) is a progressive condition that is associated with a number of serious cardiovascular comorbidities. These are caused by the accumulation of uremic retention solutes (URSs) such as protein-bound uremic toxins, which include indoxyl sulfate, p-cresyl sulfate and trimethylamine-N-oxide. These toxins are metabolized from dietary precursors by the gut microbiota and liver enzymes. Elevated levels of uremic toxins intensify renal and cardiovascular tissue damage by augmenting oxidative stress and inflammation, culminating in adverse outcomes such as atherosclerosis, fibrosis and endothelial dysfunction. Kidney transplantation has been demonstrated to normalize these URS levels, thereby underscoring the pivotal role of renal function in the clearance of these toxins. Conversely, alterations in the composition of the gut microbiota may provide a potential avenue for the reduction of uremic toxin levels and the associated complications. In this review, studies from the 2020 to 2024 period were examined, and the microbiota taxa that are particularly relevant to uremic toxin production were grouped. In addition, their roles in the pathophysiology of CKD were addressed, thereby underscoring the significance of the gut–kidney axis. In the study, the taxa that have attracted attention in studies conducted in the relevant years were discussed separately according to their phylum, family, genus and species.

Abstract

Chronic kidney disease (CKD) is a progressive condition that is associated with a number of serious cardiovascular comorbidities. These are caused by the accumulation of uremic retention solutes (URSs) such as protein-bound uremic toxins, which include indoxyl sulfate, p-cresyl sulfate and trimethylamine-N-oxide. These toxins are metabolized from dietary precursors by the gut microbiota and liver enzymes. Elevated levels of uremic toxins intensify renal and cardiovascular tissue damage by augmenting oxidative stress and inflammation, culminating in adverse outcomes such as atherosclerosis, fibrosis and endothelial dysfunction. Kidney transplantation has been demonstrated to normalize these URS levels, thereby underscoring the pivotal role of renal function in the clearance of these toxins. Conversely, alterations in the composition of the gut microbiota may provide a potential avenue for the reduction of uremic toxin levels and the associated complications. In this review, studies from the 2020 to 2024 period were examined, and the microbiota taxa that are particularly relevant to uremic toxin production were grouped. In addition, their roles in the pathophysiology of CKD were addressed, thereby underscoring the significance of the gut–kidney axis. In the study, the taxa that have attracted attention in studies conducted in the relevant years were discussed separately according to their phylum, family, genus and species.

Introduction

Chronic kidney disease (CKD) patients suffer from cardiovascular comorbidities, which lead to poor survival rates. Apart from traditional risk factors, such as diabetes mellitus and hypertension, elevated circulating metabolites, known as uremic retention solutes (URSs), accelerate adverse cardiovascular-related outcomes. Epidemiological findings are supported by animal models demonstrating how high levels of URSs can damage renal and cardiovascular tissue. L-carnitine and choline, tryptophan and phenylalanine and tyrosine are dietary forerunners of TMAO (trimethylamine-N-oxide), IS (indoxyl sulfate) and PCS (p-cresyl sulfate), respectively, which are altered by the intestinal microbiota to form intermediary metabolites that are then transformed via hepatic enzymes to form the ultimate URS. These URSs are expelled via the kidneys in healthy adults, leading to low levels of circulation. Elevated blood URS levels have been observed in CKD patients experiencing kidney failure. Following a successful kidney transplant, these levels return to those of healthy adults, signifying the influence of kidney function on the elevation of serum URS levels. Potential therapies to reduce the production of URSs and thereby potentially reducing circulating levels and the associated adverse outcomes include dietary modifications and therapeutics that may modulate the gut microbiota.

CKD

The kidney is an essential organ that serves a multitude of functions, such as regulating water, salt and acid levels in the body, eliminating metabolic waste and producing hormones and prostaglandins. Due to its intricate structure, kidney diseases can be complex and are evaluated based on four units: glomerular, tubular, interstitial and vascular structures (Khan et al. 2013). CKD is a condition that results in irreversible renal dysfunction caused by damage to any of these components (Felizardo et al. 2016).

CKD significantly influences incidence and mortality from noncommunicable diseases. Between 7 and 12% of the world’s population has CKD, which has become a global medical issue (Klinkhammer & Boor 2023). In 2017, CKD caused an estimated 1.2 million deaths. By 2019, CKD had become the 18th leading cause of death and the 8th among individuals aged 50–74 (Vos et al. 2020). More than 2.5 million individuals are undergoing renal replacement therapy, with projections indicating that this figure will reach 5.4 million by 2030 (Liyanage et al. 2015), and it is projected that CKD will be the 5th leading cause of death by the year 2040 (Foreman et al. 2018). Regrettably, in various nations, access to renal replacement services is limited. This shortage of treatment options has resulted in an estimated 2.3–7.1 million adults dying prematurely (Liyanage et al. 2015).

The leading causes of CKD vary by region, with diabetes and hypertension being the most common causes (Couser et al. 2011). HIV may also contribute to the development of CKD in developing countries (Ekrikpo et al. 2018), as can exposure to toxins or heavy metals (Jha et al. 2013). The root cause is still unknown in some regions where CKD is highly prevalent (Correa-Rotter et al. 2014). On the other hand, age, gender, high blood pressure, ethnicity, socioeconomic factors and genetic factors are thought to be risk factors for CKD. Acute renal failure, glomerulonephritis, diabetes mellitus, genetic diseases, cardiovascular diseases (CVDs), multisystem diseases, drugs, urological conditions and infections are among the reported causes of CKD (Lucas & Taal 2023).

CVD is a major cause of mortality in people with CKD. For example, people with CKD appear at an increased risk of major adverse cardiovascular events, including atherothrombotic disease leading to myocardial infarction and stroke, heart failure, arrhythmias and sudden cardiac death in adults and children (Charytan 2018, Weaver & Mitsnefes 2018).

Along with diabetes mellitus and hypertension, elevated levels of URSs in the bloodstream have the potential to increase the risk of negative cardiovascular outcomes (Schiattarella et al. 2017). Three frequently researched URS metabolites are IS, PCS and TMAO (Vaziri et al. 2016, Jian et al. 2023).

The URS metabolites in question are a byproduct of the metabolic processes of L-carnitine and choline, resulting in the production of TMAO (Yu et al. 2019), tryptophan, which results in the production of IS (Imazu et al. 2017, Graboski et al. 2023), and tyrosine and phenylalanine, which results in the production of PCS (Di Paola et al. 2023). There are three steps involved in their synthesis: i) tryptophan, tyrosine, phenylalanine, L-carnitine and choline are obtained from the diet and encounter the intestinal microbiota; ii) the intestinal microbiota processes the nutrients to produce intermediate or final metabolites; and iii) metabolic intermediates undergo modification by the intestinal mucosa and/or gut microbiota before their transportation to the liver via the portal circulation. Once there, hepatic enzymes engage in further modification of these metabolites, resulting in the production of IS, PCS and TMAO, the final products (Daneshamouz et al. 2021, Di Paola et al. 2023).

Based on evidence from both animal and human studies, it is known that impaired elimination of uremic toxins from the intestinal microbiota exerts a detrimental effect on cardiovascular and renal tissues, primarily through the formation of fibrosis and impairment of endothelial function (Koppe et al. 2018). In non-CKD adults, the kidneys eliminate gut-derived URSs, causing a decrease in circulating levels. However, elevated levels of URSs in the bloodstream have been documented in individuals with renal failure. Upon successful kidney transplantation, URS concentrations return to levels similar to those found in non-CKD adult patients (Stubbs et al. 2016). However, it has also been observed that the levels of some uremic toxins increase again over time after renal transplantation (Liabeuf et al. 2018).

Microbiota

The term microbiota denotes the community of microorganisms present in a specific sample or location. While it broadly encompasses bacteria, viruses, fungi and protozoa, the term is most commonly used to refer to bacterial communities (Snelson et al. 2020). Nonetheless, it also includes commensal populations of archaea and protists (Matijašić et al. 2020). The basis of studies conducted in the field of microbiota in recent years is the determination of the factors affecting the microbiota in health or disease. When an organ precedes the term microbiota, it refers to the microbial community in that area like the ‘gut microbiota’ that we generally refer to in this study. A healthy microbiota needs to be diverse and in a dynamic balance. Unless the change caused by external factors is large enough, the microbiota tends to return to a stable state. However, severe and continuous change, along with the underlying genetic predisposition and medications used due to chronic diseases, cause an imbalance in the flora called dysbiosis. Many factors play a role in the emergence of dysbiosis. Inflammation and past infections, diet and xenobiotics, genetic structure, familial transmission, disruption of circadian rhythm, pregnancy, maternal malnutrition and physical damage are the leading factors among these factors.

The gut microbiota has recently become a subject of remarkable research due to its relationship with kidney diseases. The initial elucidation and definition of the ‘gut–kidney axis’ theory clarified the possible mechanisms by which gut microbiota-derived metabolites mediate this relationship and that changes in the composition or dysbiosis of the gut microbiota may influence these metabolites, thereby affecting the pathogenesis of CKD (Meijers & Evenepoel 2011, Wu et al. 2011). It is noteworthy that an increasing number of studies emphasize the critical role of the gut microbiota in the progression of CKD (Wu et al. 2011, Natarajan et al. 2014, Yang et al. 2018, Ren et al. 2020).

Uremic toxins, metabolic products of the intestinal microbiota associated with kidney disease, are often bound to proteins. This binding makes their filtration through the glomerular barrier more difficult. Accumulation of these urotoxins is a sign of decreased kidney function (Luo et al. 2022). The production of these metabolites is also the primary means by which the microbiota exerts its effects on the host. The effects of interventions on the microbiota are monitored by measuring these metabolites (Jin et al. 2019). As we explained earlier, frequently investigated metabolites are indoxyl sulfate, p-cresyl sulfate and trimethylamine-N-oxide (Vaziri et al. 2016, Jian et al. 2023). Comprehensive information regarding uremic toxins can be found in the relevant sections.

We performed a comprehensive literature review focusing on studies addressing microbiota and associated uremic toxins published between 2020 and 2024. This review identified pivotal research relevant to specific aspects of microbiota–toxin dynamics. The findings from these studies are systematically presented under the section titled ‘key findings from screening: microbiota–toxin interactions’ accompanied by detailed tables and explanatory notes to ensure clarity and comprehensiveness. Fungal and viral communities were not included in this study due to the primary focus on bacterial taxa. In addition, the current literature on the role of fungi and viruses in CKD-related microbiome alterations remains limited, requiring further comprehensive investigations.

URS

Individuals who suffer from CKD frequently expel harmful substances called uremic toxins through their urine. This can result in a range of health problems including renal fibrosis, anemia, bone diseases and CVDs. Further analysis of the pathophysiology of uremia has revealed that oxidative stress and inflammation are both factors that contribute to and are caused by this condition in CKD patients (Mutsaers et al. 2013, Hoyer & Nahrendorf 2019).

In cases of CKD, the body typically eliminates uremic toxins through urine to prevent the onset of various ailments such as anemia, renal fibrosis, bone diseases and CVDs. Upon closer inspection, it becomes apparent that uremia can both be a consequence and a catalyst of inflammation and oxidative stress in patients grappling with CKD (Zemaitis et al. 2024).

In recent years, there has been growing acceptance of the toxicity of uremic solutes. To date, over 150 molecules have been documented. IS, a highly researched protein-bound uremic toxin, is directly linked to cardiovascular events. Further research reveals that IS contributes to the advancement of renal fibrosis, bone deformities and neurological degradation (Hoyer & Nahrendorf 2019). In a study of TMAO, it has been shown that circulating TMAO levels increase due to CKD, which increases vascular oxidative stress and inflammation, contributing to endothelial dysfunction and CVD (Li et al. 2018).

The taxa associated with the increase or decrease of the relevant uremic toxins are indicated in the respective tables where applicable.

Indoxyl sulfate

Indoxyl sulfate (IS) is a harmful metabolite that can build up in the body as a result of the breakdown of dietary tryptophan. Specifically, the tryptophanase enzyme found in intestinal bacteria, such as Escherichia coli, converts tryptophan to indole, which then transforms in the liver to become IS. Once it has been absorbed by the intestines, the kidneys are responsible for removing it from the proximal tubules (Imazu et al. 2017).

IS is a metabolite that predominantly binds to serum albumin in the bloodstream, with over 90% of the compound being attached to this protein (Niwa & Shimizu 2012). Due to this robust binding, dialysis is not an effective means of removing the molecule from the body. Notably, as CKD progresses, the likelihood and severity of inflammation also increase (Niwa & Shimizu 2012, Lu et al. 2023).

IS is recognized to be efficient in the development of atherosclerosis and vascular inflammation by increasing oxidative stress in the endothelium. In individuals with CKD, IS levels increase inversely with glomerular filtration rate, and IS levels had an inverse association with renal function and a direct relationship with aortic calcification and mortality (Caldiroli et al. 2021, Wu et al. 2021).

Studies have shown that IS can have a notable impact on the emergence of CVD in patients with CKD, serving as a critical connection between these two conditions (Kaminski et al. 2019, Lin et al. 2020, Lu et al. 2023). In patients with stage 3–4 CKD, URSs, such as IS and PCS, have been found to elevate levels of inflammatory markers, such as interleukin-6 and glutathione peroxidase. In addition, these toxins have been linked to systemic inflammation and atherosclerosis (Lau et al. 2015). In addition, an old study discovered that IS can initiate inflammatory responses by inhibiting lymphoblast and interleukin-2 (Kawashima 1989).

A very important study on IS and its cardiovascular relationships was conducted in 2006 (Yamamoto et al. 2006). As a result of the study, it was determined that there was a significant correlation between IS and the development of atherosclerosis and vascular inflammation. Their findings showed that IS increased oxidative stress in the endothelium (Yamamoto et al. 2006). Later studies involving 139 patients in 2009 showed a direct relationship between IS levels and CKD as the glomerular filtration rate decreases. In this group, IS levels were also associated with aortic calcification and mortality (Barreto et al. 2009).

Exposure to IS has been observed to increase the expression and activation of ERK, P38 mitogen-activated protein kinases and NFkB, which have the potential to impact cardiac remodeling. In addition, renin receptors and angiotensin receptors are activated by IS, leading to harmful effects on cardiomyocytes, cardiac fibroblasts and cardiac endothelial cells. These negative effects ultimately contribute to cardiovascular dysfunction (Imazu et al. 2017).

During the initial phases of CKD, elevated IS levels have been associated with left ventricular dysfunction, coronary atherosclerosis, coronary stent restenosis and cardiac fatality (Ravid et al. 2021). Nonetheless, in advanced CKD, like that experienced by hemodialysis patients, the relationship between high IS levels and cardiovascular incidents, and cardiac death, is not straightforward. Studies discovered that heightened IS levels were not connected to cardiovascular morbidity and mortality, potentially due to end-organ damage already caused by advanced CKD and the reduced impact of uremic toxins (Ackley et al. 2017, Ravid et al. 2021, Li et al. 2022c , Takkavatakarn et al. 2022).

Numerous studies have shown that IS and PCS, which are protein-bound uremic toxins, can lead to the development of CKD by causing oxidative stress in renal tubular cells. Moreover, these toxins can also contribute to CVDs associated with CKD by inducing oxidative stress in endothelial cells and vascular smooth muscle cells. A vital study revealed that IS and PCS both significantly contribute to the advancement of CKD and its associated complications (Asai et al. 2018).

A study found that IS has pro-fibrotic, pro-hypertrophic and pro-inflammatory effects on NCF (neonatal rat cardiac fibroblasts), NCM (neonatal rat cardiac myocytes),and THP-1 (human monocytic cell line) cells. This suggests that reducing IS or its activated pathways could be a promising approach to treat CVDs associated with CKD (Lekawanvijit et al. 2009).

In a study, it was shown that IS levels were high for CKD and end-stage renal disease (ESRD), but decreased in the first phase of renal transplantation (1 month) and could remain at the current level without increasing again in the early transplant phase (within 12 months) (Liabeuf et al. 2018). In another study, it was reported that IS levels increased as a result of malnutrition in renal transplant patients and as a result, it was possible to increase renal dysfunction (Huang et al. 2012).

A review of more recent literature suggests that similar views are still valid that IS levels are associated with CKD progression (Cheng et al. 2020, Holle et al. 2020, Lu et al. 2021). Similarly, studies have been conducted to determine the relationship between IS and CVDs, with a particular focus on patients with CKD. In these studies, elevated levels of IS observed in patients with CKD have not been associated with heart failure (Iwasaki et al. 2024); however, they have been linked to vascular calcification (Gao et al. 2024) and atherosclerotic mechanisms (Wakamatsu et al. 2024).

P-cresyl sulfate

P-cresyl sulfate (PCS) is a tyrosine metabolite produced by the gut microbiota. It is processed in four steps: the first three of which are carried out in gut microbes to form intermediates, namely 4-hydroxyphenylpyruvate, 4-hydroxyphenylacetate and p-cresol. The final step is the formation of PCS in the gut mucosa or liver. PCS, like IS, induces NADPH oxidase activity and reactive oxygen species production, which contributes to direct cytotoxicity to cardiomyocytes, facilitating cardiac apoptosis and resulting in diastolic dysfunction (Zhao & Wang 2020).

PCS plays a significant role in CKD-related pathophysiology by activating protumorigenic processes such as chronic systemic inflammation, increased oxidative stress through free radical production and immune dysfunction (Simeoni et al. 2024a ). It is also known that circulating PCS levels are an independent marker for predicting the risk of CKD progression and various clinical events by exerting prooxidant and pro-inflammatory effects on complex organ systems (Chang et al. 2020).

A study investigated the potential links between blood antioxidant levels, oxidative stress, arterial thickness and various inflammatory parameters. The findings showed a correlation between serum-free and total IS and PCS levels and inflammatory markers, and a positive association was observed between PCS levels and IL-6 and arterial stiffness (Rossi et al. 2014).

The fermentation of the amino acid tyrosine forms p-cresol. This fermentation process is carried out by bacteria such as Bifidobacterium, Clostridium difficile, Faecalibacterium prausnitzii, Lactobacillus and Subdoligranulum. Then p-cresol reacts with sulfate in the liver and gets converted into PCS (Nallu et al. 2017).

Studies have indicated that PCS and IS have the potential to harm kidney cells by triggering specific pathophysiological alterations (Mutsaers et al. 2015). These alterations can arise through a range of mechanisms, including oxidative stress, cytokine and inflammatory gene expression, renal fibrosis, nephrosclerosis, differentiation of epithelial cells into mesenchymal cells, activation of the renin–angiotensin–aldosterone system (Delgado-Valero et al. 2021, Sapian et al. 2022, Zhang et al. 2023), methylation of the Klotho gene and the induction of intercellular adhesion molecule-1 (ICAM-1). These effects not only cause renal damage but also have cytotoxic effects that lead to cardiovascular and inflammatory changes. Several studies suggest that these toxins may serve as dependable biomarkers for predicting CKD-related cardiovascular and inflammatory consequences (Biagi et al. 2010, Cigarran Guldris et al. 2017, Delgado-Valero et al. 2021).

Recent studies also report that PCS, like IS, has a role in CKD (Corradi et al. 2024) and is also associated with CVD (Zwaenepoel et al. 2024). Studies also show that PCS correlates with diabetes (Oladi-Ghadikolaei et al. 2023). There are also recent studies reporting the association of IS and PCS with cancer progression (Simeoni et al. 2024a , b ).

Trimethylamine-N-oxide

Trimethylamine-N-oxide (TMAO) is a gut microbiota-dependent degradation product of dietary trimethylamine precursors (mainly choline and carnitine). Its metabolism consists of three steps: first, intestinal microbial degradation of dietary trimethylamine precursors; second, conversion of trimethylamine to TMAO by liver enzymes; and finally, elimination of TMAO by the kidney. Trimethylamine is oxidized in the liver by FMO (flavin-containing monooxygenases), especially FMO3, to produce TMAO (Yu et al. 2019).

The mechanisms by which TMAO may exacerbate kidney damage and worsen nephropathy are not fully understood. However, the potential of TMAO to aggravate kidney disease by activating the inflammatory response should be considered (Zixin et al. 2022). TMAO activates the NLRP3 inflammasome, leading to the release of IL-1β and IL-18, thereby accelerating renal inflammation (Fang et al. 2021). Beyond intensifying inflammation, TMAO is also known to contribute to oxidative stress in CKD patients (Ribeiro et al. 2024). High TMAO concentrations are known to induce vascular oxidative stress and inflammation, reducing NO production, which may trigger endothelial dysfunction and CKD-related complications such as CVD (Li et al. 2019).

The concentration of TMAO in human blood plasma is increased in patients with renal failure. It can range from 3 mmol/L in healthy individuals to 40 mmol/L in those with renal failure. However, information on the amount of TMAO found in other tissues is limited (Koeth et al. 2013). Research has indicated that individuals who suffer from advanced kidney disease frequently exhibit elevated TMA and TMAO levels within their system. This may be attributed to a decline in kidney function and a diminished capacity to eliminate TMAO from the bloodstream. Furthermore, kidney impairment stemming from inadequate blood flow to the kidneys could also play a role in heightening TMAO levels in both urine and plasma (Ufnal et al. 2014).

Recent research suggests that elevated levels of TMAO in the bloodstream may increase the likelihood of developing CVD, regardless of the presence of other known risk factors such as diabetes or renal failure (Zixin et al. 2022). However, it is worth noting that individuals with high TMAO levels exhibit additional risk factors such as hypertension, diabetes and impaired renal function (Gruppen et al. 2017).

In a study, TMAO levels were measured in the healthy control group, individuals with CKD in the 3rd, 4th and 5th stages of the disease, ESRD patients and transplantation patients. As a result of the study, it was observed that TMAO levels were normal in healthy individuals and increased as CKD stages progressed. The mean concentration in ESRD patients was reported to be approximately 30 times higher than the mean in the control group with normal renal function. In addition, TMAO concentrations were measured before and 3 months after the transplant in six individuals who had successful renal transplantation, and it was reported that a rapid decrease was encountered (Stubbs et al. 2016). Similarly, a different study observed a significant decline in TMAO levels following kidney transplantation, which remained stable for at least 2 years posttransplantation (Missailidis et al. 2016).

In a prospective study, the levels of various uremic toxins were measured in 51 kidney transplant recipients at four time points: at the time of transplantation and then on day 7, month 3 and month 12. The results showed that all analyzed uremic toxins dropped significantly within the first 7 days posttransplant. Among patients with CKD, serum TMAO levels showed a marked decrease by day 7 compared to healthy individuals. However, this decrease became much less significant by month 3 and beyond (Poesen et al. 2016).

Key findings from screening: microbiota–toxin interactions

This study is not a meta-analysis study, but is planned as a general evaluation of studies conducted in the period 2020–2024. This review was created by examining approximately 600 studies as a result of searching the terms ‘uremic toxins, TMAO, PCS, IS and microbiota’ in ScienceDirect and PubMed. Studies containing uremic toxins or their metabolites (such as IS and PCS or tryptophan and indole derivatives instead) were evaluated. The tables presented in this study were prepared based on the key bacterial taxa identified in the reviewed literature. The selection of these taxa was guided by their association with uremic toxins or related complications, as highlighted in the studies. Specifically, bacterial taxa demonstrating notable changes in composition were prioritized. These changes in microbiome density, as reported in the reviewed studies, formed the basis for the analysis. Taxonomic groups at the phylum, family and genus levels were considered. From each study, the 5–7 taxa exhibiting the most significant changes in microbiota composition were included, and the corresponding studies were referenced in the tables. Consequently, pie charts were generated to represent the taxa most frequently analyzed across the studies. Explanations for certain taxa have been provided within the tables.

Please refer to Table 1 for the phylum group, Table 2 for the family and Table 3 for the genus. The studies from which the relevant elements were taken are referenced in these tables. The graphs illustrate which bacterial taxonomic group is mentioned more frequently based on the numbers of these reference studies. The frequency of bacterial taxa, based on the reference counts from the compiled studies, is presented in pie charts. Fig. 1 illustrates the frequency of taxa from Table 1, Fig. 2 corresponds to Table 2, and Fig. 3 represents the data from Table 3.

Table 1

Bacterial groups that attracted attention on the basis of phylum in the studies carried out in 2020–2024.

Phylum Annotations References
Acidobacteria Associated with diabetic kidney disease (DKD), which is also related to TMAO Yang et al. (2023)
Actinobacteria One of the most abundant in the microbiota in CKD; potential roles in the increase of IS and PCS Wiese et al. (2024), Tran et al. (2023), Pan et al. (2020), Lin et al. (2022), Sun et al. (2023), Shi et al. (2022), Liu et al. (2022a), Haonon et al. (2022), Pang et al. (2024), Li et al. (2020), Lakshmanan et al. (2021), Yan et al. (2023), Qin et al. (2023), Moon et al. (2022), Mo et al. (2024), Zhang et al. (2024a), Jiang et al. (2024), Bian et al. (2024), Cui et al. (2024), Yang et al. (2023)
Archaeal phyla Decrease in TMAO production Mafra et al. (2022), Kemp et al. (2024)
Bacteroidetes Associated with increased levels of TMAO, PCS and IS Wiese et al. (2024), Shi et al. (2024), Biruete et al. (2021), Tran et al. (2023), Pan et al. (2020), Yi et al. (2023), Lin et al. (2022), Sun et al. (2023), Shi et al. (2022), Zhu et al. (2023), Liu et al. (2022a), Haonon et al. 2022, Pang et al. 2024, Das et al. (2022), Li et al. (2020), Lakshmanan et al. (2021), Yan et al. (2023), Du et al. (2020), Qin et al. (2023), Moon et al. (2022), Mo et al. (2024), Zhang et al. (2024a), Jiang et al. (2024), Zhang et al. (2024b), Cui et al. (2024), Yang et al. (2023)
Campilobacterota Yi et al. (2023)
Cyanobacteria Sun et al. (2023), Pang et al. (2024), Yang et al. (2023)
Desulfobacterota Yi et al. (2023), Lin et al. (2022), Mo et al. (2024)
Epsilonbactereota Yan et al. (2023), Huang et al. (2023)
Euryarchaeota Haonon et al. (2022)
Firmicutes (Bacillota) Firmicutes/bacteroidetes ratio is considered an important index of gut composition, and this ratio is altered (increased) in CKD; role in tryptophan metabolism (increase in IS); potential increases in other uremic toxins Wiese et al. (2024), Shi et al. (2024), Tran et al. (2023), Pan et al. (2020), Yi et al. (2023), Sun et al. (2023), Shi et al. (2022), Liu et al. (2022a), Haonon et al. 2022, Pang et al. 2024, Li et al. (2020), Lakshmanan et al. (2021), Yan et al. (2023), Du et al. (2020), Qin et al. (2023), Li et al. (2022b), Moon et al. (2022), Mo et al. (2024), Zhang et al. (2024a), Attaye et al. (2023), Huang et al. (2023), Jiang et al. (2024), Zhang et al. (2024b), Bian et al. (2024), Cui et al. (2024)
Fusobacteria Increase in CKD; potential role in PCS production Tran et al. (2023), Lin et al. (2022), Sun et al. (2023), Liu et al. (2022a), Li et al. (2020), Du et al. (2020)
Lentisphaerae Lakshmanan et al. (2021)
Oscillospirales McFarlane et al. (2022)
Patescibacteria Pang et al. (2024), Bian et al. (2024)
Proteobacteria (Pseudomonadota) Increase in CKD; potential roles in TMAO and IS production Wiese et al. (2024), Pan et al. (2020), Yi et al. (2023), Lin et al. (2022), Sun et al. (2023), Shi et al. (2022), Liu et al. (2022a), Haonon et al. (2022), Pang et al. (2024), Das et al. (2022), Li et al. (2020), Lakshmanan et al. (2021), Yan et al. (2023), Du et al. (2020), Qin et al. (2023), Moon et al. (2022), Zhang et al. (2024a), Huang et al. (2023), Zhang et al. (2024b), Bian et al. (2024), Cui et al. (2024)
Saccharibacteria Haonon et al. (2022), Lakshmanan et al. (2021)
Tenericutes Lakshmanan et al. (2021), Du et al. (2020), Zhang et al. (2024b)
Verrucomicrobia Increase in CKD (although some species show a decrease, generally an increase); potential roles in TMAO increase and CVS-related effects; increase in PCS production Wiese et al. (2024), Biruete et al. (2021), Tran et al. (2023), Shi et al. (2022), Haonon et al. (2022), Pang et al. (2024), Das et al. (2022), Lakshmanan et al. (2021), Li et al. (2022b), Moon et al. (2022), Mo et al. (2024), Zhang et al. (2024a), Huang et al. (2023), Zhang et al. (2024b), Yang et al. (2023)

DKD, diabetic kidney disease; CKD, chronic kidney disease.

Figure 1
Figure 1

Graph of the most mentioned bacteria by phylum in the studies conducted in 2020–2024.

Citation: Microbiota and Host 3, 1; 10.1530/MAH-24-0012

Table 2

Bacterial groups that attracted attention based on family in the studies carried out in 2020–2024.

Family Annotation References
Alcaligenaceae Pan et al. (2020)
Bacteroidaceae In healthy adults, approximately 20–80% of the gut microbiota is associated with the Bacteroidetes phylum, which includes the genera Parabacteroides, Bacteroides, Alistipes and Prevotella. In general, a decrease in CKD is noted; however, some studies report no difference or even an increase compared to healthy controls. Possible roles in uremic toxin production Asgharian et al. (2022), Liu et al. (2022b), Tran et al. (2023), Pan et al. (2020), Pivari et al. (2022)
Bifidobacteriaceae Asgharian et al. (2022)
Christensenellaceae Pivari et al. (2022)
Clostridiaceae Liu et al. (2022a), Bian et al. (2024)
Coriobacteriaceae Jiang et al. (2024)
Cyanobiaceae Hsu et al. (2021)
Enterobacteriaceae An increase in CKD supports PCS production. TMAO promotes the proliferation of this family Sun et al. (2023), Pivari et al. (2022)
Erysipelotrichaceae An increase in CKD is correlated with high TMAO levels. In some species, it is associated with increased IS and PCS levels Liu et al. (2022b), Pan et al. (2020), Sohn et al. (2024), Hsu et al. (2021), Bian et al. (2024)
Fusobacteraceae Tran et al. (2023), Sun et al. (2023)
Lachnospiraceae In CKD, some species show a decrease while others show an increase. Certain species are associated with uremic toxin production Liu et al. (2022b), Tran et al. (2023), Yan et al. (2023), Zhu et al. (2023), Huang et al. (2023), Jiang et al. (2024), Bian et al. (2024)
Lactobacillaceae Beneficial taxa show a decrease in CKD. Their increase in gut content offers promising potential for CKD improvement and the reduction of uremic toxins Asgharian et al. (2022), Pan et al. (2020), Liu et al. (2022a), Yan et al. (2023), Huang et al. (2023), Bian et al. (2024)
Methanosperaceae Cason et al. (2020)
Micrococcaceae Liu et al. (2022a)
Moraxellaceae Liu et al. (2022a)
Muribaculaceae Wu et al. (2024), Huang et al. (2023)
Oscillospiraceae Yan et al. (2023)
Peptostreptococcacea Tran et al. (2023), Bian et al. (2024)
Prevotellaceae The taxon potentially exerting a protective role in CKD demonstrates a negative correlation with CRP, suggesting that its reduction in CKD may contribute to increased inflammation. Furthermore, this taxon exhibits negative correlations with TMAO, IS and PCS levels Asgharian et al. (2022), Tran et al. (2023), Pan et al. (2020), Liu et al. (2022a), Pivari et al. (2022)
Rikenellaceae Liu et al. (2022b), Jiang et al. (2024)
Ruminococcaceae In patients with CKD, an increased abundance is observed, with involvement in the production of IS, PCS and TMAO. Liu et al. (2022b), Hsu et al. (2022), Tran et al. (2023), Pan et al. (2020), Sohn et al. (2024), Yan et al. (2023), Attaye et al. (2023)
Staphylococcaceae Yan et al. (2023)

CKD, chronic kidney disease.

Figure 2
Figure 2

Graph of the most mentioned bacteria by family in the studies conducted in 2020–2024.

Citation: Microbiota and Host 3, 1; 10.1530/MAH-24-0012

Table 3

Bacterial groups that attracted attention based on genus in the studies carried out in 2020–2024.

Genus Annotations References
Acetatifactor Zhang et al. (2024b)
Agathobacter Mitrović et al. (2023)
Akkermansia In some species, CKD is associated with an increase, while in others, a decrease, with potential roles in the production of uremic toxins Biruete et al. (2021), Hsu et al. (2022), Shi et al. (2022), Hsu et al. (2021), Li et al. (2022a), Mo et al. (2024), Wu et al. (2024), Rashidi et al. (2021), Zhang et al. (2024b), Cui et al. (2024)
Alistipes As CKD stages progress, gut abundance increases, correlating with TMAO production and potential roles in the synthesis of PCS and IS precursors Yi et al. (2023), Pang et al. (2024), Das et al. (2022), Zhang et al. (2024a), Attaye et al. (2023), Zhu et al. (2023), Huang et al. (2023)
Allobaculum Shi et al. (2024), Liu et al. (2022b)
Alloprevotella Zhu et al. (2023), Wu et al. (2024)
Anaerosporobacter Tran et al. (2023)
Anaerostipes Sohn et al. (2024)
Bacteroides One of the prominent genera abundant in the gut composition of CKD patients, with strong potential roles in TMAO, PCS and IS production Biruete et al. (2021), Liu et al. (2022b), Yi et al. (2023), Lin et al. (2022), Sun et al. (2023), Zhu et al. (2023), Pang et al. (2024), Ebrahim et al. (2022), Pivari et al. (2022), Das et al. (2022), Tran et al. (2023), Li et al. (2020), Zhang et al. (2024a), Attaye et al. (2023), Wu et al. (2024), Rashidi et al. (2021), Jiang et al. (2024), Zhang et al. (2024b), Cui et al. (2024)
Barnesiella Zhang et al. (2024b)
Bifidobacteria Decreased in CKD, showing a negative correlation with PCS production. Potential role in reducing TMAO levels Stuivenberg et al. (2022), Sohn et al. (2024), Lee et al. (2020), Shi et al. (2022), Liu et al. (2022a), McFarlane et al. (2021), Das et al. (2022), Mitrović et al. (2023), Zhu et al. (2023), Jiang et al. (2024), Cui et al. (2024)
Blautia The role of Blautia in CKD is complex, exhibiting both protective and pathogenic potential Yi et al. (2023), Lin et al. (2022), McFarlane et al. (2021), Ebrahim et al. (2022), Li et al. (2020), Mitrović et al. (2023), Attaye et al. (2023), Cui et al. (2024), Pan et al. (2020)
Butyricicoccus Das et al. (2022)
Butyricimonas Das et al. (2022), Mo et al. (2024)
Catenibacterium Tran et al. (2023)
Clostridium Pan et al. (2023), Pang et al. (2024), Zhu et al. (2023), Zhang et al. (2024b), Bian et al. (2024)
Colidextribacter Jiang et al. (2024)
Corynebacterium Pan et al. (2020)
Coprococcus Mo et al. (2024)
Desulfovibrio The relationship between the Desulfovibrio genus and eGFR varies significantly across studies; however, the prevailing consensus suggests that Desulfovibrio may metabolize renoprotective short-chain fatty acids Kang et al. (2020), Shi et al. (2022), Das et al. (2022), Wu et al. (2024), Cason et al. (2020), Huang et al. (2023), Jiang et al. (2024), Zhang et al. (2024b)
Dorea Mitrović et al. (2023), Attaye et al. (2023)
Eisenbergiella Zhang et al. (2024b)
Enterococcus Das et al. (2022), Rashidi et al. (2021)
Escherichia, Shigella Lin et al. (2022), Pivari et al. (2022), Li et al. (2020), Wu et al. (2024), Cui et al. (2024)
Faecalibacterium Although some studies report the opposite, the general consensus is that gut microbial abundance is reduced in CKD patients. This reduction is positively correlated with systemic inflammation. Negatively correlated with IS and PCS Yi et al. (2023), Lin et al. (2022), Sun et al. (2023), Liu et al. (2022a), Ebrahim et al. (2022), Tran et al. (2023), Li et al. (2020), Du et al. (2020), Cui et al. (2024)
Flavobacterium Pan et al. (2020)
Flavonifactor (Eubacterium) Pang et al. (2024), Das et al. (2022), Mitrović et al. (2023), Mo et al. (2024), Zhang et al. (2024a), Attaye et al. (2023), Wu et al. (2024)
Fusobacterium Zhu et al. (2023)
Fusicatenibacter Tran et al. (2023), Mitrović et al. (2023)
Gordonibacter Hsu et al. (2022)
Helicobacter Wu et al. (2024), Huang et al. (2023), Jiang et al. (2024)
Intestinibacter Sohn et al. (2024)
Klebsiella Sun et al. (2023)
Lachnospira A decrease in gut abundance is associated with various diseases, including CKD. Negatively correlated with TMAO plasma levels Liu et al. (2022b), Hsu et al. (2022), Lin et al. (2022), Sohn et al. (2024), Zhu et al. (2023), Hsu et al. (2021), Pang et al. (2024), Pivari et al. (2022)
Lactobacillus Lactobacillus species and other acid-producing bacteria may suppress aerobic bacteria, restore intestinal microbiota balance and reduce uremic toxins in CKD patients Stuivenberg et al. (2022), Moon et al. (2022), Yi et al. (2023), Lee et al. (2020), Shi et al. (2022), Pang et al. (2024), McFarlane et al. (2021), Mitrović et al. (2023), Wu et al. (2024), Rashidi et al. (2021), Jiang et al. (2024), Zhang et al. (2024b), Bian et al. (2024)
Lactococcus Jiang et al. (2024)
Marvinbryantia Shi et al. (2024)
Megamonas Lin et al. (2022), Sun et al. (2023)
Moryella Sohn et al. (2024)
Mucispirillum Mo et al. (2024)
Myroides Pan et al. (2020)
Negative bacillus Sohn et al. (2024)
Oligella Pan et al. (2020)
Oscillibacter Attaye et al. (2023)
Oscillospira Shi et al. (2022)
Parabacteroides Yi et al. (2023), Sun et al. (2023), Mo et al. (2024), Rashidi et al. (2021)
Peptoclostridium Tran et al. (2023)
Peptococcus Cason et al. (2020)
Pirellula Wang et al. (2023)
Prevotella In CKD, gut microbial abundance decreases overall, yet some species within certain taxa exhibit a paradoxical positive correlation with TMAO production, likely due to metabolic adaptations to the dysbiotic environment McFarlane et al. (2022), Lin et al. (2022), Ebrahim et al. (2022), Du et al. (2020), Zhang et al. (2024a), Cason et al. (2020)
Pseudomonas Lin et al. (2022)
Rikenella Liu et al. (2022b)
Rombutsia Hsu et al. (2022), Yi et al. (2023), Bian et al. (2024)
Roseburia Hsu et al. (2022), Ebrahim et al. (2022), Das et al. (2022), Attaye et al. (2023), Pan et al. (2020)
Ruminococcus In CKD, certain microbial species exhibit increased abundance, while others show a decrease. A positive correlation is observed in choline synthesis (see TMAO) Liu et al. (2022b), Yi et al. (2023), Lin et al. (2022), Liu et al. (2022a), Hsu et al. (2021), Ebrahim et al. (2022), Li et al. (2020), Mitrović et al. (2023), Zhang et al. (2024a), Wang et al. (2023)
Sarcina Tran et al. (2023)
Solobacterium Wang et al. (2023)
Stenotrophomonas Wang et al. (2023)
Streptococcus In CKD, significant increases and risk factors for infection are observed with TMA production (see TMAO) McFarlane et al. (2021), Mitrović et al. (2023), Du et al. (2020), Rashidi et al. (2021), Cui et al. (2024)
Subdoligranulum Mitrović et al. (2023), Pivari et al. (2022)
Syntrophococcus Wang et al. (2023)
Terrisporobacter Sohn et al. (2024)
Turicibacter Sohn et al. (2024), Hsu et al. (2021), Bian et al. (2024)

CKD, chronic kidney disease.

Figure 3
Figure 3

Graph of the most mentioned bacteria by genus in the studies conducted in 2020–2024.

Citation: Microbiota and Host 3, 1; 10.1530/MAH-24-0012

It is known that metabolic disorders, including CKD, affect the intestinal flora, thereby increasing intestinal permeability and causing dysbiosis, leading to systemic inflammatory conditions (Anders et al. 2013). Although research in this area does not have a very old history, it is possible to say that studies investigating the relationship between intestinal microbial composition and CKD have been carried out for a significant period of time.

Despite the existence of numerous studies encompassing a vast array of data, a single study has indicated that the intestinal density of Proteobacteria, Enterobacteria, Escherichia coli, Acinetobacter, Proteus species and Clostridium is elevated in CKD patients relative to healthy controls. Conversely, the intestinal density of Lactobacillus and Bifidobacterium species has been demonstrated to be diminished in CKD patients (Guldris et al. 2017).

A study revealed that CKD patients have a decrease in the abundance of Lactobacillaceae and Prevotellaceae, while the levels of Enterobacter and Enterococcus are significantly higher (Vaziri et al. 2013). Another study found that the intestinal microbiota in end-stage renal disease patients is associated with inflammation and renal function, with a decrease in the proportion of bacteria ranging from Prevotella to Bacteroides (Jiang et al. 2017). Both dialysis and non-dialysis CKD patients have been found to have alterations in their gut bacteria. Peritoneal dialysis patients show a decrease in the abundance of Firmicutes (Bacillota) and Actinobacteria, while hemodialysis patients show an increase in the abundance of Bacteroides (McIntyre et al. 2011).

These changes in the microbiota appear to be closely related to the presence of uremic toxins in CKD patients (Pluznick 2016). Animal models of CKDs have shown that excessive uremia can lead to dysbiosis in the gut and dysfunction of the intestinal barrier and bacterial translocation (Andersen et al. 2017). A recent study was conducted to analyze the microbial diversity present in the intestinal flora. In this study, a reduction in the diversity of the intestinal flora and alterations in the communities were observed in peritoneal dialysis patients, in whom microbial diversity was found to be positively correlated with albumin level. A total of 20 gut flora phyla were identified in 166 fecal samples, which were divided into three dominant gut types: Bacteroides, Firmicutes (Bacillota) and Proteobacteria. Further analysis revealed the presence of 198 genera, of which 86 exhibited a significant difference in abundance. At the genus level, Faecalibacterium and butyrate-producing taxa, namely Bifidobacteriaceae and Prevotellaceae, were observed to be the dominant genera in the control, CKD and hemodialysis groups, although not necessarily respectively. Conversely, the dominant genera in the peritoneal dialysis patients group were those containing urease, forming indole and p-cresol, namely Escherichia, at the genus level and Enterobacteriaceae and Enterococcaceae at the family level (Hu et al. 2020).

In patients with normal gut health, many bacterial genera are responsible for the production of uremic toxins. IS is typically derived from the precursor indole, which is extracted from tryptophan by tryptophanase. This enzyme is found in the genera Citrobacter, Proteus and Escherichia. The liver enzyme cytochrome P450-2E1 (CYP2E1) is responsible for the absorption and processing of indole into indoxyl. Subsequently, indoxyl is sulfated by sulfotransferase (SULT), resulting in the formation of IS as the reaction product. Several studies demonstrated that an increase in dietary tryptophan results in elevated IS production (Banoglu et al. 2001, Lauriola et al. 2023).

It is established that PCS is the result of a conjugation reaction between sulfate and p-cresol in the intestinal wall (Vandaele 2023). P-cresol is a phenol that is formed from tyrosine and phenylalanine by anaerobic commensals, including Bacteroides, Lactobacillus, Clostridium and Bifidobacterium (Lauriola et al. 2023).

TMAO is one of the uremic toxins of intestinal origin. A study reported that TMAO levels were associated with inflammatory markers. An increased Firmicutes (Bacillota)/Bacteroidota ratio resulting from increased TMAO concentrations, increased Streptococcus, Pseudomonas and unclassified Clostridia UCG 014 density, and a decreased Rothia and RB41 density were reported to be effective in this relationship (Xie et al. 2024). The use of a dietary supplement formulated with the bacterial species Lactobacillus plantarum and Bifidobacterium lactis has been shown to significantly reduce TMAO levels in the body. Given the established role of elevated TMAO concentrations as a risk factor for CVDs, this reduction may contribute to a cardioprotective effect (Qiu et al. 2017, 2018).

It has been previously proposed that TMAO is produced from TMA, which is generated within the intestine. A review of the literature reveals that numerous bacterial species are involved in TMA production, including Clostridium difficile, Proteus species and Shigella species (Rath et al. 2017). Further research has indicated that several bacterial genera, including Clostridium, Lachnospira, Ruminococcus, Desulfovibrio and Candidatus Arthromitus, have the potential to influence circulating TMAO levels (Zhu et al. 2016, Fu et al. 2020). It can be argued that there are significant differences in the composition of the gut microbiota between individuals with low and high levels of TMAO production. Studies have also identified the gut bacteria Emergencia timonensis and Ihubacter massiliensis as key species responsible for the conversion of carnitine to TMAO (Wu et al. 2019).

A substantial body of research exists examining the correlation between specific species and uremic toxins. This expansive framework, however, is beyond the scope of the present study and merits a dedicated review in its own right. In this investigation, we have compiled a list of species that have been administered as supplements with the objective of influencing the composition of the microbiota, or the relationship between these species and the microbiota, and have presented this information in a table with references (see Table 4).

Table 4

The list of significant bacterial species proposed for uremic toxins and microbiota composition.

Species Annotations References
Akkermansia muciniphila Improves renal function, restores the gut microbiome, enhances the intestinal mucosal barrier, reduces systemic inflammation and mitigates interstitial fibrosis in rats with CKD. It may reduce TMAO production Xu et al. (2024), Pei et al. (2023)
Bifidobacterium longum A beneficial bacterium in CKD that reduces TMAO production De Mauri et al. (2020), Lúcio et al. (2024), Wang et al. (2022)
Bifidobacterium animals subs lactis It may reduce PCS production Zhu et al. (2023)
Bifidobacterium lactis A beneficial bacterium in CKD known for reducing inflammation and uremic toxins, particularly IS. Mitrović et al. (2023)
Bacteroides pleibius Potential roles in IS and PCS production Lin et al. (2022)
Bacteroides ovatus
Bacteroides fragilis
Bacteroides caccae
Clostridium coccoides Positively correlated with PCS production Miyata et al. (2021)
Clostridium leptum
Clostridium perfringens Potential roles in PCS production Pan et al. (2023), Lee et al. (2020)
Clostridium sporogenes Pan et al. (2023), Zhu et al. (2023)
Enterococcus faecalis Improved renal failure symptoms, including the accumulation of uremic toxins Takemura et al. (2024)
E. coli An increase in uremic toxin production and a decrease in the abundance of beneficial bacteria contribute to the worsening of CKD. Lin et al. (2022), Lee et al. (2020)
Faecalibacterium prausnitzii Potential role in reducing uremic toxin production in CKD recovery Li et al. (2022a)
Fusobacterium nucleatum Inhibitory effect on PCS production Zhu et al. (2023)
Haemophylus parainfluenza Associated with elevated levels of IS Stanford et al. (2021)
Lactobacillus casei In CKD, a known species that reduces inflammation and uremic toxins, particularly indoxyl sulfate (IS) Mitrović et al. (2023)
Lactobacillus acidophilus In CKD, a known species that reduces inflammation and uremic toxins, particularly indoxyl sulfate (IS) Mitrović et al. (2023), Park et al. (2024)
Lactobacillus reuteri Potential role in the reduction of uremic toxins De Mauri et al. (2020)
Lactobacillus plantarum One of the species important in CKD recovery Huang et al. (2021), Nissen et al. (2024), Pan et al. (2023)
Lactobacillus paracasei Another important species in CKD recovery Huang et al. (2021)
Lactobacillus plantarum subs plantarum A reduction in renal damage observed in CKD, improvement in BUN levels and a potential decrease in IS production Lee et al. (2020)
Lactobacillus paracasei subs paracasei
Lactobacillus pentosus Potential important roles in the reduction of IS. Lim et al. (2021)
Lactobacillus lactis subs lactis Lim et al. (2021)
Lactobacillus salivarius Lim et al. (2021)
Lactiplantibacillus paraplantarum Decreased serum phosphate levels and reduced blood indoxyl sulfate concentration Moon et al. (2022)
Streptococcus salivarius subs thermophilus A reduction in renal damage observed in CKD, improvement in BUN levels and a potential decrease in IS production Lee et al. (2020)

CKD,chronic kidney disease.

Conclusion

A plethora of studies have addressed the relationship between microbiota and CKD from disparate angles, yielding a multitude of results. It appears to be a formidable challenge to consolidate these findings into a unified framework. The objective of this study was to establish a comprehensive framework for the consideration of taxonomic changes. In addition, we have provided a separate table with references for a limited number of species-based studies, which illustrate specific examples from the broader context.

Uremic toxins have been demonstrated to play a pivotal role in the pathogenesis of CKD and in the development of numerous other diseases. Accordingly, our study was designed within the context of uremic toxins and microbiota, with an expanded framework encompassing uremic toxins and CKD. The review revealed a need for more specialized studies to clarify the data on uremic toxins, related metabolites, microbiota and related diseases. It is anticipated that future studies targeting the composition of the microbiota will yield promising results for many diseases, including CKD. Given the importance of disease pathways in determining therapeutic targets, it can be said that microbiota and microbiota-derived metabolic products represent an important field of study that requires further investigation.

Declaration of interest

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of this work.

Funding

This work did not receive any specific grant from any funding agency in the public, commercial or not-for-profit sector.

Author contribution statement

ZR took the lead in conceptualizing the review topic and formulating the study idea. She conducted a significant portion of the literature review, analyzed the findings, synthesized the results and drafted the manuscript. In addition, ZR coordinated the writing process and ensured the logical flow and coherence of the text. GAP contributed to the literature search, assisted in data collection and analysis and collaborated on drafting specific manuscript sections. GAP also reviewed and refined the written content to ensure accuracy and clarity. IMV was involved in performing the final checks on the manuscript, ensuring its alignment with the journal’s guidelines, and preparing it for submission. IMV also provided critical feedback throughout the process to enhance the overall quality and impact of the review.

References

  • Ackley W , Soiefer L , Etinger A , et al. 2017 Uremic retention solutes. In Aspects In Dialysis, pp 927. London: IntechOpen.

  • Anders H-J , Andersen K & Stecher B 2013 The intestinal microbiota, a leaky gut, and abnormal immunity in kidney disease. Kidney Int 83 10101016. (https://doi.org/10.1038/ki.2012.440)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Andersen K , Kesper MS , Marschner JA , et al. 2017 Intestinal dysbiosis, barrier dysfunction, and bacterial translocation account for CKD–related systemic inflammation. J Am Soc Nephrol 28 7683. (https://doi.org/10.1681/asn.2015111285)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Asai H , Hirata J & Watanabe-Akanuma M 2018 Indoxyl glucuronide, a protein-bound uremic toxin, inhibits hypoxia-inducible factor–dependent erythropoietin expression through activation of aryl hydrocarbon receptor. Biochem Biophys Res Commun 504 538544. (https://doi.org/10.1016/j.bbrc.2018.09.018)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Asgharian M , Gholizadeh P , Samadi Kafil H , et al. 2022 Correlation of inflammatory biomarkers with the diversity of Bacteroidaceae, Bifidobacteriaceae, Prevotellaceae and Lactobacillaceae families in the intestinal microbiota of patients with end stage renal disease. Adv Med Sci 67 304310. (https://doi.org/10.1016/j.advms.2022.07.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Attaye I , Lassen PB , Adriouch S , et al. 2023 Protein supplementation changes gut microbial diversity and derived metabolites in subjects with type 2 diabetes. iScience 26 107471. (https://doi.org/10.1016/j.isci.2023.107471)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Banoglu E , Jha GG & King RS 2001 Hepatic microsomal metabolism of indole to indoxyl, a precursor of indoxyl sulfate. Eur J Drug Metab Pharmacokinet 26 235240. (https://doi.org/10.1007/bf03226377)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barreto FC , Barreto DV , Liabeuf S , et al. 2009 Serum indoxyl sulfate is associated with vascular disease and mortality in chronic kidney disease patients. Clin J Am Soc Nephrol 4 15511558. (https://doi.org/10.2215/cjn.03980609)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Biagi E , Nylund L , Candela M , et al. 2010 Through ageing, and beyond: gut microbiota and inflammatory status in seniors and centenarians. PLoS One 5 e10667. (https://doi.org/10.1371/journal.pone.0010667)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bian M , Zhu C , Nie A , et al. 2024 Guizhi Shaoyao Zhimu Decoction ameliorates gouty arthritis in rats via altering gut microbiota and improving metabolic profile. Phytomedicine 131 155800. (https://doi.org/10.1016/j.phymed.2024.155800)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Biruete A , Cross T-WL , Allen JM , et al. 2021 Effect of dietary inulin supplementation on the gut microbiota composition and derived metabolites of individuals undergoing hemodialysis: a pilot study. J Ren Nutr 31 512522. (https://doi.org/10.1053/j.jrn.2020.10.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Caldiroli L , Armelloni S , Eskander A , et al. 2021 Association between the uremic toxins indoxyl-sulfate and p-cresyl-sulfate with sarcopenia and malnutrition in elderly patients with advanced chronic kidney disease. Exp Gerontol 147 111266. (https://doi.org/10.1016/j.exger.2021.111266)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cason CA , Kuntz TM , Chen EB , et al. 2020 Microbiota composition modulates inflammation and neointimal hyperplasia after arterial angioplasty. J Vasc Surg 71 13781389.e3. (https://doi.org/10.1016/j.jvs.2019.06.208)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chang J-F , Hsieh C-Y , Liou J-C , et al. 2020 Scavenging intracellular ROS attenuates p-cresyl sulfate-triggered osteogenesis through MAPK signaling pathway and NF-κB activation in human arterial smooth muscle cells. Toxins 12 472. (https://doi.org/10.3390/toxins12080472)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Charytan DM 2018 Introduction: cardiovascular disease in chronic kidney disease. Semin Nephrol 38 541. (https://doi.org/10.1016/j.semnephrol.2018.08.008)

  • Cheng T-H , Ma M-C , Liao M-T , et al. 2020 Indoxyl sulfate, a tubular toxin, contributes to the development of chronic kidney disease. Toxins 12 684. (https://doi.org/10.3390/toxins12110684)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cigarran Guldris S , González Parra E & Cases Amenós A 2017 Gut microbiota in chronic kidney disease. Revista de la Sociedad Española de Nefrología 37 919. (https://doi.org/10.1016/j.nefro.2016.05.008)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Corradi V , Caprara C , Barzon E , et al. 2024 A possible role of P-cresyl sulfate and indoxyl sulfate as biomarkers in the prediction of renal function according to the GFR (G) categories. Integr Med Nephrol Androl 11 e24e00002. (https://doi.org/10.1097/imna-d-24-00002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Correa-Rotter R , Wesseling C & Johnson RJ 2014 CKD of unknown origin in Central America: the case for a Mesoamerican nephropathy. Am J Kidney Dis 63 506520. (https://doi.org/10.1053/j.ajkd.2013.10.062)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Couser WG , Remuzzi G , Mendis S , et al. 2011 The contribution of chronic kidney disease to the global burden of major noncommunicable diseases. Kidney Int 80 12581270. (https://doi.org/10.1038/ki.2011.368)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cui Y , Kuang D , Wang J , et al. 2024 Effect of soluble dietary fiber on gut microbiota and derived metabolites in stage 3 to 5 chronic kidney disease patients: a randomized controlled trial. J Funct Foods 116 106181. (https://doi.org/10.1016/j.jff.2024.106181)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Daneshamouz S , Eduok U , Abdelrasoul A , et al. 2021 Protein-bound uremic toxins (PBUTs) in chronic kidney disease (CKD) patients: production pathway, challenges and recent advances in renal PBUTs clearance. NanoImpact 21 100299. (https://doi.org/10.1016/j.impact.2021.100299)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Das S , Song Z , Han H , et al. 2022 Intestinal osteopontin protects from alcohol-induced liver injury by preserving the gut microbiome and the intestinal barrier function. CMGH 14 813839. (https://doi.org/10.1016/j.jcmgh.2022.06.012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Delgado-Valero B , Cachofeiro V & Martínez-Martínez E 2021 Fibrosis, the bad actor in cardiorenal syndromes: mechanisms involved. Cells 10 1824. (https://doi.org/10.3390/cells10071824)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Du Z , Wang J , Lu Y , et al. 2020 The cardiac protection of Baoyuan decoction via gut-heart axis metabolic pathway. Phytomedicine 79 153322. (https://doi.org/10.1016/j.phymed.2020.153322)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ebrahim Z , Proost S , Tito RY , et al. 2022 The effect of ß-glucan prebiotic on kidney function, uremic toxins and gut microbiome in stage 3 to 5 chronic kidney disease (CKD) predialysis participants: a randomized controlled trial. Nutrients 14 805. (https://doi.org/10.3390/nu14040805)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ekrikpo UE , Kengne AP , Bello AK , et al. 2018 Chronic kidney disease in the global adult HIV-infected population: a systematic review and meta-analysis. PLoS One 13 e0195443. (https://doi.org/10.1371/journal.pone.0195443)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fang Q , Zheng B , Liu N , et al. 2021 Trimethylamine N-oxide exacerbates renal inflammation and fibrosis in rats with diabetic kidney disease. Front Physiol 12 682482. (https://doi.org/10.3389/fphys.2021.682482)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Felizardo RJF , Castoldi A , Andrade-Oliveira V , et al. 2016 The microbiota and chronic kidney diseases: a double-edged sword. Clin Translational Immunol 5 e86. (https://doi.org/10.1038/cti.2016.36)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Foreman KJ , Marquez N , Dolgert A , et al. 2018 Forecasting life expectancy, years of life lost, and all-cause and cause-specific mortality for 250 causes of death: reference and alternative scenarios for 2016–40 for 195 countries and territories. Lancet 392 20522090. (https://doi.org/10.1016/s0140-6736(18)31694-5)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fu BC , Hullar MAJ , Randolph TW , et al. 2020 Associations of plasma trimethylamine N-oxide, choline, carnitine, and betaine with inflammatory and cardiometabolic risk biomarkers and the fecal microbiome in the Multiethnic Cohort Adiposity Phenotype Study. Am J Clin Nutr 111 12261234. (https://doi.org/10.1093/ajcn/nqaa015)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gao Y , Zhao C-J , Liu Q , et al. 2024 Relationship between serum indoxyl sulfate and Klotho protein and vascular calcification in patients with chronic kidney disease stages 3–5. Int J Endocrinol 2024 8229604. (https://doi.org/10.1155/2024/8229604)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Graboski AL , Kowalewski ME , Simpson JB , et al. 2023 Mechanism-based inhibition of gut microbial tryptophanases reduces serum indoxyl sulfate. Cell Chem Biol 30 14021413.e7. (https://doi.org/10.1016/j.chembiol.2023.07.015)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gruppen EG , Garcia E , Connelly MA , et al. 2017 TMAO is associated with mortality: impact of modestly impaired renal function. Sci Rep 7 13781. (https://doi.org/10.1038/s41598-017-13739-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guldris SC , Parra EG & Amenós AC 2017 Gut microbiota in chronic kidney disease. Nefrologia 37 919. (https://doi.org/10.1016/j.nefroe.2017.01.017)

  • Haonon O , Liu Z , Dangtakot R , et al. 2022 Opisthorchis viverrini infection induces metabolic disturbances in hamsters fed with high fat/high fructose diets: implications for liver and kidney pathologies. J Nutr Biochem 107 109053. (https://doi.org/10.1016/j.jnutbio.2022.109053)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Holle J , Kirchner M , Okun J , et al. 2020 Serum indoxyl sulfate concentrations associate with progression of chronic kidney disease in children. PLoS One 15 e0240446. (https://doi.org/10.1371/journal.pone.0240446)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hoyer FF & Nahrendorf M 2019 Uremic toxins activate macrophages. Circulation 139 97100. (https://doi.org/10.1161/circulationaha.118.037308)

  • Hsu CN , Hou CY , Chang-Chien GP , et al. 2021 Maternal resveratrol therapy protected adult rat offspring against hypertension programmed by combined exposures to asymmetric dimethylarginine and trimethylamine-N-oxide. J Nutr Biochem 93 108630. (https://doi.org/10.1016/j.jnutbio.2021.108630)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hsu CN , Yu HR , Lin IC , et al. 2022 Sodium butyrate modulates blood pressure and gut microbiota in maternal tryptophan-free diet-induced hypertension rat offspring. J Nutr Biochem 108 109090. (https://doi.org/10.1016/j.jnutbio.2022.109090)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hu J , Zhong X , Yan J , et al. 2020 High-throughput sequencing analysis of intestinal flora changes in ESRD and CKD patients. BMC Nephrol 21 12. (https://doi.org/10.1186/s12882-019-1668-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang H , Li K , Lee Y , et al. 2021 Preventive effects of Lactobacillus mixture against chronic kidney disease progression through enhancement of beneficial bacteria and downregulation of gut-derived uremic toxins. J Agric Food Chem 69 73537366. (https://doi.org/10.1021/acs.jafc.1c01547)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang S-T , Shu K-H , Cheng C-H , et al. 2012 Serum total p-cresol and indoxyl sulfate correlated with stage of chronic kidney disease in renal transplant recipients. In Transplantation Proceedings, vol 44, 3rd ed. pp 621624. New York: Elsevier.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang Z , Fang Z , Wu A , et al. 2023 Fermented Psidium guajava leaves regulate the gut microbiota and improve metabolic alterations in diabetic mice. Food Biosci 51 102201. (https://doi.org/10.1016/j.fbio.2022.102201)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Imazu M , Takahama H , Shindo K , et al. 2017 A pathophysiological role of plasma indoxyl sulfate in patients with heart failure. Int J Gerontol 11 6266. (https://doi.org/10.1016/j.ijge.2016.05.010)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Iwasaki K , Miyoshi T , Urabe C , et al. 2024 Impact of serum indoxyl sulfate on one-year adverse events in chronic kidney disease patients with heart failure. J Clin Med 13 4384. (https://doi.org/10.3390/jcm13154384)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jha V , Garcia-Garcia G , Iseki K , et al. 2013 Chronic kidney disease: global dimension and perspectives. Lancet 382 260272. (https://doi.org/10.1016/s0140-6736(13)60687-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jian S , Yang K , Zhang L , et al. 2023 The modulation effects of plant-derived bioactive ingredients on chronic kidney disease: focus on the gut–kidney axis. Food Front 4 262282. (https://doi.org/10.1002/fft2.209)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jiang S , Xie S , Lv D , et al. 2017 Alteration of the gut microbiota in Chinese population with chronic kidney disease. Sci Rep 7 2870. (https://doi.org/10.1038/s41598-017-02989-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jiang S , Wei X , Wang H , et al. 2024 Biotransformed bear bile powder alleviates diet-induced nonalcoholic fatty liver disease in mice by regulating the gut microbiota and reversing lipid metabolism. Arab J Chem 17 105578. (https://doi.org/10.1016/j.arabjc.2023.105578)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jin Q , Black A , Kales SN , et al. 2019 Metabolomics and microbiomes as potential tools to evaluate the effects of the Mediterranean diet. Nutrients 11 207. (https://doi.org/10.3390/nu11010207)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kaminski TW , Pawlak K , Karbowska M , et al. 2019 The impact of antihypertensive pharmacotherapy on interplay between protein-bound uremic toxin (indoxyl sulfate) and markers of inflammation in patients with chronic kidney disease. Int Urol Nephrol 51 491502. (https://doi.org/10.1007/s11255-018-02064-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kang D-W , Adams JB , Vargason T , et al. 2020 Distinct fecal and plasma metabolites in children with autism spectrum disorders and their modulation after microbiota transfer therapy. mSphere 5 e00314-20. (https://doi.org/10.1128/msphere.00314-20)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kawashima Y 1989 Study on the uremic protein binding inhibitors as uremic toxin: toxic effect on erythroid colony formation, lymphocyte blast formation and renal function. The Japanese Journal of Nephrology 31 11511161. (https://doi.org/10.14842/jpnjnephrol1959.31.1151)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kemp JA , Schultz J , Modolon F , et al. 2024 Is there a correlation between TMAO plasma levels and archaea in the gut of patients undergoing hemodialysis? Int Urol Nephrol 17. (https://doi.org/10.1007/s11255-024-04273-5)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Khan KNM , Hard GC & Alden CL 2013 Chapter 47 – kidney. In Haschek and Rousseaux’s Handbook of Toxicologic Pathology, 3rd edn. pp 16671773. Eds WM Haschek , CG Rousseaux & MA Wallig . Boston: Academic Press. (https://doi.org/10.1016/B978-0-12-415759-0.00047-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Klinkhammer BM & Boor P 2023 Kidney fibrosis: emerging diagnostic and therapeutic strategies. Mol Aspect Med 93 101206. (https://doi.org/10.1016/j.mam.2023.101206)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Koeth RA , Wang Z , Levison BS , et al. 2013 Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med 19 576585. (https://doi.org/10.1038/nm.3145)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Koppe L , Fouque D & Soulage CO 2018 The role of gut microbiota and diet on uremic retention solutes production in the context of chronic kidney disease. Toxins 10 155. (https://doi.org/10.3390/toxins10040155)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lakshmanan AP , Al Za’abi M , Ali BH , et al. 2021 The influence of the prebiotic gum acacia on the intestinal microbiome composition in rats with experimental chronic kidney disease. Biomed Pharmacother 133 110992. (https://doi.org/10.1016/j.biopha.2020.110992)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lau WL , Kalantar-Zadeh K & Vaziri ND 2015 The gut as a source of inflammation in chronic kidney disease. Nephron 130 9298. (https://doi.org/10.1159/000381990)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lauriola M , Farré R , Evenepoel P , et al. 2023a Food-derived uremic toxins in chronic kidney disease. Toxins 15 116. (https://doi.org/10.3390/toxins15020116)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lee YJ , Li KY , Wang PJ , et al. 2020 Alleviating chronic kidney disease progression through modulating the critical genus of gut microbiota in a cisplatin-induced Lanyu pig model. J Food Drug Anal 28 103114. (https://doi.org/10.38212/2224-6614.1225)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lekawanvijit S , Adrahtas A , Kelly DJ , et al. 2009 Does indoxyl sulfate, a uremic toxin, have direct effects on cardiac fibroblasts & myocytes? Heart Lung Circ 18 S174. (https://doi.org/10.1016/j.hlc.2009.05.395)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li T , Gua C , Wu B , et al. 2018 Increased circulating trimethylamine N-oxide contributes to endothelial dysfunction in a rat model of chronic kidney disease. Biochem Biophys Res Commun 495 20712077. (https://doi.org/10.1016/j.bbrc.2017.12.069)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li X , Geng J , Zhao J , et al. 2019 Trimethylamine N-oxide exacerbates cardiac fibrosis via activating the NLRP3 inflammasome. Front Physiol 10 866. (https://doi.org/10.3389/fphys.2019.00866)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li L , Xiong Q , Zhao J , et al. 2020 Inulin-type fructan intervention restricts the increase in gut microbiome-generated indole in patients with peritoneal dialysis: a randomized crossover study. Am J Clin Nutr 111 10871099. (https://doi.org/10.1093/ajcn/nqz337)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li H-B , Xu M-L , Xu X-D , et al. 2022a Faecalibacterium prausnitzii attenuates CKD via butyrate-renal GPR43 axis. Circ Res 131 e120e134. (https://doi.org/10.1161/circresaha.122.320184)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li J , Morrow C , Barnes S , et al. 2022b Gut microbiome composition and serum metabolome profile among individuals with spinal cord injury and normal glucose tolerance or prediabetes/type 2 diabetes. Arch Phys Med Rehabil 103 702710. (https://doi.org/10.1016/j.apmr.2021.03.043)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li Z , Ke G , Song L , et al. 2022c Association between cardiac outcomes and indoxyl sulfate levels in hemodialysis patients: a cross-sectional study. Kidney Blood Press Res 47 239246. (https://doi.org/10.1159/000521422)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liabeuf S , Cheddani L & Massy Z 2018 Uremic toxins and clinical outcomes: the impact of kidney transplantation. Toxins 10 229. (https://doi.org/10.3390/toxins10060229)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lim PS , Wang HF , Lee MC , et al. 2021 The efficacy of lactobacillus-containing probiotic supplementation in hemodialysis patients: a randomized, double-blind, placebo-controlled trial. J Ren Nutr 31 189198. (https://doi.org/10.1053/j.jrn.2020.07.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lin TY , Chou HH , Huang HL , et al. 2020 Indoxyl sulfate and incident peripheral artery disease in hemodialysis patients. Toxins 12 696. (https://doi.org/10.3390/toxins12110696)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lin X , Liang W , Li L , et al. 2022 The accumulation of gut microbiome–derived indoxyl sulfate and P-cresyl sulfate in patients with end-stage renal disease. J Ren Nutr 32 578586. (https://doi.org/10.1053/j.jrn.2021.09.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu J , Gao LD , Fu B , et al. 2022a Efficacy and safety of Zicuiyin decoction on diabetic kidney disease: a multicenter, randomized controlled trial. Phytomedicine 100 154079. (https://doi.org/10.1016/j.phymed.2022.154079)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu T , Lu X , Gao W , et al. 2022b Cardioprotection effect of Yiqi–Huoxue–Jiangzhuo formula in a chronic kidney disease mouse model associated with gut microbiota modulation and NLRP3 inflammasome inhibition. Biomed Pharmacother 152 113159. (https://doi.org/10.1016/j.biopha.2022.113159)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liyanage T , Ninomiya T , Jha V , et al. 2015 Worldwide access to treatment for end-stage kidney disease: a systematic review. Lancet 385 19751982. (https://doi.org/10.1016/s0140-6736(14)61601-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lu C-L , Zheng C-M , Lu K-C , et al. 2021 Indoxyl-sulfate-induced redox imbalance in chronic kidney disease. Antioxidants 10 936. (https://doi.org/10.3390/antiox10060936)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lu C , Wu L , Tang M-Y , et al. 2023 Indoxyl sulfate in atherosclerosis. Toxicol Lett 383 204212. (https://doi.org/10.1016/j.toxlet.2023.07.001)

  • Lucas B & Taal MW 2023 Epidemiology and causes of chronic kidney disease. Medicine 51 165169. (https://doi.org/10.1016/j.mpmed.2022.12.003)

  • Lúcio HG , Lopes Rd CSO , Gomes MJC , et al. 2024 A symbiotic meal containing extruded sorghum and probiotic (Bifidobacterium longum) ameliorated intestinal health markers in individuals with chronic kidney disease: a secondary analysis of a subsample from a previous randomized and controlled clinical trial. Nutrients 16 1852. (https://doi.org/10.3390/nu16121852)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Luo L , Luo J , Cai Y , et al. 2022 Inulin-type fructans change the gut microbiota and prevent the development of diabetic nephropathy. Pharmacol Res 183 106367. (https://doi.org/10.1016/j.phrs.2022.106367)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mafra D , Ribeiro M , Fonseca L , et al. 2022 Archaea from the gut microbiota of humans: could be linked to chronic diseases? Anaerobe 77 102629. (https://doi.org/10.1016/j.anaerobe.2022.102629)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Matijašić M , Meštrović T , Čipčić Paljetak H , et al. 2020 Gut microbiota beyond bacteria – mycobiome, virome, archaeome, and eukaryotic parasites in IBD. Int J Mol Sci 21 2668. (https://doi.org/10.3390/ijms21082668)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • De Mauri A , Carrera D , Bagnati M , et al. 2020 Probiotics-addicted low-protein diet for microbiota modulation in patients with advanced chronic kidney disease (ProLowCKD): a protocol of placebo-controlled randomized trial. J Funct Foods 74 104133. (https://doi.org/10.1016/j.jff.2020.104133)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • McFarlane C , Krishnasamy R , Stanton T , et al. 2021 Synbiotics easing renal failure by improving gut microbiology ii (Synergy ii): a feasibility randomized controlled trial. Nutrients 13 4481. (https://doi.org/10.3390/nu13124481)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • McFarlane C , Krishnasamy R , Stanton T , et al. 2022 Diet quality and protein-bound uraemic toxins: investigation of novel risk factors and the role of microbiome in chronic kidney disease. J Ren Nutr 32 542551. (https://doi.org/10.1053/j.jrn.2021.10.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • McIntyre CW , Harrison LEA , Eldehni MT , et al. 2011 Circulating endotoxemia: a novel factor in systemic inflammation and cardiovascular disease in chronic kidney disease. Clin J Am Soc Nephrol 6 133141. (https://doi.org/10.2215/cjn.04610510)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Meijers BKI & Evenepoel P 2011 The gut–kidney axis: indoxyl sulfate, p-cresyl sulfate and CKD progression. Nephrol Dial Transplant 26 759761. (https://doi.org/10.1093/ndt/gfq818)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Missailidis C , Hällqvist J , Qureshi AR , et al. 2016 Serum trimethylamine-N-oxide is strongly related to renal function and predicts outcome in chronic kidney disease. PLoS One 11 e0141738. (https://doi.org/10.1371/journal.pone.0141738)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mitrović M , Stanković-Popović V , Tolinački M , et al. 2023 The impact of synbiotic treatment on the levels of gut-derived uremic toxins, inflammation, and gut microbiome of chronic kidney disease patients – a randomized trial. J Ren Nutr 33 278288. (https://doi.org/10.1053/j.jrn.2022.07.008)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Miyata N , Hata T , Takakura S , et al. 2021 Metabolomics profile of Japanese female patients with restricting-type anorexia nervosa. Physiol Behav 228 113204. (https://doi.org/10.1016/j.physbeh.2020.113204)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mo X , Cheng R , Shen L , et al. 2024 High-fat diet induces sarcopenic obesity in natural aging rats through the gut–trimethylamine N-oxide–muscle axis. J Adv Res S2090-1232 00205214. (https://doi.org/10.1016/j.jare.2024.05.015)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Moon SJ , Hwang J , Kang WK , et al. 2022 Administration of the probiotic Lactiplantibacillus paraplantarum is effective in controlling hyperphosphatemia in 5/6 nephrectomy rat model. Life Sci 306 120856. (https://doi.org/10.1016/j.lfs.2022.120856)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mutsaers HAM , Wilmer MJG , Reijnders D , et al. 2013 Uremic toxins inhibit renal metabolic capacity through interference with glucuronidation and mitochondrial respiration. Biochim Biophys Acta 1832 142150. (https://doi.org/10.1016/j.bbadis.2012.09.006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mutsaers HAM , Stribos EGD , Glorieux G , et al. 2015 Chronic kidney disease and fibrosis: the role of uremic retention solutes. Front Med 2 60. (https://doi.org/10.3389/fmed.2015.00060)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nallu A , Sharma S , Ramezani A , et al. 2017 Gut microbiome in chronic kidney disease: challenges and opportunities. Transl Res 179 2437. (https://doi.org/10.1016/j.trsl.2016.04.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Natarajan R , Pechenyak B , Vyas U , et al. 2014 Randomized controlled trial of strain-specific probiotic formulation (renadyl) in dialysis patients. BioMed Res Int 2014 568571. (https://doi.org/10.1155/2014/568571)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nissen L , Prete R , Casciano F , et al. 2024 Modulation of human colon microbiota by naturally debittered olive patè enriched with Lactiplantibacillus plantarum in an in vitro intestinal model. LWT 198 116014. (https://doi.org/10.1016/j.lwt.2024.116014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Niwa T & Shimizu H 2012 Indoxyl sulfate induces nephrovascular senescence. J Ren Nutr 22 102106. (https://doi.org/10.1053/j.jrn.2011.10.032)

  • Oladi-Ghadikolaei R , Aliasgharzadeh A , Shayanfar A , et al. 2023 Serum levels of indoxyl sulfate and P-cresol in type II diabetic patients with and without nephropathy. Iranian J Kidney Dis 17 126134. (https://doi.org/10.52547/ijkd.7266)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pan L , Han P , Ma S , et al. 2020 Abnormal metabolism of gut microbiota reveals the possible molecular mechanism of nephropathy induced by hyperuricemia. Acta Pharm Sin B 10 249261. (https://doi.org/10.1016/j.apsb.2019.10.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pan L , Yu H , Fu J , et al. 2023 Berberine ameliorates chronic kidney disease through inhibiting the production of gut-derived uremic toxins in the gut microbiota. Acta Pharm Sin B 13 15371553. (https://doi.org/10.1016/j.apsb.2022.12.010)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pang X , Guo Z , Ao L , et al. 2024 Metabolomics combined with intestinal microbiota analysis to reveal the effects of Inonotus hispidus on chronic kidney disease caused by high-protein diet. Food Biosci 61 104354. (https://doi.org/10.1016/j.fbio.2024.104354)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Di Paola R , De A , Izhar R , et al. 2023 Possible effects of uremic toxins p-cresol, indoxyl sulfate, p-cresyl sulfate on the development and progression of colon cancer in patients with chronic renal failure. Genes 14 1257. (https://doi.org/10.3390/genes14061257)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Park J , Nam KH , Nam BY , et al. 2024 Lactobacillus acidophilus KBL409 protects against kidney injury via improving mitochondrial function with chronic kidney disease. Eur J Nutr 63 21212135. (https://doi.org/10.1007/s00394-024-03408-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pei T , Hu R , Wang F , et al. 2023 Akkermansia muciniphila ameliorates chronic kidney disease interstitial fibrosis via the gut-renal axis. Microb Pathog 174 105891. (https://doi.org/10.1016/j.micpath.2022.105891)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pivari F , Mingione A , Piazzini G , et al. 2022 Curcumin supplementation (Meriva®) modulates inflammation, lipid peroxidation and gut microbiota composition in chronic kidney disease. Nutrients 14 231. (https://doi.org/10.3390/nu14010231)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pluznick JL 2016 Gut microbiota in renal physiology: focus on short-chain fatty acids and their receptors. Kidney Int 90 11911198. (https://doi.org/10.1016/j.kint.2016.06.033)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Poesen R , Evenepoel P , de Loor H , et al. 2016 The influence of renal transplantation on retained microbial–human co-metabolites. Nephrol Dial Transplant 31 17211729. (https://doi.org/10.1093/ndt/gfw009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Qin S , Zhang K , Ding X , et al. 2023 Microbiome-metabolomics analysis insight into the effects of dietary resistant starch on intestinal integrity. Food Chem 401 134148. (https://doi.org/10.1016/j.foodchem.2022.134148)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Qiu L , Yang D , Tao X , et al. 2017 Enterobacter aerogenes ZDY01 attenuates choline-induced trimethylamine N-oxide levels by remodeling gut microbiota in mice. J Microbiol Biotechnol 27 14911499. (https://doi.org/10.4014/jmb.1703.03039)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Qiu L , Tao X , Xiong H , et al. 2018 Lactobacillus plantarum ZDY04 exhibits a strain-specific property of lowering TMAO via the modulation of gut microbiota in mice. Food Funct 9 42994309. (https://doi.org/10.1039/c8fo00349a)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rashidi A , Ebadi M , Rehman TU , et al. 2021 Altered microbiota-host metabolic cross talk preceding neutropenic fever in patients with acute leukemia. Blood Adv 5 39373950. (https://doi.org/10.1182/bloodadvances.2021004973)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rath S , Heidrich B , Pieper DH , et al. 2017 Uncovering the trimethylamine-producing bacteria of the human gut microbiota. Microbiome 5 5414. (https://doi.org/10.1186/s40168-017-0271-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ravid JD , Kamel MH & Chitalia VC 2021 Uraemic solutes as therapeutic targets in CKD-associated cardiovascular disease. Nat Rev Nephrol 17 402416. (https://doi.org/10.1038/s41581-021-00408-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ren Z , Fan Y , Li A , et al. 2020 Alterations of the human gut microbiome in chronic kidney disease. Adv Sci 7 2001936. (https://doi.org/10.1002/advs.202001936)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ribeiro M , Kemp JA , Cardozo L , et al. 2024 Trimethylamine N-oxide (TMAO) plasma levels in patients with different stages of chronic kidney disease. Toxins 17 15. (https://doi.org/10.3390/toxins17010015)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rossi M , Campbell KL , Johnson DW , et al. 2014 Protein-bound uremic toxins, inflammation and oxidative stress: a cross-sectional study in stage 3–4 chronic kidney disease. Arch Med Res 45 309317. (https://doi.org/10.1016/j.arcmed.2014.04.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sapian S , Budin SB , Taib IS , et al. 2022 Role of polyphenol in regulating oxidative stress, inflammation, fibrosis, and apoptosis in diabetic nephropathy. Endocr Metab Immune Disord Drug Targets 22 453470. (https://doi.org/10.2174/1871530321666211119144309)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schiattarella GG , Sannino A , Toscano E , et al. 2017 Gut microbe-generated metabolite trimethylamine-N-oxide as cardiovascular risk biomarker: a systematic review and dose-response meta-analysis. Eur Heart J 38 29482956. (https://doi.org/10.1093/eurheartj/ehx342)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shi C , Han X , Guo W , et al. 2022 Disturbed Gut-Liver axis indicating oral exposure to polystyrene microplastic potentially increases the risk of insulin resistance. Environ Int 164 107273. (https://doi.org/10.1016/j.envint.2022.107273)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shi W , Li Z , Wang W , et al. 2024 Dynamic gut microbiome-metabolome in cationic bovine serum albumin induced experimental immune-complex glomerulonephritis and effect of losartan and mycophenolate mofetil on microbiota modulation. J Pharm Anal 14 100931. (https://doi.org/10.1016/j.jpha.2023.12.021)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Simeoni M , Di Paola R , Abate M , et al. 2024a WCN24-804 Intestinal dysbiosis-related uremic toxins and colon cancer: p-cresyl sulphate and Indoxyl sulphate stimulate colon cancer progression in patients with CKD. Kidney Int Rep 9 S621S622. (https://doi.org/10.1016/j.ekir.2024.02.1308)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Simeoni M , DI Paola R , De A , et al. 2024b #883 P-cresyl sulphate and Indoxyl sulphate: two uremic toxins stimulating colon cancer progression in patients with chronic renal failure. Nephrol Dial Transplant 39 (Supplement 1) 883. (https://doi.org/10.1093/ndt/gfae069.216)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Snelson M , Biruete A , McFarlane C , et al. 2020 A renal clinician’s guide to the gut microbiota. J Ren Nutr 30 384395. (https://doi.org/10.1053/j.jrn.2019.11.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sohn MB , Gao B , Kendrick C , et al. 2024 Targeting gut microbiome with prebiotic in patients with CKD: the TarGut-CKD study. Kidney Int Rep 9 671685. (https://doi.org/10.1016/j.ekir.2023.12.017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Stanford J , Charlton K , Stefoska-Needham A , et al. 2021 Associations among plant-based diet quality, uremic toxins, and gut microbiota profile in adults undergoing hemodialysis therapy. J Ren Nutr 31 177188. (https://doi.org/10.1053/j.jrn.2020.07.008)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Stubbs JR , House JA , Ocque AJ , et al. 2016 Serum trimethylamine-N-oxide is elevated in CKD and correlates with coronary atherosclerosis burden. J Am Soc Nephrol 27 305313. (https://doi.org/10.1681/asn.2014111063)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Stuivenberg GA , Chmiel JA , Akouris PP , et al. 2022 Probiotic Bifidobacteria Mitigate the Deleterious effects of para-cresol in a drosophila melanogaster toxicity model. mSphere 7 e0044622. (https://doi.org/10.1128/msphere.00446-22)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sun Y , Zhang S , He H , et al. 2023 Comprehensive evaluation of the prebiotic properties of dendrobium officinale polysaccharides, β-glucan, and inulin during in vitro fermentation via multi-omics analysis. Int J Biol Macromol 253 127326. (https://doi.org/10.1016/j.ijbiomac.2023.127326)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Takemura S , Minamiyama Y , Ito N , et al. 2024 Heat-treated and/or lysozyme-treated enterococcus faecalis (FK-23) improves the progression of renal disease in a unilateral ischemia-reperfusion injury rat model. J Clin Biochem Nutr 75 7889. (https://doi.org/10.3164/jcbn.24-29)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Takkavatakarn K , Phannajit J , Udomkarnjananun S , et al. 2022 Association between indoxyl sulfate and dialysis initiation and cardiac outcomes in chronic kidney disease patients. Int J Nephrol Renovascular Dis 15 115126. (https://doi.org/10.2147/ijnrd.s354658)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tran NTD , Chaidee A , Surapinit A , et al. 2023 Strongyloides stercoralis infection reduces fusicatenibacter and anaerostipes in the gut and increases bacterial amino-acid metabolism in early-stage chronic kidney disease. Heliyon 9 e19859. (https://doi.org/10.1016/j.heliyon.2023.e19859)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ufnal M , Jazwiec R , Dadlez M , et al. 2014 Trimethylamine-N-Oxide: a carnitine-derived metabolite that prolongs the hypertensive effect of angiotensin II in rats. Can J Cardiol 30 17001705. (https://doi.org/10.1016/j.cjca.2014.09.010)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vandaele R 2023. The influence of indoxyl sulfate and p-cresyl sulfate on the renal clearance and on the plasma protein binding of drugs used in CKD. A Master dissertation for the study programme Master in Pharmaceutical Care. Ghent University, 2022-2023

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vaziri ND , Wong J , Pahl M , et al. 2013 Chronic kidney disease alters intestinal microbial flora. Kidney Int 83 308315. (https://doi.org/10.1038/ki.2012.345)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vaziri ND , Zhao Y-Y & 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 737746. (https://doi.org/10.1093/ndt/gfv095)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vos T , Lim SS , Abbafati C , et al. 2020 Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet 396 12041222. (https://doi.org/10.1016/s0140-6736(20)30925-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wakamatsu T , Yamamoto S , Yoshida S , et al. 2024 Indoxyl sulfate-induced macrophage toxicity and therapeutic strategies in uremic atherosclerosis. Toxins 16 254. (https://doi.org/10.3390/toxins16060254)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang Q , Guo M , Liu Y , et al. 2022 Bifidobacterium breve and Bifidobacterium longum attenuate choline-induced plasma trimethylamine N-oxide production by modulating gut microbiota in mice. Nutrients 14 1222. (https://doi.org/10.3390/nu14061222)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang B , Zhang B , Zhou L , et al. 2023 Multi-omics reveals diet-induced metabolic disorders and liver inflammation via microbiota-gut-liver axis. J Nutr Biochem 111 109183. (https://doi.org/10.1016/j.jnutbio.2022.109183)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Weaver DJ & Mitsnefes M 2018 Cardiovascular disease in children and adolescents with chronic kidney disease. Semin Nephrol 38 559569. (https://doi.org/10.1016/j.semnephrol.2018.08.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wiese GN , Biruete A , Stremke ER , et al. 2024 Gut microbiota and uremic retention solutes in adults with moderate CKD: a 6-day controlled feeding study. J Ren Nutr 34 2634. (https://doi.org/10.1053/j.jrn.2023.06.011)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wu I-W , Hsu K-H , Lee C-C , et al. 2011 P-cresyl sulphate and indoxyl sulphate predict progression of chronic kidney disease. Nephrol Dial Transplant 26 938947. (https://doi.org/10.1093/ndt/gfq580)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wu W-K , Chen C-C , Liu P-Y , et al. 2019 Identification of TMAO-producer phenotype and host–diet–gut dysbiosis by carnitine challenge test in human and germ-free mice. Gut 68 14391449. (https://doi.org/10.1136/gutjnl-2018-317155)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wu P-H , Lin Y-T , Chiu Y-W , et al. 2021 The relationship of indoxyl sulfate and p-cresyl sulfate with target cardiovascular proteins in hemodialysis patients. Sci Rep 11 3786. (https://doi.org/10.1038/s41598-021-83383-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wu C , Wu C , Peng L , et al. 2024 Multi-omics approaches for the understanding of therapeutic mechanism for Huang-Qi-Long-Dan Granule against ischemic stroke. Pharmacol Res 205 107229. (https://doi.org/10.1016/j.phrs.2024.107229)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Xie S , Deng N , Fang L , et al. 2024 TMAO is involved in kidney-yang deficiency syndrome diarrhea by mediating the “gut-kidney axis”. Heliyon 10 e35461. (https://doi.org/10.1016/j.heliyon.2024.e35461)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Xu H , Bian X , Wang H , et al. 2024 Akkermansia muciniphila postbiotic administration mitigates choline-induced plasma Trimethylamine-N-Oxide production in mice. Appl Biol Chem 67 52. (https://doi.org/10.1186/s13765-024-00905-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yamamoto H , Tsuruoka S , Ioka T , et al. 2006 Indoxyl sulfate stimulates proliferation of rat vascular smooth muscle cells. Kidney Int 69 17801785. (https://doi.org/10.1038/sj.ki.5000340)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yan T , Shi L , Liu T , et al. 2023 Diet-rich in wheat bran modulates tryptophan metabolism and AhR/IL-22 signalling mediated metabolic health and gut dysbacteriosis: a novel prebiotic-like activity of wheat bran. Food Res Int 163 112179. (https://doi.org/10.1016/j.foodres.2022.112179)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yang T , Richards EM , Pepine CJ , et al. 2018 The gut microbiota and the brain–gut–kidney axis in hypertension and chronic kidney disease. Nat Rev Nephrol 14 442456. (https://doi.org/10.1038/s41581-018-0018-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yang Y , Ma C , Li S , et al. 2023 Urinary microbiota and serum metabolite analysis in patients with diabetic kidney disease. Heliyon 9 e17040. (https://doi.org/10.1016/j.heliyon.2023.e17040)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yi ZY , Peng YJ , Hui BP , et al. 2023 Zuogui-Jiangtang-Yishen decoction prevents diabetic kidney disease: intervene pyroptosis induced by trimethylamine n-oxide through the mROS-NLRP3 axis. Phytomedicine 114 154775. (https://doi.org/10.1016/j.phymed.2023.154775)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yu D , Shu X , Rivera ES , et al. 2019 Urinary levels of trimethylamine-N-oxide and incident coronary heart disease: a prospective investigation among urban Chinese adults. J Am Heart Assoc 8 e010606. (https://doi.org/10.1161/jaha.118.010606)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang M , Cui R , Zhou Y , et al. 2023 Accumulation of renal fibrosis in hyperuricemia rats is attributed to the recruitment of mast cells, activation of the TGF-β1/smad2/3 pathway, and aggravation of oxidative stress. Int J Mol Sci 24 10839. (https://doi.org/10.3390/ijms241310839)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang B , Zhang X , Luo Z , et al. 2024a Microbiome and metabolome dysbiosis analysis in impaired glucose tolerance for the prediction of progression to diabetes mellitus. J Genet Genomics 51 7586. (https://doi.org/10.1016/j.jgg.2023.08.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang Y , Han L , Dong J , et al. 2024b Shaoyao decoction improves damp-heat colitis by activating the AHR/IL-22/STAT3 pathway through tryptophan metabolism driven by gut microbiota. J Ethnopharmacol 326 117874. (https://doi.org/10.1016/j.jep.2024.117874)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zemaitis MR , Foris LA , Katta S , et al. 2024 Uremia. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing. Available from: https://www.ncbi.nlm.nih.gov/books/NBK441859/

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhao Y & Wang Z 2020 Gut microbiome and cardiovascular disease. Curr Opin Cardiol 35 207218. (https://doi.org/10.1097/hco.0000000000000720)

  • Zhu W , Gregory JC , Org E , et al. 2016 Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell 165 111124. (https://doi.org/10.1016/j.cell.2016.02.011)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhu N , Duan H , Feng Y , et al. 2023 Magnesium lithospermate B ameliorates diabetic nephropathy by suppressing the uremic toxin formation mediated by gut microbiota. Eur J Pharmacol 953 175812. (https://doi.org/10.1016/j.ejphar.2023.175812)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zixin Y , Lulu C , Xiangchang Z , et al. 2022 TMAO as a potential biomarker and therapeutic target for chronic kidney disease: a review. Front Pharmacol 13 929262. (https://doi.org/10.3389/fphar.2022.929262)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zwaenepoel B , De Backer T , Glorieux G , et al. 2024 Predictive value of protein-bound uremic toxins for heart failure in patients with chronic kidney disease. ESC Heart Fail 11 466474. (https://doi.org/10.1002/ehf2.14566)

    • PubMed
    • Search Google Scholar
    • Export Citation

 

  • Collapse
  • Expand
Get Permissions
  • Figure 1

    Graph of the most mentioned bacteria by phylum in the studies conducted in 2020–2024.

  • Figure 2

    Graph of the most mentioned bacteria by family in the studies conducted in 2020–2024.

  • Figure 3

    Graph of the most mentioned bacteria by genus in the studies conducted in 2020–2024.

  • Ackley W , Soiefer L , Etinger A , et al. 2017 Uremic retention solutes. In Aspects In Dialysis, pp 927. London: IntechOpen.

  • Anders H-J , Andersen K & Stecher B 2013 The intestinal microbiota, a leaky gut, and abnormal immunity in kidney disease. Kidney Int 83 10101016. (https://doi.org/10.1038/ki.2012.440)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Andersen K , Kesper MS , Marschner JA , et al. 2017 Intestinal dysbiosis, barrier dysfunction, and bacterial translocation account for CKD–related systemic inflammation. J Am Soc Nephrol 28 7683. (https://doi.org/10.1681/asn.2015111285)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Asai H , Hirata J & Watanabe-Akanuma M 2018 Indoxyl glucuronide, a protein-bound uremic toxin, inhibits hypoxia-inducible factor–dependent erythropoietin expression through activation of aryl hydrocarbon receptor. Biochem Biophys Res Commun 504 538544. (https://doi.org/10.1016/j.bbrc.2018.09.018)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Asgharian M , Gholizadeh P , Samadi Kafil H , et al. 2022 Correlation of inflammatory biomarkers with the diversity of Bacteroidaceae, Bifidobacteriaceae, Prevotellaceae and Lactobacillaceae families in the intestinal microbiota of patients with end stage renal disease. Adv Med Sci 67 304310. (https://doi.org/10.1016/j.advms.2022.07.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Attaye I , Lassen PB , Adriouch S , et al. 2023 Protein supplementation changes gut microbial diversity and derived metabolites in subjects with type 2 diabetes. iScience 26 107471. (https://doi.org/10.1016/j.isci.2023.107471)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Banoglu E , Jha GG & King RS 2001 Hepatic microsomal metabolism of indole to indoxyl, a precursor of indoxyl sulfate. Eur J Drug Metab Pharmacokinet 26 235240. (https://doi.org/10.1007/bf03226377)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barreto FC , Barreto DV , Liabeuf S , et al. 2009 Serum indoxyl sulfate is associated with vascular disease and mortality in chronic kidney disease patients. Clin J Am Soc Nephrol 4 15511558. (https://doi.org/10.2215/cjn.03980609)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Biagi E , Nylund L , Candela M , et al. 2010 Through ageing, and beyond: gut microbiota and inflammatory status in seniors and centenarians. PLoS One 5 e10667. (https://doi.org/10.1371/journal.pone.0010667)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bian M , Zhu C , Nie A , et al. 2024 Guizhi Shaoyao Zhimu Decoction ameliorates gouty arthritis in rats via altering gut microbiota and improving metabolic profile. Phytomedicine 131 155800. (https://doi.org/10.1016/j.phymed.2024.155800)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Biruete A , Cross T-WL , Allen JM , et al. 2021 Effect of dietary inulin supplementation on the gut microbiota composition and derived metabolites of individuals undergoing hemodialysis: a pilot study. J Ren Nutr 31 512522. (https://doi.org/10.1053/j.jrn.2020.10.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Caldiroli L , Armelloni S , Eskander A , et al. 2021 Association between the uremic toxins indoxyl-sulfate and p-cresyl-sulfate with sarcopenia and malnutrition in elderly patients with advanced chronic kidney disease. Exp Gerontol 147 111266. (https://doi.org/10.1016/j.exger.2021.111266)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cason CA , Kuntz TM , Chen EB , et al. 2020 Microbiota composition modulates inflammation and neointimal hyperplasia after arterial angioplasty. J Vasc Surg 71 13781389.e3. (https://doi.org/10.1016/j.jvs.2019.06.208)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chang J-F , Hsieh C-Y , Liou J-C , et al. 2020 Scavenging intracellular ROS attenuates p-cresyl sulfate-triggered osteogenesis through MAPK signaling pathway and NF-κB activation in human arterial smooth muscle cells. Toxins 12 472. (https://doi.org/10.3390/toxins12080472)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Charytan DM 2018 Introduction: cardiovascular disease in chronic kidney disease. Semin Nephrol 38 541. (https://doi.org/10.1016/j.semnephrol.2018.08.008)

  • Cheng T-H , Ma M-C , Liao M-T , et al. 2020 Indoxyl sulfate, a tubular toxin, contributes to the development of chronic kidney disease. Toxins 12 684. (https://doi.org/10.3390/toxins12110684)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cigarran Guldris S , González Parra E & Cases Amenós A 2017 Gut microbiota in chronic kidney disease. Revista de la Sociedad Española de Nefrología 37 919. (https://doi.org/10.1016/j.nefro.2016.05.008)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Corradi V , Caprara C , Barzon E , et al. 2024 A possible role of P-cresyl sulfate and indoxyl sulfate as biomarkers in the prediction of renal function according to the GFR (G) categories. Integr Med Nephrol Androl 11 e24e00002. (https://doi.org/10.1097/imna-d-24-00002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Correa-Rotter R , Wesseling C & Johnson RJ 2014 CKD of unknown origin in Central America: the case for a Mesoamerican nephropathy. Am J Kidney Dis 63 506520. (https://doi.org/10.1053/j.ajkd.2013.10.062)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Couser WG , Remuzzi G , Mendis S , et al. 2011 The contribution of chronic kidney disease to the global burden of major noncommunicable diseases. Kidney Int 80 12581270. (https://doi.org/10.1038/ki.2011.368)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cui Y , Kuang D , Wang J , et al. 2024 Effect of soluble dietary fiber on gut microbiota and derived metabolites in stage 3 to 5 chronic kidney disease patients: a randomized controlled trial. J Funct Foods 116 106181. (https://doi.org/10.1016/j.jff.2024.106181)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Daneshamouz S , Eduok U , Abdelrasoul A , et al. 2021 Protein-bound uremic toxins (PBUTs) in chronic kidney disease (CKD) patients: production pathway, challenges and recent advances in renal PBUTs clearance. NanoImpact 21 100299. (https://doi.org/10.1016/j.impact.2021.100299)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Das S , Song Z , Han H , et al. 2022 Intestinal osteopontin protects from alcohol-induced liver injury by preserving the gut microbiome and the intestinal barrier function. CMGH 14 813839. (https://doi.org/10.1016/j.jcmgh.2022.06.012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Delgado-Valero B , Cachofeiro V & Martínez-Martínez E 2021 Fibrosis, the bad actor in cardiorenal syndromes: mechanisms involved. Cells 10 1824. (https://doi.org/10.3390/cells10071824)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Du Z , Wang J , Lu Y , et al. 2020 The cardiac protection of Baoyuan decoction via gut-heart axis metabolic pathway. Phytomedicine 79 153322. (https://doi.org/10.1016/j.phymed.2020.153322)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ebrahim Z , Proost S , Tito RY , et al. 2022 The effect of ß-glucan prebiotic on kidney function, uremic toxins and gut microbiome in stage 3 to 5 chronic kidney disease (CKD) predialysis participants: a randomized controlled trial. Nutrients 14 805. (https://doi.org/10.3390/nu14040805)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ekrikpo UE , Kengne AP , Bello AK , et al. 2018 Chronic kidney disease in the global adult HIV-infected population: a systematic review and meta-analysis. PLoS One 13 e0195443. (https://doi.org/10.1371/journal.pone.0195443)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fang Q , Zheng B , Liu N , et al. 2021 Trimethylamine N-oxide exacerbates renal inflammation and fibrosis in rats with diabetic kidney disease. Front Physiol 12 682482. (https://doi.org/10.3389/fphys.2021.682482)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Felizardo RJF , Castoldi A , Andrade-Oliveira V , et al. 2016 The microbiota and chronic kidney diseases: a double-edged sword. Clin Translational Immunol 5 e86. (https://doi.org/10.1038/cti.2016.36)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Foreman KJ , Marquez N , Dolgert A , et al. 2018 Forecasting life expectancy, years of life lost, and all-cause and cause-specific mortality for 250 causes of death: reference and alternative scenarios for 2016–40 for 195 countries and territories. Lancet 392 20522090. (https://doi.org/10.1016/s0140-6736(18)31694-5)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fu BC , Hullar MAJ , Randolph TW , et al. 2020 Associations of plasma trimethylamine N-oxide, choline, carnitine, and betaine with inflammatory and cardiometabolic risk biomarkers and the fecal microbiome in the Multiethnic Cohort Adiposity Phenotype Study. Am J Clin Nutr 111 12261234. (https://doi.org/10.1093/ajcn/nqaa015)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gao Y , Zhao C-J , Liu Q , et al. 2024 Relationship between serum indoxyl sulfate and Klotho protein and vascular calcification in patients with chronic kidney disease stages 3–5. Int J Endocrinol 2024 8229604. (https://doi.org/10.1155/2024/8229604)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Graboski AL , Kowalewski ME , Simpson JB , et al. 2023 Mechanism-based inhibition of gut microbial tryptophanases reduces serum indoxyl sulfate. Cell Chem Biol 30 14021413.e7. (https://doi.org/10.1016/j.chembiol.2023.07.015)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gruppen EG , Garcia E , Connelly MA , et al. 2017 TMAO is associated with mortality: impact of modestly impaired renal function. Sci Rep 7 13781. (https://doi.org/10.1038/s41598-017-13739-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guldris SC , Parra EG & Amenós AC 2017 Gut microbiota in chronic kidney disease. Nefrologia 37 919. (https://doi.org/10.1016/j.nefroe.2017.01.017)

  • Haonon O , Liu Z , Dangtakot R , et al. 2022 Opisthorchis viverrini infection induces metabolic disturbances in hamsters fed with high fat/high fructose diets: implications for liver and kidney pathologies. J Nutr Biochem 107 109053. (https://doi.org/10.1016/j.jnutbio.2022.109053)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Holle J , Kirchner M , Okun J , et al. 2020 Serum indoxyl sulfate concentrations associate with progression of chronic kidney disease in children. PLoS One 15 e0240446. (https://doi.org/10.1371/journal.pone.0240446)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hoyer FF & Nahrendorf M 2019 Uremic toxins activate macrophages. Circulation 139 97100. (https://doi.org/10.1161/circulationaha.118.037308)

  • Hsu CN , Hou CY , Chang-Chien GP , et al. 2021 Maternal resveratrol therapy protected adult rat offspring against hypertension programmed by combined exposures to asymmetric dimethylarginine and trimethylamine-N-oxide. J Nutr Biochem 93 108630. (https://doi.org/10.1016/j.jnutbio.2021.108630)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hsu CN , Yu HR , Lin IC , et al. 2022 Sodium butyrate modulates blood pressure and gut microbiota in maternal tryptophan-free diet-induced hypertension rat offspring. J Nutr Biochem 108 109090. (https://doi.org/10.1016/j.jnutbio.2022.109090)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hu J , Zhong X , Yan J , et al. 2020 High-throughput sequencing analysis of intestinal flora changes in ESRD and CKD patients. BMC Nephrol 21 12. (https://doi.org/10.1186/s12882-019-1668-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang H , Li K , Lee Y , et al. 2021 Preventive effects of Lactobacillus mixture against chronic kidney disease progression through enhancement of beneficial bacteria and downregulation of gut-derived uremic toxins. J Agric Food Chem 69 73537366. (https://doi.org/10.1021/acs.jafc.1c01547)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang S-T , Shu K-H , Cheng C-H , et al. 2012 Serum total p-cresol and indoxyl sulfate correlated with stage of chronic kidney disease in renal transplant recipients. In Transplantation Proceedings, vol 44, 3rd ed. pp 621624. New York: Elsevier.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang Z , Fang Z , Wu A , et al. 2023 Fermented Psidium guajava leaves regulate the gut microbiota and improve metabolic alterations in diabetic mice. Food Biosci 51 102201. (https://doi.org/10.1016/j.fbio.2022.102201)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Imazu M , Takahama H , Shindo K , et al. 2017 A pathophysiological role of plasma indoxyl sulfate in patients with heart failure. Int J Gerontol 11 6266. (https://doi.org/10.1016/j.ijge.2016.05.010)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Iwasaki K , Miyoshi T , Urabe C , et al. 2024 Impact of serum indoxyl sulfate on one-year adverse events in chronic kidney disease patients with heart failure. J Clin Med 13 4384. (https://doi.org/10.3390/jcm13154384)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jha V , Garcia-Garcia G , Iseki K , et al. 2013 Chronic kidney disease: global dimension and perspectives. Lancet 382 260272. (https://doi.org/10.1016/s0140-6736(13)60687-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jian S , Yang K , Zhang L , et al. 2023 The modulation effects of plant-derived bioactive ingredients on chronic kidney disease: focus on the gut–kidney axis. Food Front 4 262282. (https://doi.org/10.1002/fft2.209)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jiang S , Xie S , Lv D , et al. 2017 Alteration of the gut microbiota in Chinese population with chronic kidney disease. Sci Rep 7 2870. (https://doi.org/10.1038/s41598-017-02989-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jiang S , Wei X , Wang H , et al. 2024 Biotransformed bear bile powder alleviates diet-induced nonalcoholic fatty liver disease in mice by regulating the gut microbiota and reversing lipid metabolism. Arab J Chem 17 105578. (https://doi.org/10.1016/j.arabjc.2023.105578)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jin Q , Black A , Kales SN , et al. 2019 Metabolomics and microbiomes as potential tools to evaluate the effects of the Mediterranean diet. Nutrients 11 207. (https://doi.org/10.3390/nu11010207)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kaminski TW , Pawlak K , Karbowska M , et al. 2019 The impact of antihypertensive pharmacotherapy on interplay between protein-bound uremic toxin (indoxyl sulfate) and markers of inflammation in patients with chronic kidney disease. Int Urol Nephrol 51 491502. (https://doi.org/10.1007/s11255-018-02064-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kang D-W , Adams JB , Vargason T , et al. 2020 Distinct fecal and plasma metabolites in children with autism spectrum disorders and their modulation after microbiota transfer therapy. mSphere 5 e00314-20. (https://doi.org/10.1128/msphere.00314-20)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kawashima Y 1989 Study on the uremic protein binding inhibitors as uremic toxin: toxic effect on erythroid colony formation, lymphocyte blast formation and renal function. The Japanese Journal of Nephrology 31 11511161. (https://doi.org/10.14842/jpnjnephrol1959.31.1151)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kemp JA , Schultz J , Modolon F , et al. 2024 Is there a correlation between TMAO plasma levels and archaea in the gut of patients undergoing hemodialysis? Int Urol Nephrol 17. (https://doi.org/10.1007/s11255-024-04273-5)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Khan KNM , Hard GC & Alden CL 2013 Chapter 47 – kidney. In Haschek and Rousseaux’s Handbook of Toxicologic Pathology, 3rd edn. pp 16671773. Eds WM Haschek , CG Rousseaux & MA Wallig . Boston: Academic Press. (https://doi.org/10.1016/B978-0-12-415759-0.00047-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Klinkhammer BM & Boor P 2023 Kidney fibrosis: emerging diagnostic and therapeutic strategies. Mol Aspect Med 93 101206. (https://doi.org/10.1016/j.mam.2023.101206)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Koeth RA , Wang Z , Levison BS , et al. 2013 Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med 19 576585. (https://doi.org/10.1038/nm.3145)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Koppe L , Fouque D & Soulage CO 2018 The role of gut microbiota and diet on uremic retention solutes production in the context of chronic kidney disease. Toxins 10 155. (https://doi.org/10.3390/toxins10040155)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lakshmanan AP , Al Za’abi M , Ali BH , et al. 2021 The influence of the prebiotic gum acacia on the intestinal microbiome composition in rats with experimental chronic kidney disease. Biomed Pharmacother 133 110992. (https://doi.org/10.1016/j.biopha.2020.110992)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lau WL , Kalantar-Zadeh K & Vaziri ND 2015 The gut as a source of inflammation in chronic kidney disease. Nephron 130 9298. (https://doi.org/10.1159/000381990)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lauriola M , Farré R , Evenepoel P , et al. 2023a Food-derived uremic toxins in chronic kidney disease. Toxins 15 116. (https://doi.org/10.3390/toxins15020116)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lee YJ , Li KY , Wang PJ , et al. 2020 Alleviating chronic kidney disease progression through modulating the critical genus of gut microbiota in a cisplatin-induced Lanyu pig model. J Food Drug Anal 28 103114. (https://doi.org/10.38212/2224-6614.1225)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lekawanvijit S , Adrahtas A , Kelly DJ , et al. 2009 Does indoxyl sulfate, a uremic toxin, have direct effects on cardiac fibroblasts & myocytes? Heart Lung Circ 18 S174. (https://doi.org/10.1016/j.hlc.2009.05.395)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li T , Gua C , Wu B , et al. 2018 Increased circulating trimethylamine N-oxide contributes to endothelial dysfunction in a rat model of chronic kidney disease. Biochem Biophys Res Commun 495 20712077. (https://doi.org/10.1016/j.bbrc.2017.12.069)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li X , Geng J , Zhao J , et al. 2019 Trimethylamine N-oxide exacerbates cardiac fibrosis via activating the NLRP3 inflammasome. Front Physiol 10 866. (https://doi.org/10.3389/fphys.2019.00866)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li L , Xiong Q , Zhao J , et al. 2020 Inulin-type fructan intervention restricts the increase in gut microbiome-generated indole in patients with peritoneal dialysis: a randomized crossover study. Am J Clin Nutr 111 10871099. (https://doi.org/10.1093/ajcn/nqz337)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li H-B , Xu M-L , Xu X-D , et al. 2022a Faecalibacterium prausnitzii attenuates CKD via butyrate-renal GPR43 axis. Circ Res 131 e120e134. (https://doi.org/10.1161/circresaha.122.320184)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li J , Morrow C , Barnes S , et al. 2022b Gut microbiome composition and serum metabolome profile among individuals with spinal cord injury and normal glucose tolerance or prediabetes/type 2 diabetes. Arch Phys Med Rehabil 103 702710. (https://doi.org/10.1016/j.apmr.2021.03.043)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li Z , Ke G , Song L , et al. 2022c Association between cardiac outcomes and indoxyl sulfate levels in hemodialysis patients: a cross-sectional study. Kidney Blood Press Res 47 239246. (https://doi.org/10.1159/000521422)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liabeuf S , Cheddani L & Massy Z 2018 Uremic toxins and clinical outcomes: the impact of kidney transplantation. Toxins 10 229. (https://doi.org/10.3390/toxins10060229)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lim PS , Wang HF , Lee MC , et al. 2021 The efficacy of lactobacillus-containing probiotic supplementation in hemodialysis patients: a randomized, double-blind, placebo-controlled trial. J Ren Nutr 31 189198. (https://doi.org/10.1053/j.jrn.2020.07.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lin TY , Chou HH , Huang HL , et al. 2020 Indoxyl sulfate and incident peripheral artery disease in hemodialysis patients. Toxins 12 696. (https://doi.org/10.3390/toxins12110696)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lin X , Liang W , Li L , et al. 2022 The accumulation of gut microbiome–derived indoxyl sulfate and P-cresyl sulfate in patients with end-stage renal disease. J Ren Nutr 32 578586. (https://doi.org/10.1053/j.jrn.2021.09.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu J , Gao LD , Fu B , et al. 2022a Efficacy and safety of Zicuiyin decoction on diabetic kidney disease: a multicenter, randomized controlled trial. Phytomedicine 100 154079. (https://doi.org/10.1016/j.phymed.2022.154079)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu T , Lu X , Gao W , et al. 2022b Cardioprotection effect of Yiqi–Huoxue–Jiangzhuo formula in a chronic kidney disease mouse model associated with gut microbiota modulation and NLRP3 inflammasome inhibition. Biomed Pharmacother 152 113159. (https://doi.org/10.1016/j.biopha.2022.113159)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liyanage T , Ninomiya T , Jha V , et al. 2015 Worldwide access to treatment for end-stage kidney disease: a systematic review. Lancet 385 19751982. (https://doi.org/10.1016/s0140-6736(14)61601-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lu C-L , Zheng C-M , Lu K-C , et al. 2021 Indoxyl-sulfate-induced redox imbalance in chronic kidney disease. Antioxidants 10 936. (https://doi.org/10.3390/antiox10060936)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lu C , Wu L , Tang M-Y , et al. 2023 Indoxyl sulfate in atherosclerosis. Toxicol Lett 383 204212. (https://doi.org/10.1016/j.toxlet.2023.07.001)

  • Lucas B & Taal MW 2023 Epidemiology and causes of chronic kidney disease. Medicine 51 165169. (https://doi.org/10.1016/j.mpmed.2022.12.003)

  • Lúcio HG , Lopes Rd CSO , Gomes MJC , et al. 2024 A symbiotic meal containing extruded sorghum and probiotic (Bifidobacterium longum) ameliorated intestinal health markers in individuals with chronic kidney disease: a secondary analysis of a subsample from a previous randomized and controlled clinical trial. Nutrients 16 1852. (https://doi.org/10.3390/nu16121852)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Luo L , Luo J , Cai Y , et al. 2022 Inulin-type fructans change the gut microbiota and prevent the development of diabetic nephropathy. Pharmacol Res 183 106367. (https://doi.org/10.1016/j.phrs.2022.106367)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mafra D , Ribeiro M , Fonseca L , et al. 2022 Archaea from the gut microbiota of humans: could be linked to chronic diseases? Anaerobe 77 102629. (https://doi.org/10.1016/j.anaerobe.2022.102629)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Matijašić M , Meštrović T , Čipčić Paljetak H , et al. 2020 Gut microbiota beyond bacteria – mycobiome, virome, archaeome, and eukaryotic parasites in IBD. Int J Mol Sci 21 2668. (https://doi.org/10.3390/ijms21082668)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • De Mauri A , Carrera D , Bagnati M , et al. 2020 Probiotics-addicted low-protein diet for microbiota modulation in patients with advanced chronic kidney disease (ProLowCKD): a protocol of placebo-controlled randomized trial. J Funct Foods 74 104133. (https://doi.org/10.1016/j.jff.2020.104133)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • McFarlane C , Krishnasamy R , Stanton T , et al. 2021 Synbiotics easing renal failure by improving gut microbiology ii (Synergy ii): a feasibility randomized controlled trial. Nutrients 13 4481. (https://doi.org/10.3390/nu13124481)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • McFarlane C , Krishnasamy R , Stanton T , et al. 2022 Diet quality and protein-bound uraemic toxins: investigation of novel risk factors and the role of microbiome in chronic kidney disease. J Ren Nutr 32 542551. (https://doi.org/10.1053/j.jrn.2021.10.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • McIntyre CW , Harrison LEA , Eldehni MT , et al. 2011 Circulating endotoxemia: a novel factor in systemic inflammation and cardiovascular disease in chronic kidney disease. Clin J Am Soc Nephrol 6 133141. (https://doi.org/10.2215/cjn.04610510)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Meijers BKI & Evenepoel P 2011 The gut–kidney axis: indoxyl sulfate, p-cresyl sulfate and CKD progression. Nephrol Dial Transplant 26 759761. (https://doi.org/10.1093/ndt/gfq818)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Missailidis C , Hällqvist J , Qureshi AR , et al. 2016 Serum trimethylamine-N-oxide is strongly related to renal function and predicts outcome in chronic kidney disease. PLoS One 11 e0141738. (https://doi.org/10.1371/journal.pone.0141738)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mitrović M , Stanković-Popović V , Tolinački M , et al. 2023 The impact of synbiotic treatment on the levels of gut-derived uremic toxins, inflammation, and gut microbiome of chronic kidney disease patients – a randomized trial. J Ren Nutr 33 278288. (https://doi.org/10.1053/j.jrn.2022.07.008)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Miyata N , Hata T , Takakura S , et al. 2021 Metabolomics profile of Japanese female patients with restricting-type anorexia nervosa. Physiol Behav 228 113204. (https://doi.org/10.1016/j.physbeh.2020.113204)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mo X , Cheng R , Shen L , et al. 2024 High-fat diet induces sarcopenic obesity in natural aging rats through the gut–trimethylamine N-oxide–muscle axis. J Adv Res S2090-1232 00205214. (https://doi.org/10.1016/j.jare.2024.05.015)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Moon SJ , Hwang J , Kang WK , et al. 2022 Administration of the probiotic Lactiplantibacillus paraplantarum is effective in controlling hyperphosphatemia in 5/6 nephrectomy rat model. Life Sci 306 120856. (https://doi.org/10.1016/j.lfs.2022.120856)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mutsaers HAM , Wilmer MJG , Reijnders D , et al. 2013 Uremic toxins inhibit renal metabolic capacity through interference with glucuronidation and mitochondrial respiration. Biochim Biophys Acta 1832 142150. (https://doi.org/10.1016/j.bbadis.2012.09.006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mutsaers HAM , Stribos EGD , Glorieux G , et al. 2015 Chronic kidney disease and fibrosis: the role of uremic retention solutes. Front Med 2 60. (https://doi.org/10.3389/fmed.2015.00060)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nallu A , Sharma S , Ramezani A , et al. 2017 Gut microbiome in chronic kidney disease: challenges and opportunities. Transl Res 179 2437. (https://doi.org/10.1016/j.trsl.2016.04.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Natarajan R , Pechenyak B , Vyas U , et al. 2014 Randomized controlled trial of strain-specific probiotic formulation (renadyl) in dialysis patients. BioMed Res Int 2014 568571. (https://doi.org/10.1155/2014/568571)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nissen L , Prete R , Casciano F , et al. 2024 Modulation of human colon microbiota by naturally debittered olive patè enriched with Lactiplantibacillus plantarum in an in vitro intestinal model. LWT 198 116014. (https://doi.org/10.1016/j.lwt.2024.116014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Niwa T & Shimizu H 2012 Indoxyl sulfate induces nephrovascular senescence. J Ren Nutr 22 102106. (https://doi.org/10.1053/j.jrn.2011.10.032)

  • Oladi-Ghadikolaei R , Aliasgharzadeh A , Shayanfar A , et al. 2023 Serum levels of indoxyl sulfate and P-cresol in type II diabetic patients with and without nephropathy. Iranian J Kidney Dis 17 126134. (https://doi.org/10.52547/ijkd.7266)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pan L , Han P , Ma S , et al. 2020 Abnormal metabolism of gut microbiota reveals the possible molecular mechanism of nephropathy induced by hyperuricemia. Acta Pharm Sin B 10 249261. (https://doi.org/10.1016/j.apsb.2019.10.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pan L , Yu H , Fu J , et al. 2023 Berberine ameliorates chronic kidney disease through inhibiting the production of gut-derived uremic toxins in the gut microbiota. Acta Pharm Sin B 13 15371553. (https://doi.org/10.1016/j.apsb.2022.12.010)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pang X , Guo Z , Ao L , et al. 2024 Metabolomics combined with intestinal microbiota analysis to reveal the effects of Inonotus hispidus on chronic kidney disease caused by high-protein diet. Food Biosci 61 104354. (https://doi.org/10.1016/j.fbio.2024.104354)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Di Paola R , De A , Izhar R , et al. 2023 Possible effects of uremic toxins p-cresol, indoxyl sulfate, p-cresyl sulfate on the development and progression of colon cancer in patients with chronic renal failure. Genes 14 1257. (https://doi.org/10.3390/genes14061257)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Park J , Nam KH , Nam BY , et al. 2024 Lactobacillus acidophilus KBL409 protects against kidney injury via improving mitochondrial function with chronic kidney disease. Eur J Nutr 63 21212135. (https://doi.org/10.1007/s00394-024-03408-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pei T , Hu R , Wang F , et al. 2023 Akkermansia muciniphila ameliorates chronic kidney disease interstitial fibrosis via the gut-renal axis. Microb Pathog 174 105891. (https://doi.org/10.1016/j.micpath.2022.105891)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pivari F , Mingione A , Piazzini G , et al. 2022 Curcumin supplementation (Meriva®) modulates inflammation, lipid peroxidation and gut microbiota composition in chronic kidney disease. Nutrients 14 231. (https://doi.org/10.3390/nu14010231)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pluznick JL 2016 Gut microbiota in renal physiology: focus on short-chain fatty acids and their receptors. Kidney Int 90 11911198. (https://doi.org/10.1016/j.kint.2016.06.033)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Poesen R , Evenepoel P , de Loor H , et al. 2016 The influence of renal transplantation on retained microbial–human co-metabolites. Nephrol Dial Transplant 31 17211729. (https://doi.org/10.1093/ndt/gfw009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Qin S , Zhang K , Ding X , et al. 2023 Microbiome-metabolomics analysis insight into the effects of dietary resistant starch on intestinal integrity. Food Chem 401 134148. (https://doi.org/10.1016/j.foodchem.2022.134148)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Qiu L , Yang D , Tao X , et al. 2017 Enterobacter aerogenes ZDY01 attenuates choline-induced trimethylamine N-oxide levels by remodeling gut microbiota in mice. J Microbiol Biotechnol 27 14911499. (https://doi.org/10.4014/jmb.1703.03039)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Qiu L , Tao X , Xiong H , et al. 2018 Lactobacillus plantarum ZDY04 exhibits a strain-specific property of lowering TMAO via the modulation of gut microbiota in mice. Food Funct 9 42994309. (https://doi.org/10.1039/c8fo00349a)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rashidi A , Ebadi M , Rehman TU , et al. 2021 Altered microbiota-host metabolic cross talk preceding neutropenic fever in patients with acute leukemia. Blood Adv 5 39373950. (https://doi.org/10.1182/bloodadvances.2021004973)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rath S , Heidrich B , Pieper DH , et al. 2017 Uncovering the trimethylamine-producing bacteria of the human gut microbiota. Microbiome 5 5414. (https://doi.org/10.1186/s40168-017-0271-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ravid JD , Kamel MH & Chitalia VC 2021 Uraemic solutes as therapeutic targets in CKD-associated cardiovascular disease. Nat Rev Nephrol 17 402416. (https://doi.org/10.1038/s41581-021-00408-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ren Z , Fan Y , Li A , et al. 2020 Alterations of the human gut microbiome in chronic kidney disease. Adv Sci 7 2001936. (https://doi.org/10.1002/advs.202001936)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ribeiro M , Kemp JA , Cardozo L , et al. 2024 Trimethylamine N-oxide (TMAO) plasma levels in patients with different stages of chronic kidney disease. Toxins 17 15. (https://doi.org/10.3390/toxins17010015)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rossi M , Campbell KL , Johnson DW , et al. 2014 Protein-bound uremic toxins, inflammation and oxidative stress: a cross-sectional study in stage 3–4 chronic kidney disease. Arch Med Res 45 309317. (https://doi.org/10.1016/j.arcmed.2014.04.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sapian S , Budin SB , Taib IS , et al. 2022 Role of polyphenol in regulating oxidative stress, inflammation, fibrosis, and apoptosis in diabetic nephropathy. Endocr Metab Immune Disord Drug Targets 22 453470. (https://doi.org/10.2174/1871530321666211119144309)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schiattarella GG , Sannino A , Toscano E , et al. 2017 Gut microbe-generated metabolite trimethylamine-N-oxide as cardiovascular risk biomarker: a systematic review and dose-response meta-analysis. Eur Heart J 38 29482956. (https://doi.org/10.1093/eurheartj/ehx342)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shi C , Han X , Guo W , et al. 2022 Disturbed Gut-Liver axis indicating oral exposure to polystyrene microplastic potentially increases the risk of insulin resistance. Environ Int 164 107273. (https://doi.org/10.1016/j.envint.2022.107273)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shi W , Li Z , Wang W , et al. 2024 Dynamic gut microbiome-metabolome in cationic bovine serum albumin induced experimental immune-complex glomerulonephritis and effect of losartan and mycophenolate mofetil on microbiota modulation. J Pharm Anal 14 100931. (https://doi.org/10.1016/j.jpha.2023.12.021)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Simeoni M , Di Paola R , Abate M , et al. 2024a WCN24-804 Intestinal dysbiosis-related uremic toxins and colon cancer: p-cresyl sulphate and Indoxyl sulphate stimulate colon cancer progression in patients with CKD. Kidney Int Rep 9 S621S622. (https://doi.org/10.1016/j.ekir.2024.02.1308)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Simeoni M , DI Paola R , De A , et al. 2024b #883 P-cresyl sulphate and Indoxyl sulphate: two uremic toxins stimulating colon cancer progression in patients with chronic renal failure. Nephrol Dial Transplant 39 (Supplement 1) 883. (https://doi.org/10.1093/ndt/gfae069.216)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Snelson M , Biruete A , McFarlane C , et al. 2020 A renal clinician’s guide to the gut microbiota. J Ren Nutr 30 384395. (https://doi.org/10.1053/j.jrn.2019.11.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sohn MB , Gao B , Kendrick C , et al. 2024 Targeting gut microbiome with prebiotic in patients with CKD: the TarGut-CKD study. Kidney Int Rep 9 671685. (https://doi.org/10.1016/j.ekir.2023.12.017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Stanford J , Charlton K , Stefoska-Needham A , et al. 2021 Associations among plant-based diet quality, uremic toxins, and gut microbiota profile in adults undergoing hemodialysis therapy. J Ren Nutr 31 177188. (https://doi.org/10.1053/j.jrn.2020.07.008)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Stubbs JR , House JA , Ocque AJ , et al. 2016 Serum trimethylamine-N-oxide is elevated in CKD and correlates with coronary atherosclerosis burden. J Am Soc Nephrol 27 305313. (https://doi.org/10.1681/asn.2014111063)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Stuivenberg GA , Chmiel JA , Akouris PP , et al. 2022 Probiotic Bifidobacteria Mitigate the Deleterious effects of para-cresol in a drosophila melanogaster toxicity model. mSphere 7 e0044622. (https://doi.org/10.1128/msphere.00446-22)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sun Y , Zhang S , He H , et al. 2023 Comprehensive evaluation of the prebiotic properties of dendrobium officinale polysaccharides, β-glucan, and inulin during in vitro fermentation via multi-omics analysis. Int J Biol Macromol 253 127326. (https://doi.org/10.1016/j.ijbiomac.2023.127326)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Takemura S , Minamiyama Y , Ito N , et al. 2024 Heat-treated and/or lysozyme-treated enterococcus faecalis (FK-23) improves the progression of renal disease in a unilateral ischemia-reperfusion injury rat model. J Clin Biochem Nutr 75 7889. (https://doi.org/10.3164/jcbn.24-29)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Takkavatakarn K , Phannajit J , Udomkarnjananun S , et al. 2022 Association between indoxyl sulfate and dialysis initiation and cardiac outcomes in chronic kidney disease patients. Int J Nephrol Renovascular Dis 15 115126. (https://doi.org/10.2147/ijnrd.s354658)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tran NTD , Chaidee A , Surapinit A , et al. 2023 Strongyloides stercoralis infection reduces fusicatenibacter and anaerostipes in the gut and increases bacterial amino-acid metabolism in early-stage chronic kidney disease. Heliyon 9 e19859. (https://doi.org/10.1016/j.heliyon.2023.e19859)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ufnal M , Jazwiec R , Dadlez M , et al. 2014 Trimethylamine-N-Oxide: a carnitine-derived metabolite that prolongs the hypertensive effect of angiotensin II in rats. Can J Cardiol 30 17001705. (https://doi.org/10.1016/j.cjca.2014.09.010)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vandaele R 2023. The influence of indoxyl sulfate and p-cresyl sulfate on the renal clearance and on the plasma protein binding of drugs used in CKD. A Master dissertation for the study programme Master in Pharmaceutical Care. Ghent University, 2022-2023

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vaziri ND , Wong J , Pahl M , et al. 2013 Chronic kidney disease alters intestinal microbial flora. Kidney Int 83 308315. (https://doi.org/10.1038/ki.2012.345)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vaziri ND , Zhao Y-Y & 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 737746. (https://doi.org/10.1093/ndt/gfv095)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vos T , Lim SS , Abbafati C , et al. 2020 Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet 396 12041222. (https://doi.org/10.1016/s0140-6736(20)30925-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wakamatsu T , Yamamoto S , Yoshida S , et al. 2024 Indoxyl sulfate-induced macrophage toxicity and therapeutic strategies in uremic atherosclerosis. Toxins 16 254. (https://doi.org/10.3390/toxins16060254)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang Q , Guo M , Liu Y , et al. 2022 Bifidobacterium breve and Bifidobacterium longum attenuate choline-induced plasma trimethylamine N-oxide production by modulating gut microbiota in mice. Nutrients 14 1222. (https://doi.org/10.3390/nu14061222)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang B , Zhang B , Zhou L , et al. 2023 Multi-omics reveals diet-induced metabolic disorders and liver inflammation via microbiota-gut-liver axis. J Nutr Biochem 111 109183. (https://doi.org/10.1016/j.jnutbio.2022.109183)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Weaver DJ & Mitsnefes M 2018 Cardiovascular disease in children and adolescents with chronic kidney disease. Semin Nephrol 38 559569. (https://doi.org/10.1016/j.semnephrol.2018.08.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wiese GN , Biruete A , Stremke ER , et al. 2024 Gut microbiota and uremic retention solutes in adults with moderate CKD: a 6-day controlled feeding study. J Ren Nutr 34 2634. (https://doi.org/10.1053/j.jrn.2023.06.011)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wu I-W , Hsu K-H , Lee C-C , et al. 2011 P-cresyl sulphate and indoxyl sulphate predict progression of chronic kidney disease. Nephrol Dial Transplant 26 938947. (https://doi.org/10.1093/ndt/gfq580)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wu W-K , Chen C-C , Liu P-Y , et al. 2019 Identification of TMAO-producer phenotype and host–diet–gut dysbiosis by carnitine challenge test in human and germ-free mice. Gut 68 14391449. (https://doi.org/10.1136/gutjnl-2018-317155)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wu P-H , Lin Y-T , Chiu Y-W , et al. 2021 The relationship of indoxyl sulfate and p-cresyl sulfate with target cardiovascular proteins in hemodialysis patients. Sci Rep 11 3786. (https://doi.org/10.1038/s41598-021-83383-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wu C , Wu C , Peng L , et al. 2024 Multi-omics approaches for the understanding of therapeutic mechanism for Huang-Qi-Long-Dan Granule against ischemic stroke. Pharmacol Res 205 107229. (https://doi.org/10.1016/j.phrs.2024.107229)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Xie S , Deng N , Fang L , et al. 2024 TMAO is involved in kidney-yang deficiency syndrome diarrhea by mediating the “gut-kidney axis”. Heliyon 10 e35461. (https://doi.org/10.1016/j.heliyon.2024.e35461)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Xu H , Bian X , Wang H , et al. 2024 Akkermansia muciniphila postbiotic administration mitigates choline-induced plasma Trimethylamine-N-Oxide production in mice. Appl Biol Chem 67 52. (https://doi.org/10.1186/s13765-024-00905-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yamamoto H , Tsuruoka S , Ioka T , et al. 2006 Indoxyl sulfate stimulates proliferation of rat vascular smooth muscle cells. Kidney Int 69 17801785. (https://doi.org/10.1038/sj.ki.5000340)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yan T , Shi L , Liu T , et al. 2023 Diet-rich in wheat bran modulates tryptophan metabolism and AhR/IL-22 signalling mediated metabolic health and gut dysbacteriosis: a novel prebiotic-like activity of wheat bran. Food Res Int 163 112179. (https://doi.org/10.1016/j.foodres.2022.112179)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yang T , Richards EM , Pepine CJ , et al. 2018 The gut microbiota and the brain–gut–kidney axis in hypertension and chronic kidney disease. Nat Rev Nephrol 14 442456. (https://doi.org/10.1038/s41581-018-0018-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yang Y , Ma C , Li S , et al. 2023 Urinary microbiota and serum metabolite analysis in patients with diabetic kidney disease. Heliyon 9 e17040. (https://doi.org/10.1016/j.heliyon.2023.e17040)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yi ZY , Peng YJ , Hui BP , et al. 2023 Zuogui-Jiangtang-Yishen decoction prevents diabetic kidney disease: intervene pyroptosis induced by trimethylamine n-oxide through the mROS-NLRP3 axis. Phytomedicine 114 154775. (https://doi.org/10.1016/j.phymed.2023.154775)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yu D , Shu X , Rivera ES , et al. 2019 Urinary levels of trimethylamine-N-oxide and incident coronary heart disease: a prospective investigation among urban Chinese adults. J Am Heart Assoc 8 e010606. (https://doi.org/10.1161/jaha.118.010606)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang M , Cui R , Zhou Y , et al. 2023 Accumulation of renal fibrosis in hyperuricemia rats is attributed to the recruitment of mast cells, activation of the TGF-β1/smad2/3 pathway, and aggravation of oxidative stress. Int J Mol Sci 24 10839. (https://doi.org/10.3390/ijms241310839)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang B , Zhang X , Luo Z , et al. 2024a Microbiome and metabolome dysbiosis analysis in impaired glucose tolerance for the prediction of progression to diabetes mellitus. J Genet Genomics 51 7586. (https://doi.org/10.1016/j.jgg.2023.08.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang Y , Han L , Dong J , et al. 2024b Shaoyao decoction improves damp-heat colitis by activating the AHR/IL-22/STAT3 pathway through tryptophan metabolism driven by gut microbiota. J Ethnopharmacol 326 117874. (https://doi.org/10.1016/j.jep.2024.117874)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zemaitis MR , Foris LA , Katta S , et al. 2024 Uremia. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing. Available from: https://www.ncbi.nlm.nih.gov/books/NBK441859/

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhao Y & Wang Z 2020 Gut microbiome and cardiovascular disease. Curr Opin Cardiol 35 207218. (https://doi.org/10.1097/hco.0000000000000720)

  • Zhu W , Gregory JC , Org E , et al. 2016 Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell 165 111124. (https://doi.org/10.1016/j.cell.2016.02.011)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhu N , Duan H , Feng Y , et al. 2023 Magnesium lithospermate B ameliorates diabetic nephropathy by suppressing the uremic toxin formation mediated by gut microbiota. Eur J Pharmacol 953 175812. (https://doi.org/10.1016/j.ejphar.2023.175812)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zixin Y , Lulu C , Xiangchang Z , et al. 2022 TMAO as a potential biomarker and therapeutic target for chronic kidney disease: a review. Front Pharmacol 13 929262. (https://doi.org/10.3389/fphar.2022.929262)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zwaenepoel B , De Backer T , Glorieux G , et al. 2024 Predictive value of protein-bound uremic toxins for heart failure in patients with chronic kidney disease. ESC Heart Fail 11 466474. (https://doi.org/10.1002/ehf2.14566)

    • PubMed
    • Search Google Scholar
    • Export Citation