Startseite Applying mass spectrometry-based assays to explore gut microbial metabolism and associations with disease
Artikel Öffentlich zugänglich

Applying mass spectrometry-based assays to explore gut microbial metabolism and associations with disease

  • Liam M. Heaney ORCID logo EMAIL logo
Veröffentlicht/Copyright: 22. Oktober 2019
Veröffentlichen auch Sie bei De Gruyter Brill

Abstract

The workings of the gut microbiome have gained increasing interest in recent years through the mounting evidence that the microbiota plays an influential role in human health and disease. A principal focus of this research seeks to further understand the production of metabolic by-products produced by bacteria resident in the gut, and the subsequent interaction of these metabolites on host physiology and pathophysiology of disease. Gut bacterial metabolites of interest are predominately formed via metabolic breakdown of dietary compounds including choline and ʟ-carnitine (trimethylamine N-oxide), amino acids (phenol- and indole-containing uremic toxins) and non-digestible dietary fibers (short-chain fatty acids). Investigations have been accelerated through the application of mass spectrometry (MS)-based assays to quantitatively assess the concentration of these metabolites in laboratory- and animal-based experiments, as well as for direct circulating measurements in clinical research populations. This review seeks to explore the impact of these metabolites on disease, as well as to introduce the application of MS for those less accustomed to its use as a clinical tool, highlighting pertinent research related to its use for measurements of gut bacteria-mediated metabolites to further understand their associations with disease.

Introduction

The average human body plays host to an estimated 4×1013 bacteria, a quantity which is approximately equal to the total number of host human cells [1], [2]. These commensal bacteria are present both on and within the human biological system and can be found in multiple sites including, but not limited to, the oral cavity, respiratory tract, skin, genital regions and gastrointestinal tract [3], [4]. The latter, commonly referred to as the gut microbiome, has been perhaps the most well-studied when investigating the impact of the microbiota on health and disease [5]. Although the co-existence of bacteria and host generally leads to a symbiotic relationship that maintains a mutual status of biochemical homeostasis, incidences where the genetic or metabolic landscape are altered have been associated with ill-health and disease [6]. The change in genetic diversity of the microbiome is often termed as ‘dysbiosis’ and is related to an alteration in the numbers or species of bacteria present within the ecosystem [5]. These changes have been associated with the development and progression of multiple conditions including cardiovascular disease, chronic kidney disease (CKD), non-alcoholic fatty liver disease and diabetes, inflammatory bowel disease, amongst many others [7], [8], [9], [10]. Metabolic changes, of which will be the focus of this discussion, relate to shifts in bacterial metabolism that either increase/decrease or alter the metabolite by-products released as part of their routine cellular workings. These molecules are released into the gut lumen and are capable of crossing the gut epithelial barrier to enter the systemic circulation of the host [11]. As many of these metabolites are categorized as small molecule metabolites, they are particularly well-suited to subsequent analysis by hyphenated mass spectrometry (MS) techniques such as gas chromatography-MS (GC-MS) and liquid chromatography-MS (LC-MS). This review aims to provide a brief introduction to MS for readers who are not familiar with the technology, and to present an overview of how MS-based assays are providing novel and informative data into the metabolism of the gut microbiota through the discussion of selected small molecule gut metabolites. Details are given on how these metabolites are being measured by MS for clinical research with the view to understand their utility as clinical biomarker measurements and lead the way to warrant future translation into routine laboratory medicine.

Basics of MS and its benefits to clinical chemistry

MS is an analytical technology based on the measurement of the molecular mass of a charged molecule, referred to as an ion. The MS instrument can manipulate and/or monitor the flight path of an ion, subsequently assigning a value which is a calculation of the molecular mass divided by the charge state of the ion. This is termed the mass-to-charge ratio and is symbolized by m/z. For small molecule MS, the charge state of the ion is commonly at +1 or −1 (often through the addition [+1] or loss [−1] of a proton [H+ ion]), and therefore the m/z value relates to the approximate molecular mass of the measured analyte. However, in some circumstances (predominantly for measurement of larger molecules such as peptides), the charge state can be increased (e.g. +2, +3, etc.) and so the m/z value will reflect a value that is lower than the molecular mass of the analyte of interest. There are multiple approaches in which to ionize a molecule and they are typically referred to as soft ionization (where the molecule remains predominantly intact) and hard ionization (where the molecule is deconstructed into smaller fragments), with the chosen method based on the instrumentation and suitability to the analyte(s) of interest; see an excellent summary by Gary Siuzdak for more technical information on these processes [12]. The response (intensity) of the measured ion(s) is then plotted against m/z to produce a mass spectrum which can be investigated to understand the presence and intensity for an analyte(s) of interest.

As clinical biochemical analyses are performed on complex matrices such as urine, plasma and serum, in most circumstances the application of MS alone is not adequate to provide a definitive answer to a clinical question. If all molecules present in the mixture enter the mass spectrometer at the same time (known as direct infusion), the instrument will return a response for all ions that are produced which makes it difficult to isolate the analyte of interest and obtain reproducible data (albeit a technique that was traditionally applied for newborn screening [13]). Furthermore, for soft ionization techniques such as electrospray (commonly known as ESI) there is a competition for charge in the ionization source. This means that molecules with greater affinity to achieve a charged state will ‘swamp’ the analysis by suppressing the ionization capability of other molecules. One major issue with this sequestering of charge is that molecules that dominate the ionization space will not be of equal concentration across samples. Therefore, the ion suppression present for one patient sample will not equal that of another, further complicating the analysis and producing what is known as the matrix effect. One way to overcome this phenomenon is to manipulate the molecules so that they enter the mass spectrometer at different times across the analytical run. This is predominantly achieved using a separation technique in which the sample is initially passed through prior to entry into the mass spectrometer. For quantitative bioanalysis, this is typically achieved using chromatographic techniques such as GC and LC. Comprehensive development and optimization of the separation techniques allows the analyte(s) of interest to elute within a given time window with partial or full separation from molecules that are competing for ionization. Moreover, the analyte(s) elutes from the column over a short time period following a Gaussian distribution profile, allowing the area under the curve to be calculated and compared back to calibration curves to provide accurate quantitative data. In order to further adjust for ion suppression, stable isotope labeled (SIL) versions of the analyte(s) (employing multiple incorporation of 2D, 13C or 15N atoms) are included at a known concentration. These SIL compounds behave identically within the chromatographic space to enter the mass spectrometer at the same point as the unlabeled molecule but have an increased molecular mass and can be identified in isolation. Any ion suppression that occurs will do so to both the unlabeled and SIL compound and thus the unlabeled data can be adjusted accordingly against the known SIL concentration.

A further strength of MS for biochemical analyses comes from the application of multiple stage analysis referred to as tandem-MS, or MS/MS. MS can be performed in an open (full) scan process where no prioritization of ions is implemented which can lead to elevated levels of background noise and subsequently a reduction in analytical sensitivity. However, applying a multiple stage selection and filter of ions of interest helps ensure that a specific biomolecule is analyzed. This multiple stage selection, commonly termed as selected or multiple reaction monitoring (S/MRM), employs a filtering process to select an ion(s) of interest, performs collision induced dissociation of that ion(s) to produce smaller fragment molecules, and further selects the product fragments that are specifically associated to the targeted precursor molecule; see Figure 1 for a basic overview of the advantages of MS/MS. These approaches are inherently able to diminish background noise to remove almost all interference while simultaneously improving overall selectivity. S/MRM assays can be performed on different formats of mass spectrometers but are most suited to a triple quadrupole instrument due to its capabilities for high-speed scanning and polarity switching (the ability to change between the measurement of different ions of interest and between positive and negative ion analyses, respectively), analysis over multiple orders of magnitude, two-stage ion filtering providing enhanced selectivity and sensitivity (see Table 1 and Figure 1 for more detail on this process), and an innate robustness for high-throughput workflows. In addition, triple quadrupole instruments are able to perform multiple S/MRM assays simultaneously, providing quantitative data for tens to hundreds of molecules from a single multiplexed analysis [14], [15], [16]. See Table 1 for a brief description on the triple quadrupole and alternative mass analyzers commonly employed for mass spectrometry measurement of biological samples.

Figure 1: A simplified schematic diagram to demonstrate the advantages of MS/MS (using a triple quadrupole mass spectrometer, bottom) over a single mass analyzer (top) for selected/multiple reaction monitoring applications providing highly sensitive, selective and reproducible analyses.
Figure 1:

A simplified schematic diagram to demonstrate the advantages of MS/MS (using a triple quadrupole mass spectrometer, bottom) over a single mass analyzer (top) for selected/multiple reaction monitoring applications providing highly sensitive, selective and reproducible analyses.

Table 1:

A brief description of the mass analyzers commonly employed for MS measurement of biological samples.

Mass analyzerCommon written and verbal abbreviationsBasic mechanism of measurementHigh mass accuracy?aRelative cost
QuadrupoleQuad

Single quad
Consists of two pairs of parallel roads with one pair having a static DC potential applied across it. The second pair has a superimposed, and variable, RF frequency which is set to allow ions with a specific m/z to pass through to the detector, with all other ions either colliding with the rods or being ejected from the ion flight path. This RF frequency can be rapidly altered across a single scan point when multiple ions of interest are present (this relates to the instruments scanning speed), or continuous scanning across a m/z range can be performed (i.e. continuously ramping from the lower to upper m/z value across the analysis – this is an open, or full, scan)NoLow
Triple quadrupoleTriple quad

TQ

QqQ/QQQ
An instrument with three quadrupoles positioned in series. Quadrupole 1 (Q1) is used to select a specific ion or ions of interest (precursor ion); Q2 contains an inert gas (usually N2 or Ar) and has an applied voltage to cause excitation of the ions that leads to collision induced dissociation (fragmentation); and Q3 allows specific selection of an ion or ions that are related to the fragmentation of the precursor ion (product ion)

Note: modern instruments have exchanged Q2 for a smaller non-quadrupole collision cell or have used a curved quadrupole analyzer to help reduce the laboratory footprint of the instrument
NoLow to medium
Time-of-flightToF/TOFIons are accelerated by an electric field across a flight tube that is held under vacuum. The time taken for the ion to travel from the ‘pusher’ to the detector is related to its m/z. Therefore, ions with a greater m/z value require more time to reach the detector. Increases in the ion flight path distance provide increased mass accuracy and therefore modern instruments employ a V or W wave analysis where the ions are reflected by one (V) or more (W) ion mirrors to increase the total distance travelled before hitting the detectorYesMedium
OrbitrapOrbiThe orbitrap mass analyzer works by trapping ions and orbiting them around an inner spindle-like electrode. This process produces a current which can be interpreted by the instrument to generate a m/z value, with ions of different m/z orbiting the central electrode at different frequencies. The orbitrap analyzer is capable of highly sensitive and highly accurate analyses and provides a unique ability to perform experiments applying a parallel reaction monitoring (PRM) approach. This allows precursor ions to be measured, ejected from the analyzer and fragmented in a separate collision cell, and then returned to the analyzer where the product ions can be measured. This means both precursor and product ions can be measured simultaneously and without loss from a single sample analysisYesHigh
  1. aMass accuracy relates to the ability of the analyzer to distinguish between very similar but different mass-to-charge (m/z) ratios (e.g. differences in 0.01 Da or less) and is sometimes referred to as high resolution. Instruments that are not considered as having high mass accuracy are capable of confidently measuring to the nearest Da (nearest m/z integer) and are often referred to as having nominal mass resolution.

Translation of MS into the clinical laboratory has been less rapid than one might have expected given the advantages and benefits of MS for biomarker measurements. While there is no one definitive reason for this slow uptake, a number of contributing factors exist such as: the large initial cost for instrument purchase, the requirement for employment of highly skilled/trained personnel and the often laborious sample preparation required which can be troublesome to automate [13]. There are also limitations for the translation of MS assays from research laboratories into the clinical laboratory, and from one hospital laboratory to another [17]. This difficulty stems from the fact that there are multiple MS set-ups available that do not necessarily match up on specifications such as levels of sensitivity. This means that where an assay is perfectly suited to one laboratory, it may not be transferrable to another, requiring a whole new set of experiments to be performed to optimize and validate for that laboratory. Vogeser et al. [18] recently proposed guidelines for clinically focused MS assay reporting, emphasizing the need to include more extensive information on instrument/assay parameters to allow the translation to an alternative laboratory to be more accessible. The authors further suggested the fundamental necessity to test the assay in multiple independent laboratories in order to compare results. This would allow for comparison of data obtained using different MS set-ups and laboratory personnel. If more analytical teams were to follow suit with these ideas, it would likely improve the overall uptake of MS into routine clinical measurements.

Despite the relatively restricted translation to everyday hospital analysis, research experiments completed using MS-based assays are demonstrating excellent applicability for clinical biomarker science. Over recent years, investigations into the understanding of gut microbial metabolism, via the measurement of gut microbiota-mediated metabolites, have provided novel information on disease pathogenesis, progression and risk prediction. These works have been spearheaded through the application of MS-based analyses and are building a knowledge base that has the capacity to improve diagnostic, prognostic and therapeutic monitoring of diseases that display associations with gut microbiome-mediated metabolism. A selection of the most pertinent examples of current research into gut microbiota-mediated metabolites is provided below.

Trimethylamine N-oxide (TMAO)

TMAO is a metabolite produced within the liver following gut bacterial release of its precursor, TMA. TMA is produced by bacteria as a by-product following the metabolic breakdown of quaternary ammonium-containing dietary compounds such as choline (and choline-containing lipids), ʟ-carnitine and betaine [19], [20]. The released TMA is then able to cross into the hepatic portal vein where it is transported to the liver and converted to TMAO by flavin-containing monooxygenases (e.g. FMO3) via N-oxidation [21]. TMA and TMAO have been considered important in medicine for many years through the metabolic disorder trimethylaminuria, referred to as ‘fish-odor syndrome’. This condition occurs through a genetic defect in FMO3 production, meaning that the TMA produced by gut bacteria cannot be converted to TMAO and therefore builds up within the body [22]. TMA has a strong ‘fish-like’ smell, owing to the high quantities of TMAO that are degraded to TMA in spoiled fish [23], which is then released in the sweat, urine and breath of the patient causing a strong and unpleasant body odor [24].

TMAO was first associated with alternative diseases through the finding that circulating levels were higher in cardiovascular disease patients when compared to matched control participants [19]. This discovery was only possible through the use of MS, applying a technique known as non-targeted metabolomics. Metabolomics studies take samples (often biofluids such as plasma, serum and urine) and apply GC/LC coupled to high-resolution MS (HRMS) using a full-scan approach in an attempt to measure as many compounds present in the matrix as possible [25]. HRMS instruments, including time-of-flight and orbitrap mass analyzers, are capable of measuring m/z values with extremely high accuracy (<0.001% error), providing an improved ability to identify an unknown compound by accurately isolating the response of a selected ion from its co-eluting compounds [26]. This, in turn, provides a snapshot indication of the current metabolic status of the individual, with the view to isolate evidence for associations with a (patho)physiological state of interest [26]. TMAO was further investigated in a large cohort of patients undergoing elective diagnostic cardiac catheterization, where it was observed that individuals with increased circulating levels of the metabolite were at a higher risk of a poor outcome (myocardial infarction, stroke or death), therefore demonstrating a prognostic quality of TMAO as an independent biomarker for disease risk stratification [27].

These initial studies signified the discovery and characterization of TMAO as a potential clinical biomarker and there has since been a wealth of data published on both its measurement and application. The majority of validated TMAO assays are performed by LC-MS/MS using a triple quadrupole mass spectrometer [28], [29], [30], [31]. However, additional MS-based methods have been developed to include the use of HRMS [32], [33], [34] and GC-MS-based protocols [35], [36], alongside non-MS approaches using nuclear magnetic resonance spectroscopy [37]. These MS-based assays have afforded a high-throughput (e.g. 2.5 min per sample [34]), quantitative approach to TMAO measurement and have even allowed for the added development of multiplexed assays where TMAO is measured in parallel with its precursor molecules including TMA, choline, ʟ-carnitine and betaine [38], [39], [40]. Owing to this abundance of high-quality assays within the literature, many researchers have been capable of furthering the scientific and clinical understanding of TMAO and its gut microbial-mediated origins. For example, animal models have demonstrated that the increased the presence of circulating TMAO causes an acceleration in atherosclerotic plaque formation [20] and renal fibrosis [41], enhances platelet hyperactivity [42], exacerbates pressure-overload induced heart failure [43], and promotes inflammatory-mediated endothelial dysfunction [44]. Although these mechanistic implications on TMAO and disease causality have yet to be proven in the human biological model, many researchers have found corroborating evidence to show that the severity/risk of numerous diseases is associated with elevated blood TMAO levels. Owing to the fact that TMAO was first identified in cardiovascular disease, investigations in cardiology have been the most intensely studied and published within the literature. Building on the initial data for the prognostic quality of TMAO in heart disease patients, the utility of TMAO as clinical a biomarker has been further observed for patients diagnosed with chronic and acute heart failure [45], [46], [47], [48], acute coronary syndromes including myocardial infarction [49], [50], as well as other cardiovascular related conditions such as renal disease [41], [51] and cerebral ischemic stroke [52], [53]. Further evidence has been proposed to suggest that TMAO may also provide prognostic quality for hepatic diseases [54], [55], colorectal cancer [56] and pneumonia [57].

In all, these findings indicate that the elevated production/build-up of TMAO has negative associations with a range of diseases and therefore offers a potential therapeutic target to improve overall patient health. As a promising clinical biomarker, TMAO measurements would directly benefit from increased accessibility of mass spectrometry within the clinical laboratory.

Uremic toxins

Uremia, or uremic syndrome, is a progressive condition where a reduction in renal capacity causes a range of molecules to be retained that would otherwise be effectively removed from the systemic circulation by the healthy kidney [58]. These retention solutes can include middle molecules, defined as molecules with a molecular mass of 500–60,000 Da (e.g. β2-microglobulin, parathyroid hormone) [59], and small molecules (<500 Da) that are either free water-soluble compounds (e.g. uric acid, malonaldehyde) or protein-bound molecules (e.g. indole- and phenol-based compounds) [60]. These compounds can exhibit a range of physicochemical characteristics and actions within the body, and those that are associated with pathophysiological actions are collectively termed as uremic toxins (UTs) [61]. While many of these UTs are associated with host metabolism followed by a build-up in concentration, the production of some molecules is entirely dependent on gut microbial metabolic pathways. The most prominent of the microbiota-mediated compounds are based on the metabolism of tryptophan to indole by tryptophanase and tyrosine to p-cresol (pC) by hydroxyphenylacetate decarboxylase, which are then further converted to their sulfated forms through hepatic metabolism (i.e. indoxyl sulfate [IS] and p-cresol sulfate [pCS]) [62]. These molecules have long been associated with disease and have more recently been demonstrated to exhibit a varied array of pathophysiological actions within the cardiovascular and renal systems. These actions include, but are not limited to, the promotion of cardiovascular disease risk factors including thrombotic [63], [64] and atherogenic [65] potential, mediation of blood pressure [66], [67], vascular calcification [68], cardiac fibrosis [69] and impairment of endothelial progenitor cell-mediated neovascularization [70]; as well the induction of renal fibrosis [71], inflammation [72], [73] and cellular senescence [74], [75]. Interestingly, IS has also been shown to induce endothelial cell-derived release of microvesicles that contain upregulated levels of miRNAs that are associated with the downstream promotion of inflammation, cellular senescence and apoptosis [76].

There has been an invested interest in the measurement of IS and pC in biosamples, with original analyses being pioneered with the application of high-performance liquid chromatography (HPLC) coupled to ultraviolet light or fluorescence detectors [77], [78], [79], [80]. These methods predominantly assessed a single analyte or a pair of structurally related molecules (e.g. phenol and pC), with more modern applications of HPLC allowing simultaneous measurement of phenolic (phenol, pC and pCS) and indolic (IS and indole-3-acetic acid) compounds [81]. Similarly to the TMAO story, the desire to understand gut microbial metabolism in a broader scope has driven the translation of analysis to hyphenated MS techniques to perform multiplexed approaches to UT measurements. Initial uptake of MS-based methods included the analysis of pC, pCS and p-cresol glucuronide by GC-MS [82], with measurements being more commonly performed using S/MRM assays employing LC-MS/MS. Similarly to HPLC-based assays, LC-MS/MS methods have been developed to specifically measure a single or pair of analytes [83], [84], [85], [86] or provide a wider focus on gut-derived UTs [87], as well as more complex multiplexed methods to assess additional metabolites outside of the indole and phenol classes (e.g. uric acid, TMAO, hippuric acid, kynurenic acid) [88], [89]. These analyses primarily focus on the measurement of UTs within the systemic circulation (i.e. serum, plasma), owing to the reduced ability to filter out into the urine. This has been demonstrated by the observation that less than 35% of circulating IS/pCS reduction occurs during hemodialysis [90], and that kidney transplant patients exhibit reduced circulating levels of pCS and IS that are comparable to healthy individuals [91], [92]. However, additional MS-based methods have been developed that are capable of measuring IS and pCS in saliva [93], as well as the presence of the IS precursor indole in exhaled breath gases [94], [95], [96]. However to date, the links between less traditional sample mediums and blood concentrations have yet to be explored in diseased populations where circulating UTs are of particular interest.

Renal disease has understandably received the major interest for clinical measurement and monitoring of UTs. This is due to the presence of an increased UT blood content in line with a progressive decline in renal function, as indicated by step increases in circulating levels of IS and pCS across CKD stages 1 to 5 [86]. Aside from renal disease, increased levels of IS have also been associated with more severe diastolic cardiac dysfunction (measured by E/e’), although circulating levels did not correlate with measurements of systolic dysfunction (left ventricular ejection fraction) nor with the established heart disease biomarker B-type natriuretic peptide [97]. As prognostic markers of renal disease IS and pCS have been able to demonstrate risk predictive qualities for renal decline [98], cardiovascular events [98], [99], [100], [101], [102], [103], [104] and mortality [100], [101], [102], [105], [106], [107] in CKD and hemodialysis patients. However, conflicting results are present in some investigations that demonstrate only one of the metabolites to be prognostic, or for prediction across event type (e.g. hospitalization, death, etc.) to be variable. For example, in hemodialysis studies IS has been reported as an independent predictor of outcomes where pCS was not [107], and vice-versa where pCS was predictive in place of IS [102]. These trends were reinforced in a recent meta-analysis of 11 publications investigating UTs as prognostic biomarkers in CKD. The authors observed that pCS was retained as an independent predictor of all-cause mortality and cardiovascular events, where IS was solely capable of independently predicting all-cause mortality [108]. These contrasting results provide a level of uncertainty for the true quality of clinical laboratory measurements of gut microbial-derived UTs, a point that was further reinforced by a recent large-scale study that showed that neither IS nor pCS were prognostic in hemodialysis patients [109]. Outside of renal disease, IS and pCS have been less well studied. However, initial trials have demonstrated potential utility as prognostic biomarkers in dilated cardiomyopathy [97] and heart failure [110], representing cardiovascular disease as an area that could benefit from further research into circulating UTs.

Altogether, in-depth multiplexed assays offer an excellent opportunity to more adequately understand the dynamics of UT production and build-up, with the capacity to measure multiple metabolites outweighing the downfalls where a single metabolite measurement has not shown to be reliable across patient cohorts. Current data showcase the beneficial prospects for implementation of UT measurements into routine clinical laboratory-based MS services for use in prognostic and therapeutic monitoring in conjunction with current biomarker measurements.

Short-chain fatty acids

Short-chain fatty acids (SCFAs) are volatile aliphatic carboxylic acids with a low carbon number (containing no more than five carbon atoms) and are common metabolic by-products of bacterial metabolism [111]. A list of SCFAs and their chemical information is provided in Table 2. Much like TMA, concentrated SCFAs present with an unpleasant odor and are commonly produced by bacteria on human body surfaces and thus are often described as redolent of ‘cheesy feet’ [112]. SCFAs produced by bacteria within the gastrointestinal system are metabolic by-products of non-digestible fiber fermentation, producing strait chain species (e.g. acetic, propionic, butyric acid), or protein fermentation creating branch-chained species including isobutyric, 2-methylbutyric and isovaleric acid specifically from the branch-chained amino acids valine, isoleucine and leucine [111]. Like other bacterially-produced metabolites, SCFAs can cross the gut epithelial barrier to enter the systemic circulation, albeit a large proportion remain within the gut ecosystem and are excreted within the feces [113], [114]. As SCFAs are carboxylic acids they exist in solution as dissociated forms and are therefore regularly referred to by their carboxylate anion, with acetate (C2), propionate (C3) and butyrate (C4) the principal components of total SCFAs present [115] (see Table 2 for more information on these anions).

Table 2:

A list of short-chain fatty acids containing between one and five carbon atoms.

Carbon numberNomenclatureAnionChemical structure
CommonIUPACCommonIUPAC
1Formic acidMethanoic acidFormateMethanoate
2Acetic acidEthanoic acidAcetateEthanoate
3Propionic acidPropanoic acidPropionatePropanoate
4Butyric acidButanoic acidButyrateButanoate
4Isobutyric acid2-Methylpropanoic acidIsobutyrate2-Methlypropanoate
5Valeric acidPentanoic acidValeratePentanoate
5Isovaleric acid3-Methylbutanoic acidIsovalerate3-Methylbutanoate
52-Methylbutyric acid2-Methylbutanoic acid2-Methylbutyrate2-Methylbutanoate
  1. IUPAC, International Union of Pure and Applied Chemistry: a standardized system for the naming of chemical compounds; Anion, the conjugate base of the acid that is present when in solution (i.e. in a biofluid).

The measurement of SCFAs using MS has been of interest for multiple decades, with early reports of GC/GC-MS assays as far back as the 1970s [116], [117]. More recently, there has been a flourish of published protocols, owing predominantly to the increasing interest of the impact of SCFAs in physiology and, importantly, pathophysiology. The majority of methods utilize GC-MS analysis of SCFAs derivatized with a range of different adducts and are principally applied to fecal collections [118], [119], [120]. Clinically, the measurement of fecal content of SCFAs may provide information on current or dynamic status of gut microbial metabolism, but it is not possible to provide a more accurate insight into the absorption of SCFAs into the systemic circulation, and therefore a potential impact on host cellular/tissue activity. A small but increasing number of protocols are emerging in the literature which assess circulating SCFA content, with methods including the traditional derivatized GC-MS approach [120], [121], [122], but also with further application of LC-MS systems [123], [124], [125]. Whilst derivatization of acidic metabolites generally offers greatly improved sensitivity and chromatographic reproducibility, the ability to exclude derivatization steps is of benefit to the clinical laboratory as it decreases the handling of hazardous chemicals whilst simultaneously increasing throughput and capacity. To this end, researchers have successfully developed assays that require only basic sample preparation prior to analysis. This has been afforded by use of wax-based GC columns (suitable for retention of acidic compounds) [118] as well as post-LC neutralization with ammonia, causing ammonia-adduct SCFA ions to form which can be easily measured using ESI-MS [126]. Interestingly, non-invasive sampling of SCFA levels has been proposed through the absorption of SCFAs excreted onto the surface of the skin [127]. This approach applies a chemically absorbent patch onto the skin which is later measured by MS following thermal desorption of molecules off the patch and into the analytical system. While this could provide a method to non-invasively measure circulating SCFAs in a clinical context, there are currently no data available to demonstrate the comparability of these measurements to direct quantitation in more routinely collected clinical sample types (i.e. blood and urine).

Contrary to the growing evidence of the negative impact of gut microbial metabolites on health and disease, there is an overwhelming indication that SCFAs offer protective effects. These beneficial aspects of gut metabolism have been repeatedly shown for in vitro and in vivo investigations, however, the overall impact of changes in SCFA levels in human participants are not currently well known. A strong focus of the research to date has been performed in models of renal disease, notably with the development and progression of CKD. Multiple studies have shown an interaction of SCFAs with G-protein coupled receptors on the cell surface that have shown beneficial modulations on blood pressure, inflammation, induced-tissue damage and overall immune response [114], [128], [129], [130], [131]. In particular for CKD, butyrate supplementation has demonstrated a blunting of fibrotic activity through the amelioration of TGF-β1 production in renal epithelial cells [132], [133] and via moderation of pro-inflammatory and pro-oxidative stress pathways [133], [134]. In addition to butyrate, protective effects of SCFA supplementation have been demonstrated for acetate and propionate, with suppression of TNF-α stimulated MCP-1 expression [135] and tempering of histone deacetylase activity in T cells following sepsis-induced kidney injury [136]. An in vivo model of ischemia-reperfusion of the kidneys demonstrated that supplementation of SCFAs could reduce indices of renal injury, helping to maintain renal function at a level that was comparable to pre-ischemia and reduce necrotic tissue formation by approximately 50% [129]. Fascinatingly, these positive outcomes were also achieved by supplementing mice with bacteria known to be acetate-producers, demonstrating a probiotic-like pre-treatment strategy that could reduce complications where ischemic kidney injury is common (e.g. cardiac surgery). Outside of renal disease, further beneficial effects of SCFA supplementation has been demonstrated for cardiovascular risk factors such as blood glucose homeostasis [137], reductions in atherogenic potential [138], [139], and amelioration of cardiac fibrosis and left ventricular hypertrophy in a murine model of hypertension [140]. Contrastingly, Tirosh et al. [141] demonstrated that supplementation of mice with propionate initiated an increased production of glucagon, fatty acid-binding protein 4 and norepinephrine which led to increased insulin resistance and compensatory hyperinsulinemia. This was mirrored, to some extent, in human participants who showed a reduction in circulating propionate levels that correlated with improved insulin sensitivity and overall weight loss. Whilst some investigations have started to look at the associations of circulating SCFAs with indicators of disease, few studies have looked at the use of SCFA profiling for application as diagnostic or prognostic biomarkers. One study reported that elevated plasma levels of valerate and propionate were independent predictors of coronary artery disease (CAD) in CKD patients, however, the absolute quantitative differences between those with and without CAD for these SCFAs were only around +1 μmol/L with standard deviations for both >1 μmol/L [142]. Whilst statistically these data may have shown differences, in a practical sense this separation of values between positive and negative CAD presence is far too narrow to be reliable as a clinical test. A note of interest in this study which was not emphasized by the authors was that decreased acetate levels (approximately −10 μmol/L but again with large standard deviations) showed an increased likelihood of self-reported CAD, stroke, peripheral arterial disease, congestive heart failure, or arrhythmia. Although these data are too premature to assign confidence, these results serve as preliminary evidence that increased acetate levels may be associated with reduced disease risk. For the use of SCFAs in prediction of outcomes in hospitalized patients, Weng et al. [143] observed that propionate levels were increased in sepsis patients and further elevated in patients with septic shock. Furthermore, elevated propionate was an independent predictor of mortality both in-hospital (during ICU stay) and at 28- and 90-days post-diagnosis in these patients.

Unlike TMAO and uremic toxins, accumulating evidence shows that SCFAs are beneficial in protecting from disease progression and could therefore provide a targeted therapeutic approach to improve patient outcomes. In the small number of studies performed to date on clinical samples, there is contrasting evidence that shows both increased and decreased circulating SCFAs levels could be predictive of negative outcomes and therefore it is important that further research is done in this area to include investigations to look across a range of diseases. It is apparent that the use of circulating SCFA measurements are well away from clinical translation at this stage, but accelerated research in this area could provide an interesting avenue for future clinical biomarker applications.

Outlook

MS offers numerous opportunities to improve and expand current clinical biomarker measurements, with the measurement of gut bacteria-mediated metabolites an excellent example of this future potential. The metabolites discussed in this review are by no means an exhaustive list, but act to highlight a growing interest in gut microbiome metabolites as potential clinical targets to provide an excellent resource to improve patient care, diagnosis, prognosis and therapeutic interventions/monitoring. The translation and uptake of these measurements would be facilitated through the acceleration of MS instrumentation into routine clinical laboratories, alongside the continued specialist training of laboratory scientists within this field. This focus, coupled with a continued effort in basic research of novel and potentially informative biomarkers, will allow the enhanced understanding of gut microbial metabolism to benefit patient care in the era of personalized medicine.

  1. Author contributions: The author has accepted responsibility for the entire content of this submitted manuscript and approved submission.

  2. Research funding: None declared.

  3. Employment or leadership: None declared.

  4. Honorarium: None declared.

  5. Competing interests: None declared.

  6. Ethical approval: The conducted research is a review of the literature and therefore not related to novel experiments performed using human participants or animal models.

References

1. Sender R, Fuchs S, Milo R. Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell 2016;164:337–40.10.1016/j.cell.2016.01.013Suche in Google Scholar PubMed

2. Sender R, Fuchs S, Milo R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol 2016;14: 1–14.10.1371/journal.pbio.1002533Suche in Google Scholar PubMed PubMed Central

3. Spor A, Koren O, Ley R. Unravelling the effects of the environment and host genotype on the gut microbiome. Nat Rev Microbiol 2011;9:279–90.10.1038/nrmicro2540Suche in Google Scholar PubMed

4. Faner R, Sibila O, Agustí A, Bernasconi E, Chalmers JD, HuffnagleGB, et al. The microbiome in respiratory medicine: current challenges and future perspectives. Eur Respir J 2017;49:1602086.10.1183/13993003.02086-2016Suche in Google Scholar PubMed

5. Lynch SV, Pedersen O. The human intestinal microbiome in health and disease. N Engl J Med 2016;375:2369–79.10.1056/NEJMra1600266Suche in Google Scholar PubMed

6. Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, JiaW, et al. Host-gut microbiota metabolic interactions. Science 2012;336:1262–7.10.1126/science.1223813Suche in Google Scholar PubMed

7. Brown JM, Hazen SL. Microbial modulation of cardiovascular disease. Nat Rev Microbiol 2018;16:171–81.10.1038/nrmicro.2017.149Suche in Google Scholar PubMed PubMed Central

8. Hobby GP, Karaduta O, Dusio GF, Singh M, Zybailov BL, Arthur JM. Chronic kidney disease and the gut microbiome. Am J Physiol Renal Physiol 2019;316:F1211–7.10.1152/ajprenal.00298.2018Suche in Google Scholar PubMed PubMed Central

9. Canfora EE, Meex RC, Venema K, Blaak EE. Gut microbial metabolites in obesity, NAFLD and T2DM. Nat Rev Endocrinol 2019;15:261–73.10.1038/s41574-019-0156-zSuche in Google Scholar PubMed

10. Halfvarson J, Brislawn CJ, Lamendella R, Vázquez-Baeza Y, Walters WA, Bramer LM, et al. Dynamics of the human gut microbiome in inflammatory bowel disease. Nat Microbiol 2017;2:17004.10.1038/nmicrobiol.2017.4Suche in Google Scholar PubMed PubMed Central

11. Marchesi JR, Adams DH, Fava F, Hermes GD, Hirschfield GM, Hold G, et al. The gut microbiota and host health: a new clinical frontier. Gut 2016;65:330–9.10.1136/gutjnl-2015-309990Suche in Google Scholar PubMed PubMed Central

12. Siuzdak G. An introduction to mass spectrometry ionization: an excerpt from The Expanding Role of Mass Spectrometry in Biotechnology, 2nd ed. San Diego, CA: MCC Press, 2005. J Assoc Lab Autom 2004;9:50–63.10.1016/j.jala.2004.01.004Suche in Google Scholar

13. Jannetto PJ, Fitzgerald RL. Effective use of mass spectrometry in the clinical laboratory. Clin Chem 2016;62:92–8.10.1373/clinchem.2015.248146Suche in Google Scholar PubMed

14. Minkler PE, Stoll MS, Ingalls ST, Kerner J, Hoppel CL. Validated method for the quantification of free and total carnitine, butyrobetaine, and acylcarnitines in biological samples. Anal Chem 2015;87:8994–9001.10.1021/acs.analchem.5b02198Suche in Google Scholar PubMed

15. Lau C-H, Siskos AP, Maitre L, Robinson O, Athersuch TJ, WantEJ, et al. Determinants of the urinary and serum metabolome in children from six European populations. BMC Med 2018;16:202.10.1186/s12916-018-1190-8Suche in Google Scholar PubMed PubMed Central

16. Schiffer L, Adaway JE, Arlt W, Keevil BG. A liquid chromatography-tandem mass spectrometry assay for the profiling of classical and 11-oxygenated androgens in saliva. Ann Clin Biochem 2019;56:564–73.10.1177/0004563219847498Suche in Google Scholar PubMed

17. Heaney LM, Jones DJ, Suzuki T. Mass spectrometry in medicine: a technology for the future? [Editorial] Future Sci OA 2017;3:FSO213.10.4155/fsoa-2017-0053Suche in Google Scholar PubMed PubMed Central

18. Vogeser M, Schuster C, Rockwood AL. A proposal to standardize the description of LC–MS-based measurement methods in laboratory medicine. Clin Mass Spectrom 2019;13:36–8 [Editorial].10.1016/j.clinms.2019.04.003Suche in Google Scholar PubMed PubMed Central

19. Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011;472:57–65.10.1038/nature09922Suche in Google Scholar PubMed PubMed Central

20. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, et al. Intestinal microbiota metabolism of ʟ-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med 2013;19:576–85.10.1038/nm.3145Suche in Google Scholar PubMed PubMed Central

21. Bennett BJ, Vallim TQ, Wang Z, Shih DM, Meng Y, Gregory J, et al. Trimethylamine-N-oxide, a metabolite associated with atherosclerosis, exhibits complex genetic and dietary regulation. Cell Metab 2013;17:49–60.10.1016/j.cmet.2012.12.011Suche in Google Scholar PubMed PubMed Central

22. Fennema D, Phillips IR, Shephard EA. Trimethylamine and trimethylamine N-oxide, a flavin-containing monooxygenase 3 (FMO3)-mediated host-microbiome metabolic axis implicated in health and disease. Drug Metab Dispos 2016;44:1839–50.10.1124/dmd.116.070615Suche in Google Scholar PubMed PubMed Central

23. Mohri S, Kanauchi M. Isolation of lactic acid bacteria eliminating trimethylamine (TMA) for application to fishery processing. Methods Mol Biol 2019;1887:109–17.10.1007/978-1-4939-8907-2_10Suche in Google Scholar PubMed

24. Mackay RJ, McEntyre CJ, Henderson C, Lever M, George PM. Trimethylaminuria: causes and diagnosis of a socially distressing condition. Clin Biochem Rev 2011;32:33–43.Suche in Google Scholar

25. Dunn WB, Broadhurst D, Begley P, Zelena E, Francis-Mcintyre S, Anderson N, et al. Procedures for large-scale metabolic profiling of serum and plasma using gas chromatography and liquid chromatography coupled to mass spectrometry. Nat Protoc 2011;6:1060–83.10.1038/nprot.2011.335Suche in Google Scholar PubMed

26. Heaney LM, Deighton K, Suzuki T. Non-targeted metabolomics in sport and exercise science. J Sports Sci 2019;37:959–67.10.1080/02640414.2017.1305122Suche in Google Scholar PubMed

27. Tang WH, Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med 2013;368:1575–84.10.1056/NEJMoa1109400Suche in Google Scholar PubMed PubMed Central

28. Lenky CC, McEntyre CJ, Lever M. Measurement of marine osmolytes in mammalian serum by liquid chromatography–tandem mass spectrometry. Anal Biochem 2012;420:7–12.10.1016/j.ab.2011.09.013Suche in Google Scholar PubMed

29. Wang Z, Levison BS, Hazen JE, Donahue L, Li X-M, Hazen SL. Measurement of trimethylamine-N-oxide by stable isotope dilution liquid chromatography tandem mass spectrometry. Anal Biochem 2014;455:35–40.10.1016/j.ab.2014.03.016Suche in Google Scholar PubMed PubMed Central

30. Klepacki J, Klawitter J, Klawitter J, Thurman JM, Christians U. A high-performance liquid chromatography-tandem mass spectrometry-based targeted metabolomics kidney dysfunction marker panel in human urine. Clin Chim Acta 2015;446:43–53.10.1016/j.cca.2015.04.005Suche in Google Scholar PubMed PubMed Central

31. Le TT, Shafaei A, Genoni A, Christophersen C, Devine A, Lo J, et al. Development and validation of a simple LC-MS/MS method for the simultaneous quantitative determination of trimethylamine-N-oxide and branched chain amino acids in human serum. Anal Bioanal Chem 2019;411:1019–28.10.1007/s00216-018-1522-8Suche in Google Scholar PubMed

32. Mamer OA, Choinière L, Lesimple A. Measurement of urinary trimethylamine and trimethylamime oxide by direct infusion electrospray quadrupole time-of-flight mass spectrometry. Anal Biochem 2010;406:80–2.10.1016/j.ab.2010.06.038Suche in Google Scholar

33. Kadar H, Dubus J, Dutot J, Hedjazi L, Srinivasa S, Fitch KV, et al. A multiplexed targeted assay for high-throughput quantitative analysis of serum methylamines by ultra performance liquid chromatography coupled to high resolution mass spectrometry. Arch Biochem Biophys 2016;597:12–20.10.1016/j.abb.2016.03.029Suche in Google Scholar

34. Heaney LM, Jones DJ, Mbasu RJ, Ng LL, Suzuki T. High mass accuracy assay for trimethylamine N-oxide using stable-isotope dilution with liquid chromatography coupled to orthogonal acceleration time of flight mass spectrometry with multiple reaction monitoring. Anal Bioanal Chem 2016;408:797–804.10.1007/s00216-015-9164-6Suche in Google Scholar

35. daCosta KA, Vrbanac JJ, Zeisel SH. The measurement of dimethylamine, trimethylamine, and trimethylamine N-oxide using capillary gas chromatography-mass spectrometry. Anal Biochem 1990;187:234–9.10.1016/0003-2697(90)90449-JSuche in Google Scholar

36. Fiori J, Turroni S, Candela M, Brigidi P, Gotti R. Simultaneous HS-SPME GC-MS determination of short chain fatty acids, trimethylamine and trimethylamine N-oxide for gut microbiota metabolic profile. Talanta 2018;189:573–8.10.1016/j.talanta.2018.07.051Suche in Google Scholar PubMed

37. Garcia E, Wolak-Dinsmore J, Wang Z, Li XS, Bennett DW, Connelly MA, et al. NMR quantification of trimethylamine-N-oxide in human serum and plasma in the clinical laboratory setting. Clin Biochem 2017;50:947–55.10.1016/j.clinbiochem.2017.06.003Suche in Google Scholar PubMed PubMed Central

38. Ocque AJ, Stubbs JR, Nolin TD. Development and validation of a simple UHPLC–MS/MS method for the simultaneous determination of trimethylamine N-oxide, choline, and betaine in human plasma and urine. J Pharm Biomed Anal 2015;109:128–35.10.1016/j.jpba.2015.02.040Suche in Google Scholar PubMed

39. Zhao X, Zeisel SH, Zhang S. Rapid LC-MRM-MS assay for simultaneous quantification of choline, betaine, trimethylamine, trimethylamine N-oxide, and creatinine in human plasma and urine. Electrophoresis 2015;36:2207–14.10.1002/elps.201500055Suche in Google Scholar PubMed

40. Steuer C, Schütz P, Bernasconi L, Huber AR. Simultaneous determination of phosphatidylcholine-derived quaternary ammonium compounds by a LC–MS/MS method in human blood plasma, serum and urine samples. J Chromatogr B 2016;1008:206–11.10.1016/j.jchromb.2015.12.002Suche in Google Scholar PubMed

41. Tang WH, Wang Z, Kennedy DJ, Wu Y, Buffa JA, Agatisa-Boyle B, et al. Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to both development of renal insufficiency and mortality risk in chronic kidney disease. Circ Res 2015;116:448–55.10.1161/CIRCRESAHA.116.305360Suche in Google Scholar PubMed PubMed Central

42. Zhu W, Gregory JC, Org E, Buffa JA, Gupta N, Wang Z, et al. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell 2016;165:111–24.10.1016/j.cell.2016.02.011Suche in Google Scholar PubMed PubMed Central

43. Organ CL, Otsuka H, Bhushan S, Wang Z, Bradley J, TrivediR, et al. Choline diet and its gut microbe-derived metabolite, trimethylamine N-oxide, exacerbate pressure overload-induced heart failure. Circ Heart Fail 2016;9:e002314.10.1161/CIRCHEARTFAILURE.115.002314Suche in Google Scholar PubMed PubMed Central

44. Boini KM, Hussain T, Li P-L, Koka SS. Trimethylamine-N-oxide instigates NLRP3 inflammasome activation and endothelial dysfunction. Cell Physiol Biochem 2017;44:152–62.10.1159/000484623Suche in Google Scholar PubMed PubMed Central

45. Tang WH, Wang Z, Fan Y, Levison B, Hazen JE, DonahueLM, et al. Prognostic value of elevated levels of intestinal microbe-generated metabolite trimethylamine-N-oxide in patients with heart failure: refining the gut hypothesis. J Am Coll Cardiol 2014;64:1908–14.10.1016/j.jacc.2014.02.617Suche in Google Scholar PubMed PubMed Central

46. Tang WH, Wang Z, Shrestha K, Borowski AG, Wu Y, TroughtonRW, et al. Intestinal microbiota-dependent phosphatidylcholine metabolites, diastolic dysfunction, and adverse clinical outcomes in chronic systolic heart failure. J Card Fail 2015;21:91–6.10.1016/j.cardfail.2014.11.006Suche in Google Scholar PubMed PubMed Central

47. Trøseid M, Ueland T, Hov JR, Svardal A, Gregersen I, Dahl CP, et al. Microbiota-dependent metabolite trimethylamine-N-oxide is associated with disease severity and survival of patients with chronic heart failure. J Intern Med 2015;277:717–26.10.1111/joim.12328Suche in Google Scholar PubMed

48. Suzuki T, Heaney LM, Bhandari SS, Jones DJ, Ng LL. Trimethylamine N-oxide and prognosis in acute heart failure. Heart 2016;102:841–8.10.1136/heartjnl-2015-308826Suche in Google Scholar PubMed

49. Li XS, Obeid S, Klingenberg R, Gencer B, Mach F, Räber L, et al. Gut microbiota-dependent trimethylamine N-oxide in acute coronary syndromes: a prognostic marker for incident cardiovascular events beyond traditional risk factors. Eur Heart J 2017;38:814–24.10.1093/eurheartj/ehw582Suche in Google Scholar PubMed PubMed Central

50. Suzuki T, Heaney LM, Jones DJ, Ng LL. Trimethylamine N-oxide and risk stratification after acute myocardial infarction. Clin Chem 2017;63:420–8.10.1373/clinchem.2016.264853Suche in Google Scholar PubMed

51. Missailidis C, Hällqvist J, Qureshi AR, Barany P, Heimbürger O, Lindholm B, et al. Serum trimethylamine-N-Oxide is strongly related to renal function and predicts outcome in chronic kidney disease. PLoS One 2016;11:1–14.10.1371/journal.pone.0141738Suche in Google Scholar PubMed PubMed Central

52. Nie J, Xie L, Zhao B, Li Y, Qiu B, Zhu F, et al. Serum trimethylamine N-oxide concentration is positively associated with first stroke in hypertensive patients. Stroke 2018;49:2021–8.10.1161/STROKEAHA.118.021997Suche in Google Scholar PubMed

53. Haghikia A, Li XS, Liman TG, Bledau N, Schmidt D, Zimmermann F, et al. Gut microbiota–dependent trimethylamine N-oxide predicts risk of cardiovascular events in patients with stroke and is related to proinflammatory monocytes. Arterioscler Thromb Vasc Biol 2018;38:2225–35.10.1161/ATVBAHA.118.311023Suche in Google Scholar PubMed PubMed Central

54. Kummen M, Vesterhus M, Trøseid M, Moum B, Svardal A, Boberg KM, et al. Elevated trimethylamine-N-oxide (TMAO) is associated with poor prognosis in primary sclerosing cholangitis patients with normal liver function. United Eur Gastroenterol J 2017;5:532–41.10.1177/2050640616663453Suche in Google Scholar PubMed PubMed Central

55. Barrea L, Annunziata G, Muscogiuri G, Di Somma C, Laudisio D, Maisto M, et al. Trimethylamine-N-oxide (TMAO) as novel potential biomarker of early predictors of metabolic syndrome. Nutrients 2018;10:1971.10.3390/nu10121971Suche in Google Scholar PubMed PubMed Central

56. Liu X, Liu H, Yuan C, Zhang Y, Wang W, Hu S, et al. Preoperative serum TMAO level is a new prognostic marker for colorectal cancer. Biomark Med 2017;11:443–7.10.2217/bmm-2016-0262Suche in Google Scholar PubMed

57. Ottiger M, Nickler M, Steuer C, Odermatt J, Huber A, Christ-Crain M, et al. Trimethylamine-N-oxide (TMAO) predicts fatal outcomes in community-acquired pneumonia patients without evident coronary artery disease. Eur J Intern Med 2016;36:67–73.10.1016/j.ejim.2016.08.017Suche in Google Scholar PubMed

58. Vanholder R, De Smet R, Glorieux G, Argilés A, Baurmeister U, Brunet P, et al. Review on uremic toxins: Classification, concentration, and interindividual variability. Kidney Int 2003;63:1934–43.10.1046/j.1523-1755.2003.00924.xSuche in Google Scholar PubMed

59. Winchester JF, Audia PF. extracorporeal strategies for the removal of middle molecules. Semin Dial 2006;19:110–4.10.1111/j.1525-139X.2006.00135.xSuche in Google Scholar PubMed

60. Duranton F, Cohen G, De Smet R, Rodriguez M, Jankowski J, Vanholder R, et al. Normal and pathologic concentrations of uremic toxins. J Am Soc Nephrol 2012;23:1258–70.10.1681/ASN.2011121175Suche in Google Scholar PubMed PubMed Central

61. Glassock RJ. Uremic toxins: what are they? An integrated overview of pathobiology and classification. J Ren Nutr 2008;18:2–6.10.1053/j.jrn.2007.10.003Suche in Google Scholar PubMed

62. Zhang LS, Davies SS. Microbial metabolism of dietary components to bioactive metabolites: opportunities for new therapeutic interventions. Genome Med 2016;8:46.10.1186/s13073-016-0296-xSuche in Google Scholar PubMed PubMed Central

63. Karbowska M, Kaminski T, Marcinczyk N, Misztal T, Rusak T, Smyk L, et al. The uremic toxin indoxyl sulfate accelerates thrombotic response after vascular injury in animal models. Toxins 2017;9:229.10.3390/toxins9070229Suche in Google Scholar PubMed PubMed Central

64. Karbowska M, Kaminski TW, Znorko B, Domaniewski T, Misztal T, Rusak T, et al. Indoxyl sulfate promotes arterial thrombosis in rat model via increased levels of complex TF/VII, PAI-1, platelet activation as well as decreased contents of SIRT1 and SIRT3. Front Physiol 2018;9:1623.10.3389/fphys.2018.01623Suche in Google Scholar PubMed PubMed Central

65. Jing YJ, Ni JW, Ding FH, Fang YH, Wang XQ, Wang HB, et al. p-Cresyl sulfate is associated with carotid arteriosclerosis in hemodialysis patients and promotes atherogenesis in apoE−/− mice. Kidney Int 2016;89:439–49.10.1038/ki.2015.287Suche in Google Scholar PubMed

66. Huc T, Konop M, Onyszkiewicz M, Podsadni P, Szczepańska A, Turło J, et al. Colonic indole, gut bacteria metabolite of tryptophan, increases portal blood pressure in rats. Am J Physiol Integr Comp Physiol 2018;315:R646–55.10.1152/ajpregu.00111.2018Suche in Google Scholar PubMed

67. Huć T, Nowinski A, Drapala A, Konopelski P, Ufnal M. Indole and indoxyl sulfate, gut bacteria metabolites of tryptophan, change arterial blood pressure via peripheral and central mechanisms in rats. Pharmacol Res 2018;130:172–9.10.1016/j.phrs.2017.12.025Suche in Google Scholar PubMed

68. Opdebeeck B, Maudsley S, Azmi A, De Maré A, De Leger W, Meijers B, et al. Indoxyl sulfate and p-Cresyl sulfate promote vascular calcification and associate with glucose intolerance. J Am Soc Nephrol 2019;30:751–66.10.1681/ASN.2018060609Suche in Google Scholar PubMed PubMed Central

69. Yisireyili M, Shimizu H, Saito S, Enomoto A, Nishijima F, Niwa T. Indoxyl sulfate promotes cardiac fibrosis with enhanced oxidative stress in hypertensive rats. Life Sci 2013;92:1180–5.10.1016/j.lfs.2013.05.008Suche in Google Scholar PubMed

70. Hung S-C, Kuo K-L, Huang H-L, Lin C-C, Tsai T-H, Wang C-H, et al. Indoxyl sulfate suppresses endothelial progenitor cell-mediated neovascularization. Kidney Int 2016;89:574–85.10.1016/j.kint.2015.11.020Suche in Google Scholar PubMed

71. Watanabe H, Miyamoto Y, Honda D, Tanaka H, Wu Q, Endo M, et al. p-Cresyl sulfate causes renal tubular cell damage by inducing oxidative stress by activation of NADPH oxidase. Kidney Int 2013;83:582–92.10.1038/ki.2012.448Suche in Google Scholar PubMed

72. Sun C-Y, Hsu H-H, Wu M-S. p-Cresol sulfate and indoxyl sulfate induce similar cellular inflammatory gene expressions in cultured proximal renal tubular cells. Nephrol Dial Transplant 2013;28:70–8.10.1093/ndt/gfs133Suche in Google Scholar

73. Poveda J, Sanchez-Niño MD, Glorieux G, Sanz AB, Egido J, Vanholder R, et al. p-Cresyl sulphate has pro-inflammatory and cytotoxic actions on human proximal tubular epithelial cells. Nephrol Dial Transplant 2014;29:56–64.10.1093/ndt/gft367Suche in Google Scholar

74. Shimizu H, Bolati D, Adijiang A, Enomoto A, Nishijima F, Dateki M, et al. Senescence and dysfunction of proximal tubular cells are associated with activated p53 expression by indoxyl sulfate. Am J Physiol Cell Physiol 2010;299:C1110–7.10.1152/ajpcell.00217.2010Suche in Google Scholar

75. Niwa T, Shimizu H. Indoxyl sulfate induces nephrovascular senescence. J Ren Nutr 2012;22:102–6.10.1053/j.jrn.2011.10.032Suche in Google Scholar

76. Carmona A, Guerrero F, Buendia P, Obrero T, Aljama P, Carracedo J. Microvesicles derived from indoxyl sulfate treated endothelial cells induce endothelial progenitor cells dysfunction. Front Physiol 2017;8:666.10.3389/fphys.2017.00666Suche in Google Scholar

77. Stanfel LA, Gulyassy PF, Jarrard EA. Determination of indoxyl sulfate in plasma of patients with renal failure by use of ion-pairing liquid chromatography. Clin Chem 1986;32:938–42.10.1093/clinchem/32.6.938Suche in Google Scholar

78. Niwa T, Takeda N, Tatematsu A, Maeda K. Accumulation of indoxyl sulfate, an inhibitor of drug-binding, in uremic serum as demonstrated by internal-surface reversed-phase liquid chromatography. Clin Chem 1988;34:2264–7.10.1093/clinchem/34.11.2264Suche in Google Scholar

79. Niwa T. Phenol and p-cresol accumulated in uremic serum measured by HPLC with fluorescence detection. Clin Chem 1993;39:108–11.10.1093/clinchem/39.1.108Suche in Google Scholar

80. De Smet R, David F, Sandra P, Van Kaer J, Lesaffer G, Dhondt A, et al. A sensitive HPLC method for the quantification of free and total p-cresol in patients with chronic renal failure. Clin Chim Acta 1998;278:1–21.10.1016/S0009-8981(98)00124-7Suche in Google Scholar

81. Calaf R, Cerini C, Génovésio C, Verhaeghe P, Jourde-Chiche N, Bergé-Lefranc D, et al. Determination of uremic solutes in biological fluids of chronic kidney disease patients by HPLC assay. J Chromatogr B Anal Technol Biomed Life Sci 2011;879:2281–6.10.1016/j.jchromb.2011.06.014Suche in Google Scholar PubMed

82. de Loor H, Bammens B, Evenepoel P, De Preter V, Verbeke K. Gas chromatographic-mass spectrometric analysis for measurement of p-cresol and its conjugated metabolites in uremic and normal serum. Clin Chem 2005;51:1535–8.10.1373/clinchem.2005.050781Suche in Google Scholar PubMed

83. Cuoghi A, Caiazzo M, Bellei E, Monari E, Bergamini S, Palladino G, et al. Quantification of p-cresol sulphate in human plasma by selected reaction monitoring. Anal Bioanal Chem 2012;404:2097–104.10.1007/s00216-012-6277-zSuche in Google Scholar PubMed

84. Chang S, Xujiao C, Tianyi X, Feng Z, Shouhong G, Wansheng C. LC–MS/MS method for simultaneous determination of serum p-cresyl sulfate and indoxyl sulfate in patients undergoing peritoneal dialysis. Biomed Chromatogr 2016;30:1782–8.10.1002/bmc.3753Suche in Google Scholar PubMed

85. Korytowska N, Wyczałkowska-Tomasik A, Wiśniewska A, Pączek L, Giebułtowicz J. Development of the LC-MS/MS method for determining the p-cresol level in plasma. J Pharm Biomed Anal 2019;167:149–54.10.1016/j.jpba.2019.01.041Suche in Google Scholar PubMed

86. Lin C-N, Wu I-W, Huang Y-F, Peng S-Y, Huang Y-C, Ning H-C. Measuring serum total and free indoxyl sulfate and p-cresyl sulfate in chronic kidney disease using UPLC-MS/MS. J Food Drug Anal 2019;27:502–9.10.1016/j.jfda.2018.10.008Suche in Google Scholar PubMed

87. Prokopienko AJ, West RE, Stubbs JR, Nolin TD. Development and validation of a UHPLC-MS/MS method for measurement of a gut-derived uremic toxin panel in human serum: an application in patients with kidney disease. J Pharm Biomed Anal 2019;174:618–24.10.1016/j.jpba.2019.06.033Suche in Google Scholar PubMed PubMed Central

88. Boelaert J, Lynen F, Glorieux G, Eloot S, Van Landschoot M, Waterloos M-A, et al. A novel UPLC–MS–MS method for simultaneous determination of seven uremic retention toxins with cardiovascular relevance in chronic kidney disease patients. Anal Bioanal Chem 2013;405:1937–47.10.1007/s00216-012-6636-9Suche in Google Scholar PubMed

89. de Loor H, Poesen R, De Leger W, Dehaen W, Augustijns P, Evenepoel P, et al. A liquid chromatography-tandem mass spectrometry method to measure a selected panel of uremic retention solutes derived from endogenous and colonic microbial metabolism. Anal Chim Acta 2016;936:149–56.10.1016/j.aca.2016.06.057Suche in Google Scholar PubMed

90. Itoh Y, Ezawa A, Kikuchi K, Tsuruta Y, Niwa T. Protein-bound uremic toxins in hemodialysis patients measured by liquid chromatography/tandem mass spectrometry and their effects on endothelial ROS production. Anal Bioanal Chem 2012;403:1841–50.10.1007/s00216-012-5929-3Suche in Google Scholar PubMed

91. Ligabue G, Damiano F, Cuoghi A, De Biasi S, Bellei E, Granito M, et al. P-cresol and cardiovascular risk in kidney transplant recipients. Transplant Proc 2015;47:2121–5.10.1016/j.transproceed.2015.02.033Suche in Google Scholar PubMed

92. Liabeuf S, Desjardins L, Massy ZA, Brazier F, Westeel PF, Mazouz H, et al. Levels of indoxyl sulfate in kidney transplant patients, and the relationship with hard outcomes. Circ J 2016;80:722–30.10.1253/circj.CJ-15-0949Suche in Google Scholar PubMed

93. Giebułtowicz J, Korytowska N, Sankowski B, Wroczyński P. Development and validation of a LC-MS/MS method for quantitative analysis of uraemic toxins p-cresol sulphate and indoxyl sulphate in saliva. Talanta 2016;150:593–8.10.1016/j.talanta.2015.12.075Suche in Google Scholar PubMed

94. Turner M, Guallar-Hoyas C, Kent A, Wilson I, Thomas C. Comparison of metabolomic profiles obtained using chemical ionization and electron ionization MS in exhaled breath. Bioanalysis 2011;3:2731–8.10.4155/bio.11.284Suche in Google Scholar PubMed

95. Martinez-Lozano Sinues P, Meier L, Berchtold C, Ivanov M, Sievi N, Camen G, et al. Breath analysis in real time by mass spectrometry in chronic obstructive pulmonary disease. Respiration 2014;87:301–10.10.1159/000357785Suche in Google Scholar PubMed

96. Jankowski J, Westhof T, Vaziri ND, Ingrosso D, Perna AF. Gases as uremic toxins: is there something in the air? Semin Nephrol 2014;34:135–50.10.1016/j.semnephrol.2014.02.006Suche in Google Scholar PubMed

97. Shimazu S, Hirashiki A, Okumura T, Yamada T, Okamoto R, Shinoda N, et al. Association between indoxyl sulfate and cardiac dysfunction and prognosis in patients with dilated cardiomyopathy. Circ J 2013;77:390–6.10.1253/circj.CJ-12-0715Suche in Google Scholar

98. Lin C-J, Liu H-L, Pan C-F, Chuang C-K, Jayakumar T, Wang T-J, et al. Indoxyl sulfate predicts cardiovascular disease and renal function deterioration in advanced chronic kidney disease. Arch Med Res 2012;43:451–6.10.1016/j.arcmed.2012.08.002Suche in Google Scholar PubMed

99. Meijers BK, Claes K, Bammens B, De Loor H, Viaene L, Verbeke K, et al. p-Cresol and cardiovascular risk in mild-to-moderate kidney disease. Clin J Am Soc Nephrol 2010;5:1182–9.10.2215/CJN.07971109Suche in Google Scholar PubMed PubMed Central

100. Lin C-J, Chuang C-K, Jayakumar T, Liu H-L, Pan C-F, Wang T-J, et al. Serum p-cresyl sulfate predicts cardiovascular disease and mortality in elderly hemodialysis patients. Arch Med Sci 2013;4:662–8.10.5114/aoms.2013.36901Suche in Google Scholar PubMed PubMed Central

101. Lin C, Pan C, Chuang C, Liu H, Sun F, Wang T, et al. Gastrointestinal-related uremic toxins in peritoneal dialysis: a pilot study with a 5-year follow-up. Arch Med Res 2013;44:535–41.10.1016/j.arcmed.2013.09.007Suche in Google Scholar PubMed

102. Shafi T, Meyer TW, Hostetter TH, Melamed ML, Parekh RS, Hwang S, et al. Free levels of selected organic solutes and cardiovascular morbidity and mortality in hemodialysis patients: results from the retained organic solutes and clinical outcomes (ROSCO) investigators.PLoS One 2015;10:e0126048.10.1371/journal.pone.0126048Suche in Google Scholar PubMed PubMed Central

103. Cao X-S, Chen J, Zou J-Z, Zhong Y-H, Teng J, Ji J, et al. Association of indoxyl sulfate with heart failure among patients on hemodialysis. Clin J Am Soc Nephrol 2015;10:111–9.10.2215/CJN.04730514Suche in Google Scholar PubMed PubMed Central

104. Fan P-C, Chang JC, Lin C-N, Lee C-C, Chen Y-T, Chu P-H, et al. Serum indoxyl sulfate predicts adverse cardiovascular events in patients with chronic kidney disease. J Formos Med Assoc 2019;118:1099–106.10.1016/j.jfma.2019.03.005Suche in Google Scholar PubMed

105. Wu IW, Hsu KH, Lee CC, Sun CY, Hsu HJ, Tsai CJ, et al. P-cresyl sulphate and indoxyl sulphate predict progression of chronic kidney disease. Nephrol Dial Transplant 2011;26:938–47.10.1093/ndt/gfq580Suche in Google Scholar PubMed PubMed Central

106. Wu I-W, Hsu K-H, Hsu H-J, Lee C-C, Sun C-Y, Tsai C-J, et al. Serum free p-cresyl sulfate levels predict cardiovascular and all-cause mortality in elderly hemodialysis patients – a prospective cohort study. Nephrol Dial Transplant 2012;27:1169–75.10.1093/ndt/gfr453Suche in Google Scholar PubMed

107. Melamed ML, Plantinga L, Shafi T, Parekh R, Meyer TW, Hostetter TH, et al. Retained organic solutes, patient characteristics and all-cause and cardiovascular mortality in hemodialysis: results from the retained organic solutes and clinical outcomes (ROSCO) investigators. BMC Nephrol 2013;14:134.10.1186/1471-2369-14-134Suche in Google Scholar PubMed PubMed Central

108. Lin C-J, Wu V, Wu P-C, Wu C-J. Meta-analysis of the associations of p-Cresyl sulfate (PCS) and indoxyl sulfate (IS) with cardiovascular events and all-cause mortality in patients with chronic renal failure.PLoS One 2015;10:e0132589.10.1371/journal.pone.0132589Suche in Google Scholar PubMed PubMed Central

109. Shafi T, Sirich TL, Meyer TW, Hostetter TH, Plummer NS, Hwang S, et al. Results of the HEMO Study suggest that p-cresol sulfate and indoxyl sulfate are not associated with cardiovascular outcomes. Kidney Int 2017;92:1484–92.10.1016/j.kint.2017.05.012Suche in Google Scholar PubMed PubMed Central

110. Wang C-H, Cheng M-L, Liu M-H, Shiao M-S, Hsu K-H, Huang Y-Y, et al. Increased p-cresyl sulfate level is independently associated with poor outcomes in patients with heart failure. Heart Vessels 2016;31:1100–8.10.1007/s00380-015-0702-0Suche in Google Scholar PubMed

111. Koh A, De Vadder F, Kovatcheva-Datchary P, Bäckhed F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell 2016;165:1332–45.10.1016/j.cell.2016.05.041Suche in Google Scholar PubMed

112. Stevens D, Cornmell R, Taylor D, Grimshaw SG, Riazanskaia S, Arnold DS, et al. Spatial variations in the microbial community structure and diversity of the human foot is associated with the production of odorous volatiles. FEMS Microbiol Ecol 2015;91:1–11.10.1093/femsec/fiu018Suche in Google Scholar

113. Høverstad T, Fausa O, Bjørneklett A, Bøhmer T. Short-chain fatty acids in the normal human feces. Scand J Gastroenterol 1984;19:375–81.10.1080/00365521.1984.12005738Suche in Google Scholar

114. Parada Venegas D, De la Fuente MK, Landskron G, González MJ, Quera R, Dijkstra G, et al. Short chain fatty acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front Immunol 2019;10:277.10.3389/fimmu.2019.00277Suche in Google Scholar

115. Heaney LM, Davies OG, Selby NM. Gut microbial metabolites as mediators of renal disease: do short-chain fatty acids offer some hope? Future Sci OA 2019;5:FSO384.10.4155/fsoa-2019-0013Suche in Google Scholar

116. Eldjarn L, Jellum E, Stokke O. Application of gas chromatography-mass spectrometry in routine and research in clinical chemistry. J Chromatogr A 1974;91:353–66.10.1016/S0021-9673(01)97914-2Suche in Google Scholar

117. Bachmann C, Colombo J-P, Berüter J. Short chain fatty acids in plasma and brain: Quantitative determination by gas chromatography. Clin Chim Acta 1979;92:153–9.10.1016/0009-8981(79)90109-8Suche in Google Scholar

118. Lotti C, Rubert J, Fava F, Tuohy K, Mattivi F, Vrhovsek U. Development of a fast and cost-effective gas chromatography–mass spectrometry method for the quantification of short-chain and medium-chain fatty acids in human biofluids. Anal Bioanal Chem 2017;409:5555–67.10.1007/s00216-017-0493-5Suche in Google Scholar PubMed

119. Cai J, Zhang J, Tian Y, Zhang L, Hatzakis E, Krausz KW, et al. Orthogonal comparison of GC–MS and 1H NMR spectroscopy for short chain fatty acid quantitation. Anal Chem 2017;89:7900–6.10.1021/acs.analchem.7b00848Suche in Google Scholar PubMed PubMed Central

120. Zhang S, Wang H, Zhu M-J. A sensitive GC/MS detection method for analyzing microbial metabolites short chain fatty acids in fecal and serum samples. Talanta 2019;196:249–54.10.1016/j.talanta.2018.12.049Suche in Google Scholar PubMed

121. Tsukahara T, Matsukawa N, Tomonaga S, Inoue R, Ushida K, Ochiai K. High-sensitivity detection of short-chain fatty acids in porcine ileal, cecal, portal and abdominal blood by gas chromatography-mass spectrometry. Anim Sci J 2014;85:494–8.10.1111/asj.12188Suche in Google Scholar PubMed

122. Hoving LR, Heijink M, van Harmelen V, van Dijk KW, Giera M. GC-MS analysis of short-chain fatty acids in feces, cecum content, and blood samples. Methods Mol Biol 2018;1730:247–56.10.1007/978-1-4939-7592-1_17Suche in Google Scholar PubMed

123. Zeng M, Cao H. Fast quantification of short chain fatty acids and ketone bodies by liquid chromatography-tandem mass spectrometry after facile derivatization coupled with liquid-liquid extraction. J Chromatogr B Anal Technol Biomed Life Sci 2018;1083:137–45.10.1016/j.jchromb.2018.02.040Suche in Google Scholar PubMed

124. Ma S-R, Tong Q, Zhao Z-X, Cong L, Yu J-B, Fu J, et al. Determination of berberine-upregulated endogenous short-chain fatty acids through derivatization by 2-bromoacetophenone. Anal Bioanal Chem 2019;411:3191–207.10.1007/s00216-019-01793-3Suche in Google Scholar PubMed

125. Jaochico A, Sangaraju D, Shahidi-Latham SK. A rapid derivatization based LC–MS/MS method for quantitation of short chain fatty acids in human plasma and urine. Bioanalysis 2019;11:741–53.10.4155/bio-2018-0241Suche in Google Scholar PubMed

126. van Eijk HM, Bloemen JG, Dejong CH. Application of liquid chromatography–mass spectrometry to measure short chain fatty acids in blood. J Chromatogr B 2009;877:719–24.10.1016/j.jchromb.2009.01.039Suche in Google Scholar PubMed

127. Martin HJ, Reynolds JC, Riazanskaia S, Thomas CL. High throughput volatile fatty acid skin metabolite profiling by thermal desorption secondary electrospray ionisation mass spectrometry. Analyst 2014;139:4279–86.10.1039/C4AN00134FSuche in Google Scholar PubMed

128. Pluznick JL, Protzko RJ, Gevorgyan H, Peterlin Z, Sipos A, Han J, et al. Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation. Proc Natl Acad Sci USA 2013;110:4410–5.10.1073/pnas.1215927110Suche in Google Scholar PubMed PubMed Central

129. Andrade-Oliveira V, Amano MT, Correa-Costa M, Castoldi A, Felizardo RJ, de Almeida DC, et al. Gut bacteria products prevent AKI induced by ischemia-reperfusion. J Am Soc Nephrol 2015;26:1877–88.10.1681/ASN.2014030288Suche in Google Scholar PubMed PubMed Central

130. Antunes KH, Fachi JL, de Paula R, da Silva EF, Pral LP, dos Santos AÁ, et al. Microbiota-derived acetate protects against respiratory syncytial virus infection through a GPR43-type 1 interferon response. Nat Commun 2019;10:3273.10.1038/s41467-019-11152-6Suche in Google Scholar PubMed PubMed Central

131. Qiu J, Villa M, Sanin DE, Buck MD, O’Sullivan D, Ching R, et al. Acetate promotes T cell effector function during glucose restriction. Cell Rep 2019;27:2063–74.10.1016/j.celrep.2019.04.022Suche in Google Scholar PubMed PubMed Central

132. Matsumoto N, Riley S, Fraser D, Al-Assaf S, Ishimura E, Wolever T, et al. Butyrate modulates TGF-β1 generation and function: potential renal benefit for Acacia(sen) SUPERGUM™ (gum arabic)? Kidney Int 2006;69:257–65.10.1038/sj.ki.5000028Suche in Google Scholar PubMed

133. Khan S, Jena G. Sodium butyrate, a HDAC inhibitor ameliorates eNOS, iNOS and TGF-β1-induced fibrogenesis, apoptosis and DNA damage in the kidney of juvenile diabetic rats. Food Chem Toxicol 2014;73:127–39.10.1016/j.fct.2014.08.010Suche in Google Scholar PubMed

134. MacHado RA, Constantino LD, Tomasi CD, Rojas HA, Vuolo FS, Vitto MF, et al. Sodium butyrate decreases the activation of NF-κB reducing inflammation and oxidative damage in the kidney of rats subjected to contrast-induced nephropathy. Nephrol Dial Transplant 2012;27:3136–40.10.1093/ndt/gfr807Suche in Google Scholar PubMed

135. Kobayashi M, Mikami D, Kimura H, Kamiyama K, MorikawaY, Yokoi S, et al. Short-chain fatty acids, GPR41 and GPR43 ligands, inhibit TNF-α-induced MCP-1 expression by modulating p38 and JNK signaling pathways in human renal cortical epithelial cells. Biochem Biophys Res Commun 2017;486:499–505.10.1016/j.bbrc.2017.03.071Suche in Google Scholar PubMed

136. Al-Harbi NO, Nadeem A, Ahmad SF, Alotaibi MR, AlAsmari AF, Alanazi WA, et al. Short chain fatty acid, acetate ameliorates sepsis-induced acute kidney injury by inhibition of NADPH oxidase signaling in T cells. Int Immunopharmacol 2018;58:24–31.10.1016/j.intimp.2018.02.023Suche in Google Scholar PubMed

137. Zhang W, Zhao T, Gui D, Gao C, Gu J, Gan W, et al. Sodium butyrate improves liver glycogen metabolism in type 2 diabetes mellitus. J Agric Food Chem 2019;67:7694–705.10.1021/acs.jafc.9b02083Suche in Google Scholar PubMed

138. Aguilar EC, Leonel AJ, Teixeira LG, Silva AR, Silva JF, Pelaez JM, et al. Butyrate impairs atherogenesis by reducing plaque inflammation and vulnerability and decreasing NFκB activation. Nutr Metab Cardiovasc Dis 2014;24:606–13.10.1016/j.numecd.2014.01.002Suche in Google Scholar PubMed

139. Aguilar EC, dos Santos LC, Leonel AJ, de Oliveira JS, Santos EA, Navia-Pelaez JM, et al. Oral butyrate reduces oxidative stress in atherosclerotic lesion sites by a mechanism involving NADPH oxidase down-regulation in endothelial cells. J Nutr Biochem 2016;34:99–105.10.1016/j.jnutbio.2016.05.002Suche in Google Scholar PubMed

140. Marques FZ, Nelson E, Chu PY, Horlock D, Fiedler A, Ziemann M, et al. High-fiber diet and acetate supplementation change the gut microbiota and prevent the development of hypertension and heart failure in hypertensive mice. Circulation 2017;135:964–77.10.1161/CIRCULATIONAHA.116.024545Suche in Google Scholar PubMed

141. Tirosh A, Calay ES, Tuncman G, Claiborn KC, Inouye KE, Eguchi K, et al. The short-chain fatty acid propionate increases glucagon and FABP4 production, impairing insulin action in mice and humans. Sci Transl Med 2019;11:eaav0120.10.1126/scitranslmed.aav0120Suche in Google Scholar PubMed

142. Jadoon A, Mathew AV, Byun J, Gadegbeku CA, Gipson DS, Afshinnia F, et al. Gut microbial product predicts cardiovascular risk in chronic kidney disease patients. Am J Nephrol 2018;48:269–77.10.1159/000493862Suche in Google Scholar PubMed PubMed Central

143. Weng J, Wu H, Xu Z, Xi H, Chen C, Chen D, et al. The role of propionic acid at diagnosis predicts mortality in patients with septic shock. J Crit Care 2018;43:95–101.10.1016/j.jcrc.2017.08.009Suche in Google Scholar PubMed

Received: 2019-09-18
Accepted: 2019-10-06
Published Online: 2019-10-22
Published in Print: 2020-04-28

©2020 Walter de Gruyter GmbH, Berlin/Boston

Artikel in diesem Heft

  1. Frontmatter
  2. Editorial
  3. Advancements in mass spectrometry as a tool for clinical analysis: Part I
  4. Drug adherence, testing and therapeutic monitoring
  5. Hyphenated mass spectrometry techniques for assessing medication adherence: advantages, challenges, clinical applications and future perspectives
  6. Method development for quantitative determination of seven statins including four active metabolites by means of high-resolution tandem mass spectrometry applicable for adherence testing and therapeutic drug monitoring
  7. Validation of a liquid chromatography tandem mass spectrometry (LC-MS/MS) method to detect cannabinoids in whole blood and breath
  8. THC and CBD concentrations in blood, oral fluid and urine following a single and repeated administration of “light cannabis”
  9. Identification of metabolites of peptide-derived drugs using an isotope-labeled reporter ion screening strategy
  10. Validation according to European and American regulatory agencies guidelines of an LC-MS/MS method for the quantification of free and total ropivacaine in human plasma
  11. Enhanced specificity due to method specific limits for relative ion intensities in a high-performance liquid chromatography – tandem mass spectrometry method for iohexol in human serum
  12. Small molecule biomarkers
  13. Applying mass spectrometry-based assays to explore gut microbial metabolism and associations with disease
  14. Trimethylamine-N-oxide (TMAO) determined by LC-MS/MS: distribution and correlates in the population-based PopGen cohort
  15. Development of a total serum testosterone, androstenedione, 17-hydroxyprogesterone, 11β-hydroxyandrostenedione and 11-ketotestosterone LC-MS/MS assay and its application to evaluate pre-analytical sample stability
  16. Short-term stability of free metanephrines in plasma and whole blood
  17. Validation of a rapid, comprehensive and clinically relevant amino acid profile by underivatised liquid chromatography tandem mass spectrometry
  18. UPLC-MS/MS method for determination of retinol and α-tocopherol in serum using a simple sample pretreatment and UniSpray as ionization technique to reduce matrix effects
  19. Independent association of plasma xanthine oxidoreductase activity with serum uric acid level based on stable isotope-labeled xanthine and liquid chromatography/triple quadrupole mass spectrometry: MedCity21 health examination registry
  20. Serum bile acids profiling by liquid chromatography-tandem mass spectrometry (LC-MS/MS) and its application on pediatric liver and intestinal diseases
  21. LC-MS/MS analysis of plasma glucosylsphingosine as a biomarker for diagnosis and follow-up monitoring in Gaucher disease in the Spanish population
  22. Dried blood spots and alternative sample mediums
  23. Investigating the suitability of high-resolution mass spectrometry for newborn screening: identification of hemoglobinopathies and β-thalassemias in dried blood spots
  24. Candidate reference method for determination of vitamin D from dried blood spot samples
  25. Therapeutic drug monitoring of anti-epileptic drugs – a clinical verification of volumetric absorptive micro sampling
  26. Simultaneous quantitation of five triazole anti-fungal agents by paper spray-mass spectrometry
  27. Obtaining information from the brain in a non-invasive way: determination of iron in nasal exudate to differentiate hemorrhagic and ischemic strokes
Heruntergeladen am 20.9.2025 von https://www.degruyterbrill.com/document/doi/10.1515/cclm-2019-0974/html
Button zum nach oben scrollen