Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Mar 24.
Published in final edited form as: Methods. 2024 Jan 5;222:81–99. doi: 10.1016/j.ymeth.2023.12.007

Microbiome metabolite quantification methods enabling insights into human health and disease

Jarrod Roach 1, Rohit Mital 2,#, Jacob J Haffner 3,4,#, Nathan Colwell 1,#, Randy Coats 1,#, Horvey M Palacios 3,4,#, Zongyuan Liu 1,#, Joseane L P Godinho 1,#, Monica Ness 1,#, Thilini Peramuna 1,#, Laura-Isobel McCall 1,4,5,*
PMCID: PMC11932151  NIHMSID: NIHMS2062264  PMID: 38185226

Abstract

Many of the health-associated impacts of the microbiome are mediated by its chemical activity, producing and modifying small molecules (metabolites). Thus, microbiome metabolite quantification has a central role in efforts to elucidate and measure microbiome function. In this review, we cover general considerations when designing experiments to quantify microbiome metabolites, including sample preparation, data acquisition and data processing, since these are critical to downstream data quality. We then discuss data analysis and experimental steps to demonstrate that a given metabolite feature is of microbial origin. We further discuss techniques used to quantify common microbial metabolites, including short-chain fatty acids (SCFA), secondary bile acids (BAs), tryptophan derivatives, N-acyl amides and trimethylamine N-oxide (TMAO). Lastly, we conclude with challenges and future directions for the field.

Keywords: Microbiome metabolite quantification, Instrumental methods, Mass spectrometry, Data processing, Bile acids, Short-chain fatty acids

1. Introduction

Metabolomics is defined as the study of small molecules, including those modified, created and consumed by metabolism. Metabolism is affected by factors like diet and activity level, but in various species like humans, the microorganisms that inhabit the host–collectively called the microbiome–also have a critical role [1]. Bacteria are the best-studied members of the microbiome on a metabolic level, but the microbiome also includes eukaryotes (helminths, protozoa, yeasts, fungi), archaea and viruses [2]. The microbiome may alter small molecules and their properties, including how long they remain at a given site in the body, and their effects [3]. The microbiome also produces entirely unique metabolites that do not arise without its presence [4]. Certain bacteria in the gut produce signaling molecules that affect host metabolic gene expression, as in the case of secondary bile acids [5]. The microbiome and their effects on metabolism have long-term and wide-ranging consequences for the host [6]. For instance, decreased levels of gastrointestinal short-chain fatty acids (SCFAs), a major group of gut microbiome-derived metabolites, has been associated with Alzheimer’s disease [7]. Similarly, stool samples from colorectal cancer patients have markedly lower levels of butyrate compared to those from healthy people [8]. As such, the examination of microbial metabolism is relevant to applications in human health.

Microbial metabolomics can involve annotation of detected molecules as well as quantification of these molecules. Quantification is often performed relative to reference samples, but absolute measurements may be taken if needed (for example in the case of a toxicity assessment of a certain compound, Table 1). Annotation may be required to gain biological insight, like in examining metabolic pathways. In contrast, mass spectrometry (MS)-based methods are able to use a combination of mass and retention time (or other chromatographic characteristics) to quantify a biomarker in clinical samples, even in the absence of annotation or structural elucidation.

Table 1.

Example scenarios of microbial metabolite quantification.

Project goal Absolute or relative quantification needed? Annotation needed? Example references
Determine which metabolites come from the microbiome Relative quantification may be sufficient Yes, for biological insight [4,11]
Identify microorganisms producing a specific metabolite, or conditions in which it is produced Relative quantification may be sufficient (even binary detected/not-detected information may suffice, being aware of limit of detection caveats) No [12,13]
Correlate specific microbial metabolites with clinical outcomes; biomarker identification, validation and implementation Initial studies may use relative quantification, but often absolute quantification is needed for subsequent studies Not necessarily [1416]
Determine the mechanistic role of a microbial metabolite in disease pathogenesis Initial studies may use relative quantification, but often absolute quantification is needed for subsequent studies Yes [17]
Analyze metabolism of a given drug or food product by the microbiome Relative quantification initially, which may be followed by absolute quantification, for example to assess toxicity Yes [1820]
Assess the toxicity of a specific microbial metabolite Absolute quantification Yes [21]

The purpose of this review is to serve as a primer for researchers new to metabolomics who are seeking to discover and quantify novel microbial metabolites or understand the role of well-studied microbial metabolites in human disease through the use of human studies or mouse models. We begin by discussing critical considerations for sample collection and analysis of microbial metabolites, so that quality data may be generated. Additionally, we will discuss some of the most common analytical methods used to study metabolites that are directly produced or chemically modified by the gut microbiome. From there, we will introduce means for identifying whether a metabolite is microbial in origin or produced by the mammalian host cells, though we acknowledge that some metabolites will be commonly produced by both microbiome and host, and may broadly distribute throughout the body [9]. After this, we highlight methods for determining the chemical structure of novel metabolites or the annotation of metabolites that have been previously studied. We then discuss specific quantitative methods for a wide variety of chemical families associated with the microbiome, with tools for relative or absolute quantification. Although thousands of metabolites have a microbial origin [10], we focus here on some of the most-studied classes (Figure 1), with the strongest current knowledge base with regards to human health. We conclude by introducing some of the challenges and opportunities for innovation currently associated with microbial metabolite quantification.

Figure 1. Example microbial metabolites.

Figure 1.

Figure generated using ChemDraw software.

2. Critical considerations when setting up microbial metabolite quantification studies

a. Impact of sample collection and metabolite extraction methods

Microbes can produce a wide variety of metabolites, ranging from small branched-chain amino acids to more complicated molecules like bile acids. Additionally, some metabolites are only found in specific tissues, or at greater abundance in one tissue compared to another. For example, the bile acid tyrosocholic acid can be below the limit of detection in cecum, colon, and fecal samples and thus would be missed if small intestine samples aren’t collected [11]. Thus, it is crucial to consider what sample type, sample collection method, and metabolite extraction method would work best for the metabolite(s) of interest prior to beginning an experiment.

Sample storage immediately following sample collection can have a dramatic impact on metabolite stability. Only a minority of fecal metabolites are unaffected after 24 h at room temperature, with strong effects observed on microbial metabolic activity [22][23]. Higher stability is observed in urine and serum kept at room temperature, but microbially produced branched-chain amino acids show a significant change in concentration in less than 24 hours [24,25]. Thus, immediate quenching of metabolism, for example through snap-freezing samples, is usually advisable, though these methods should be designed to minimize metabolite leakage from cells [26][27][28]. Nevertheless, immediate snap-freezing may not be practical under field conditions. Storing urine and whole blood samples on ice can help reduce changes to metabolite concentrations until they can be processed or stored into a −80°C freezer [24,29]. For fecal samples, 95% ethanol has the largest overlap of metabolites with freshly frozen samples for up to four days at room temperature [30][31]. Thus, this is a suitable alternative to snap-freezing during field collection. Fecal occult blood test (FOBT) cards have also shown acceptable performance [30]. Although RNAlater has commonly been used to preserve fecal samples for sequencing, it is usually considered incompatible with mass spectrometry due to the high salt content [3032], and is likely to decrease Nuclear Magnetic Resonance (NMR) spectroscopy sensitivity [33]. Commercial sampling kits designed for fecal metabolomics analysis are currently being developed [34]. For biofluids, volumetric absorptive microsampling (VAMS) serves as a viable means of sample collection at room temperature, with the majority of studies reporting stable metabolite concentrations for at least a few days following drying, though some metabolite levels are affected by 2 hours [3540].

Sample storage prior to metabolite extraction and data acquisition should minimize freeze-thaw cycles, which can have major impact on multiple microbial metabolite classes, including branched-chain amino acids [22,24]. Sample lyophilization can likewise lead to significant changes in the relative abundance of microbial metabolite classes such as short-chain fatty acids and branched chain amino acids [41]. Long-term storage (even without freeze-thaw) can still impact the metabolome [42]. For example, 20% of metabolite signals in dried blood spots stored at −20°C showed differential abundance by storage time, with some changes suggestive of degradation [43]. Likewise, some molecules such as lipids may oxidize during storage, unless stored under nitrogen gas [44].

b. Analytical instrumentation and data acquisition best practices

As a consequence of the variety of chemical classes produced by the microbiome, no single analytical method can analyze all microbial products. For instance, liquid chromatography-mass spectrometry (LC-MS), one of the most prolific analytical instruments in metabolomics research, is not well-suited to analyze microbially-produced short chain fatty acids without additional processing steps prior to analysis. Therefore, methods of data acquisition, which consequently affect sample preparation as well, must be considered prior to sample collection.

Data acquisition following metabolite extraction uses one of two main workflows: untargeted or targeted. Untargeted metabolomics aims to detect and quantify as many known and unknown metabolites as possible within a biological system, given experimental decisions like sample type and extraction solvent. Challenges include the high degree of diversity in chemical structures and varying concentration ranges between metabolites. In contrast, targeted metabolomics focuses on the precise and accurate measurement of a predefined set of metabolites. This is useful when studying a specific metabolic pathway or validating biomarkers identified by untargeted approaches. Other strengths include high sensitivity and specificity, quantitative and reproducible results, faster data processing and analyses, and suitability for validation studies. However, targeted approaches require prior knowledge of the metabolites to be measured and are therefore limited to the detection and quantification of known metabolites. In all cases, data acquisition in randomized order is critical to prevent the introduction of systematic effects that would confound data interpretation.

LC-MS is one of the most used methods for metabolomics analysis in microbiome research, allowing for the detection of a wide range of compounds, including polar compounds that may be challenging to detect using other methods [45][46][47]. Additionally, LC-MS based methods have large libraries of public reference data which can help with putatively annotating molecules in untargeted analysis, although these libraries tend to be composed of data collected from positive electrospray ionization, making annotation of data collected via negative electrospray ionization more challenging. While a powerful technique, LC-MS analysis can be complicated by the high degree of sample complexity, which can lead to issues with ion suppression and interference, as well as inter-run and inter-site variability in chromatography retention times.

Gas chromatography-mass spectrometry (GC-MS) is another common instrument in microbiome metabolomics research (e.g. [48][49,50][5153]), allowing for the identification of volatile and semi-volatile compounds in complex mixtures, or derivatized non-volatile compounds. This chemical class and size restriction is a major limitation of GC-MS. Furthermore, artifacts resulting from derivatization (or uneven derivatization between samples) could impair quantification [54]. On the other hand, a strength of GC-MS is the standardized electron ionization (EI) conditions, facilitating spectral comparisons between experiments over time and to reference libraries. Most GC-MS instruments currently in use or in facilities are low-mass-resolution, so they may not provide high-confidence annotations, though this is being addressed by technology such as GC-orbitrap instruments [55].

NMR can also be used for microbiome metabolomics due to its non-destructive nature, easy sample preparation in bulk material analyses, and lack of chemical derivatization requirements [56]. For example, short-chain fatty acids and branched-chain amino acids could be studied in feces by NMR using simple three-step sample preparation protocols [57][58]. NMR can also be advantageous in detecting and characterizing compounds that are challenging to analyze with LC-MS, such as sugars, organic acids, alcohols, polyols, and highly polar compounds [59][60][61]. Quantification by the addition of chemical standards, also known as quantitative NMR (qNMR), allows for the accurate measurement of NMR peak heights with the added internal standard [62]. However, qNMR can obscure the reliable measurement of peak intensities due to a high level of NMR signal overlap [63]. Low sensitivity is the inherent disadvantage and foremost challenge for the application of NMR compared with MS [64]. However, continuous developments to improve the sensitivity through the use of higher magnetic field strength [65], cryogenically cooled probes [66], and microprobes [67] have significantly enhanced the sensitivity of NMR.

Though less commonly implemented, other analytical methods such as capillary electrophoresis-MS (CE-MS) (e.g. [68][69]), direct infusion MS with no separation (e.g. [70,71]), matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI)(e.g. [72]), or desorption electrospray ionization mass spectrometry (DESI-MS) (e.g. [70]) can also be used to quantify microbial metabolites.

Overall, key considerations guiding instrumental choices are the specific goals of the experiment (see Table 1 for some example scenarios), the analytes of interest, sensitivity, precision, and accuracy needed, and whether absolute versus relative quantification is required. However, in all cases, best practices will include quality assurance/quality control steps, which should include system suitability testing. While out of the scope of this review, readers are referred to recent reviews on best quality assurance/quality control practices for targeted [73] and untargeted metabolomics [74].

c. Data processing best practices

There have been extensive reviews on data processing for metabolomics (e.g. [75][76][77][78]), and detailed protocols published on data processing tools and approaches (e.g. [79]), which naturally also apply to microbiome metabolite analysis. Here, we highlight critical points for consideration. LC-MS-based metabolomics data processing refers to “metabolite features” as a combination of chromatographic retention time and mass over charge (m/z). This recognizes that different isomers will have the same m/z but different structures, leading to different retention times. Likewise, one single structure can lead to multiple adducts or in-source fragments, so that one unique metabolite can have multiple m/z values, though at the same retention time. This complexity is one of the challenges of metabolomics data processing. Combining ion mobility with current chromatography and MS techniques is helping to distinguish between isomers (e.g. [80]).

Currently, there are many popular non-commercial platforms for untargeted analysis, such as MZmine [81], XCMS [82], OpenMS [83], and MS-DIAL [84]. For targeted analysis, example softwares include Skyline [85], MRMkit [86] and OpenMS [83]. One key strength of MZmine is its modular design, which allows users to customize the software based on their needs [81]. XCMS has two versions: the XCMS R package or XCMS Online (cloud-based version). For XCMS Online, users can upload and process LC-MS data directly without installation of software or coding skills [87]. MS-DIAL is an alternative platform, which implements deconvolution algorithms for data-independent acquisition (DIA) data sets [84]. Skyline was designed for quantitative proteomics but was later expanded to small molecules; a strength is that it is not instrument-specific [85,88]. MRMkit is an MS-based targeted analysis software that can learn retention time offset patterns for different compound classes. This information is then used to provide recommendations for peak picking in multimodal ion chromatograms, which can be complex and difficult to interpret manually [86]. OpenMS is an open-source, cross-platform software designed for high-throughput targeted and untargeted MS data. It provides a set of over 180 tools and workflows [83].

Peak detection algorithms are used as a first data processing step to identify the true peaks in the data, and to differentiate them from background noise (Figure 2). Deconvolution separates experimental data to create a pure spectrum for each component present in a sample [89,90]. MSHub is a newly developed machine learning deconvolution method that deconvolutes GC-MS data automatically and evaluates the consistency of fragmentation patterns across multiple samples [91]. DecoID is another new deconvolution method that seeks to address mixed fragmentation spectra using a LASSO regression method. This can help improve metabolite identification rates [92].

Figure 2.

Figure 2.

Workflow for LC-MS data analysis.

The prerequisite of quantitative metabolite analysis is to ensure that the same metabolites are being compared across all samples. Peak alignment, normalization and batch effect removal are important steps after peak detection. Without proper peak alignment, comparisons between samples can be misleading. The above-described software packages can perform alignment by themselves, or they can be aided by external tools such as neighbor-wise compound-specific Graphical Time Warping (ncGTW) [93]. Normalization is the process of adjusting metabolite peak areas to reduce technical variability between samples due to differences in sample preparation, instrument performance, or other factors that could lead to incorrect conclusions [94]. There are several normalization methods for MS data, such as internal standard normalization [95], total ion current (TIC) normalization [96] and probabilistic quotient normalization (PQN) [97]. PQN assumes that the alterations in concentration of one analyte will only affect part of the data, while changes in the sample concentration impact the complete data in each sample. WaveICA normalization is based on wavelet analysis, which can not only remove batch effects but also show better classification for untargeted metabolomics data [98]. hRUV is a batch effect removal tool which can remove batch effects for both intra-batch and inter-batch variation [99]. All normalization methods make assumptions, so it is important to pick a method that addresses technical variability within the specific dataset.

Most data processing workflows will remove background signals contributed by tubes, instruments, solvents, etc. This is best performed by generating an empty tube that will go through the same extraction and handling processes as actual samples (“process blanks”). Data acquired from that blank can then be used to process the acquired data. Blanks can also be added at the time of data acquisition but would not allow for the removal of contaminating features arising during the extraction steps. Common methods for background signal removal include removing metabolites that are shared with the blank (which may however remove real metabolite features in case of low-level carryover) or removing any metabolite that fails to meet a certain signal intensity fold difference threshold when compared to blanks (less affected by minor carryover effects). More complex, data-dependent approaches have also been developed [100]. Packages such as CROP [101], MetaClean [102], or NeatMS [103] can remove non-informative features contributed from the blank or false positive features introduced by data processing. Removing these figures will help minimize noise in the data and may help with statistical power and annotation.

3. Attributing microbial origin of candidate features: important considerations

Following proper data-processing, non-biological features should be minimized in the sample, leaving mainly biological metabolic features. At this point, identifying whether each remaining feature is of microbial origin can be determined through further analyses. Resources like the Microbial Metabolites Database [10] provide known microbe-metabolite relationships, though these associations do not represent the full scope of microbial chemistry. Moreover, much of untargeted metabolomics data still cannot be easily annotated [104,105]. Moving from known microbial metabolites into unknown microbe-metabolite relationships requires additional steps to identify any microbial origins. Broadly, these approaches can be grouped into software-based methods and experimental methods.

a. Software-based approaches

Correlations are among the most popular methods to estimate associations in microbiome-metabolome data. Typically, researchers select correlation techniques based on normality and/or fit of data, such as parametric (normally distributed) or nonparametric approaches. For example, Spearman’s rank correlation coefficient and Pearson’s rank correlation coefficient are both popular approaches for exploring microbiome-metabolome associations [106109]. Correlations are especially useful for quickly identifying strong associations between microbes and metabolites, offering simple guidelines for determining linear relationships between these data [110]. However, such correlation approaches assume data are independent, which is not true of microbiome-metabolome data. Moreover, the compositional nature of microbiome and metabolome data further confound correlation analyses [111,112]. Thus, correlations can be useful for identifying basic patterns within the data, but should only be used cautiously, with additional validations to eliminate false associations.

Beyond general approaches like Spearman or Pearson, correlation approaches can be expanded to perform specialized comparisons. One such correlation-based technique is weighted gene co-expression network analysis (WGCNA). Briefly, WGCNA creates a network of co-expressed genes clustered by their similarity and uses this network to report associations with other paired data [113,114]. Through integration with microbial and metabolomic data, WGCNA can identify microbe-metabolite associations by focusing on bacterial genes correlated with the metabolites of interest. For example, WGCNA has been used to examine relationships between bacteria and metabolites associated with Chagas disease [115], revealing key metabolite-altering bacterial taxa involved in Trypanosoma cruzi infection. By itself, WGCNA is a useful technique for attributing microbe-metabolite relationships, but it also represents a broader data analysis approach involving correlations.

To overcome the hurdles seen in correlations, new software tools estimate microbe-metabolite relationships while considering the compositional and dependent nature of these data. One such tool is microbe-metabolite vectors, also known as ‘mmvec’ [116]. mmvec is a neural network pipeline that predicts microbe-metabolite relationships using microbial sequence and metabolomic data. Rather than identifying associations, mmvec examines the probability of co-occurrence between microbe-metabolite pairs derived from their paired abundances [116]. However, mmvec is computationally intensive and less suited for large datasets. Additionally, there are no validation or confidence measures built into mmvec; it simply outputs the conditional probability values for all microbe-metabolite pairs [116]. Despite these limitations, mmvec is an useful alternative to correlations for exploring microbe-metabolite relationships with integrated multi-omics data [117120].

Other data analysis techniques can include Mass Spectrometry Search Tool (MASST) [13]. Akin to a Basic Local Alignment Search Tool (BLAST) search for comparing sequence data, MASST is a web-based search tool that searches inputted MS/MS data against a range of public MS/MS spectra databases. MASST uses a spectral library containing tens of thousands of annotated mass spectra of small molecules to match the m/z values of user-submitted spectra against those in the library. The output contains multiple potential matches ranked by their similarity to the user’s spectra. Importantly, sample information attributed to these public data are available when performing MASST searches, allowing researchers to identify reported sources of detected MS/MS spectra, like potential microbial sources. For example, Quinn and colleagues used MASST to run searches against public data to further confirm association between novel amino acid-conjugated bile acids and the microbiome [11].

An additional data analysis approach involves reference data-driven (RDD) analyses. RDD approaches use a controlled, standardized vocabulary when creating metadata of database references used to match against user-submitted MS/MS data. Based on these matches, researchers can get source information of their submitted MS/MS spectra through matches to these curated metadata reports of public data. Importantly, these RDD approaches maximize the utility of MS/MS spectra and allow matches against curated source information, as seen by Gauglitz et al. [121] While this particular study focuses on identifying food sources through RDD analyses, these approaches can be extended into broader and microbial contexts.

b. Experimental approaches

These data analysis-centric approaches can be complemented through experimental techniques to report microbe-metabolite relationships. One predominant experimental approach for validating microbe-metabolite associations involves culturing the candidate-producing microbe and screening for metabolite(s) of interest, usually informed by prior hypotheses of a microbe-metabolite relationship. This process isolates the microbe from its natural environment and avoids host organism modification of the metabolite of interest. Thus, analyzing the metabolites produced in the bacterial culture reports a direct microbe-metabolite relationship. Previous studies have used this technique in varied ways, ranging from culturing bacteria to reconstructing genome-wide metabolic pathways of the detected metabolites [122125][122,123] or tracing microbiome-dependent metabolites in colonized mice to metabolome profiles of cultured bacteria [126]. However, culturing conditions are meant to simulate, not recreate, the microbe’s natural environment; the expected metabolites might not necessarily be produced in all culturing experiments.

Another experimental approach involves comparisons between germ-free and colonized animal models, such as mice. These approaches compare sterile and microbially-colonized animal metabolome data, where metabolites enriched in the colonized animals likely have some microbial association. Examining different abundance patterns of metabolites can highlight microbially-modulated metabolite enrichment or depletion. Such animal-based comparison approaches are particularly useful for confirming the microbiome’s role in influencing specific metabolites, because it tests for the metabolites in the “natural” environment with the greater microbiome community. This concept can be mimicked in humans by investigating samples from humans with resected colons, as was the case with identifying the bacterial origin of 5-hydroxyindole [127], or through the administration of a poorly absorbed oral antibiotic cocktail [128130]. However, these approaches only report broad microbe-metabolite relationships without providing specific microbial sources. For example, microbe-associated metabolites might be produced by the host responding to the bacterial presence, rather than being produced by the bacterium itself. These animal-model approaches simply indicate a trend and require further work to demonstrate any metabolites are indeed produced by the microbe in question. One method to address this shortcoming is through stable isotope labeling of bacteria, which enables differentiation with host-derived unlabeled metabolites by LC-MS [9]. Uchimura et al gavaged mice with 13C-labeled E. coli to investigate the nature and spatial spread of microbiome-derived metabolites. After 2 hours, an untargeted LC-MS analysis showed labeled amino acids, fatty acids and nucleotides, produced by the microbiome, penetrate organs throughout the body. Through this, the researchers were able to determine the ratio of microbiome-produced branched-chain amino acids to host-derived in each organ [9]. Another approach, as used in experiments to identify the microbial origin of indolepropionic acid, is to knock out crucial genes associated with the production of a metabolite in cultured bacteria and then inoculate germ-free mice with the mutant bacteria to identify if the metabolite of interest is still produced [131]. Unfortunately, sterile vs. colonized animal comparisons might not always detect host-produced antagonistic pathways influencing microbial activity, such as metabolic reabsorption in the ileum. Despite these drawbacks, animal models are popular for determining microbe-metabolite relationships, such as confirming microbial metabolism of novel amino acid-conjugated bile acids [11], identifying blood metabolomic responses to gut microbial activity [4], describing longitudinal analyses of urine metabolomes throughout gut bacterial colonization [132], and linking gut microbial activity to energy and lipid storing metabolites involved in modulating host metabolism [133], to name a few.

These approaches reflect varying ways of identifying microbial origins for metabolites, but there are important considerations to note when doing these analyses. The first major consideration is how sample type and preparation method can influence observed microbiome-metabolome associations. This is particularly important from an experimental side, as evidenced by Han and colleagues observing distinct metabolomes based on sample types of colonized mice [126]. Additionally, another important consideration includes spatial dynamics of the gut. For example, metabolites with reduced abundance in the lower gastrointestinal tract might be absorbed by the gastrointestinal tract, or their depleted abundance could be the result of microbial modulation. A third consideration when investigating microbial origins of metabolites is that a detected functional gene might not always produce a metabolite in all contexts. This consideration is especially prominent when identifying a microbe-metabolite relationship through culturing. Since the culture does not perfectly mimic the microbe’s natural environment, some metabolites might not be produced in that cultured setting despite the metabolite being produced in the gut. Ultimately, identifying a microbial origin of a metabolite requires careful consideration and should ideally involve multiple complementary analyses. The approaches discussed here are not an exhaustive list but represent common ways for investigating microbial origins of metabolites. Other growing areas for identifying microbe-metabolite relationships include machine learning tools [134], especially for determining chemical properties of such microbially-derived metabolites.

4. From microbial features to chemical structures of novel microbial metabolites

Having established that a novel feature is microbial in origin, the next step is structure elucidation. Most of the candidate microbe-metabolite associations described above rely on untargeted metabolomics. An accurate structure elucidation is critical to understand the biological role of the metabolite, and its biosynthesis pathway.

The first step in the structural identification involves acquiring the metabolites in reasonable quantities, typically in the microgram to milligram range. Unless native environments provide enough of the metabolites, culture conditions may be required to accrue more. This involves routine microbiology techniques, including cultivation of microbial strains on specific media with special nutrients, cocultivation of multiple strains together and production of metabolites of interest in a specific growth phase [135][136][137]. However, as discussed above, it is important to note that the majority of microorganisms resist cultivation in the laboratory as the required conditions deviate too far from the organism’s natural environment [138]. Furthermore, metabolite production is often context dependent. As an alternative, isolation chip (iChip) incubation has been developed to facilitate growth in simulated natural environments [139]. For example, a variant of the iChip has been successfully used to cultivate anaerobic species of the human oral microbiome [140]. Another successful iteration is the gut-on-a-chip platform which enabled the coculture of 200 different aerobic and anaerobic species with human intestinal epithelium [141].

The isolation of microbial metabolites involves a wide range of LC methods based on separation capacities and selectivity [142]. Aside from isolation and separation of microbial metabolites, a major application of high-performance liquid chromatography (HPLC) methods is dereplication by hyphenated techniques including LC-NMR and LC-MS [143]. Dereplication involves identifying known compounds based on the reported data generated by MS or NMR. Most dereplication bioinformatics tools, including GNPS [144], Metlin [145], DNP [146], and NIST [147], work with integrated or online databases of accurate mass spectra, compound structures, structures of building blocks of molecules, and fragmentation routes or combination of multiple features.

After sample isolation and replication, the metabolite is ready for structural analysis. NMR is the core technique in structural elucidation, and can be supplemented with other analytical techniques such as high resolution MS, circular dichroism (vibrational and electronic), Raman optical activity, optical rotation measurements, and X-ray crystallography, depending on molecule properties [148]. 1H and 13C NMR spectra provide information about the quantitative composition of the unknown metabolite [149]. The interpretation of NMR data along with high resolution MS data is used for the determination of the molecular formula of the novel metabolite [149]. The assembly of the relative configuration is further supported by additional 2-dimensional NMR experiments such as HSQC (Heteronuclear Single Quantum Coherence), COSY (Correlation spectroscopy), and HMBC (Heteronuclear multiple bond correlation) [150]. The structure elucidation of novel metabolites may be a complicated process based on structural complexity. Spectroscopists may interpret different structures from the same initial NMR data [151], by misjudged interpretation of chemical shift annotations, misinterpretation of characteristic spectral features, and resonance overlap in NMR signals [152].

For labs without access to an NMR or without the expertise to interpret NMR data, or sometimes prior to NMR data acquisition, MS can also be used to understand the structure of an unknown metabolite. Example annotation tools for MS data include molecular networking, which groups features by MS2 spectral similarity, enabling annotation propagation to analogs with mass differences compared to library references [144][153]. MEtabolite SubStructure Auto-Recommender (MESSAR) automatically builds potential relationships between spectral data and substructure data, which are captured and learned from public spectral libraries. This approach allows MESSAR to provide multiple candidates for annotation with similar substructures, even if the exact molecule is not present in the reference database [154]. Small Molecule Accurate Recognition Technology (SMART 2.0) is a web-based tool with improved accuracy for mixture sample analysis [155]. While structures can be determined from MS/MS or MS/MS/MS data, they cannot usually provide stereochemical determination. Additionally, identification of the molecular structure from untargeted MS data alone can be challenging, as multiple molecules may have the same m/z values and fragmentation patterns. Feature annotation is a very active area of metabolomics research, and new tools are regularly being developed [156].

In general, no single technique can unambiguously assign the structure of a complex unknown compound. The synergistic combination of information obtained from different techniques, like crystallography and NMR, combined with chemical and biosynthetic logic lead to structure elucidation of novel metabolites with higher reliability [151].

5. Class-specific quantification methods for common microbial metabolites

After annotating the metabolite of interest (or choosing to forgo annotation should it not be necessary for the goal of the study), quantification should be performed next. Greater method validation is required when quantifying novel microbial metabolites that lack established methods. This would involve testing data acquisition methods using pure standards across a range of known concentrations, ideally of the same molecule or of molecules with similar functional groups, to ensure separation (if using chromatography techniques) and to assess the limit of detection/limit of quantification. Method validation with standards should be followed by assessment on representative biological samples, such as sample pools (pooled quality control [pooled QC]) or standardized biological samples (National Institute of Standards and Technology (NIST) standard reference materials). Quantification of frequently studied microbial metabolites will need less in-house method validation, since they generally rely on already-developed methods. Additionally, absolute quantification of known metabolites can be aided by diluting samples with a known amount of an isotopically labeled version of the metabolite [128,157]. In this section, we highlight quantification methods for highly studied microbial metabolites. Should a novel microbial metabolite share structural commonality with one of the metabolic classes listed below, the quantitative methods associated with that class can be used as a starting point for method development.

a. Short-chain fatty acids

Fatty acids with fewer than 8 carbon atoms are classified as short-chain fatty acids (SCFAs). The most prevalent SCFAs include acetic acid, propionic acid, and butyric acid [158,159]. When dietary fibers undergo fermentation by bacterial hydrolytic enzymes, the primary products are SCFAs. SCFAs play a crucial role in promoting the proliferation of beneficial bacteria, supplying energy to colonocytes, modulating the gut immune system, preventing autoimmune diseases, and enhancing overall gastrointestinal health [160164].

Feces, plasma, and tissue samples each offer unique information about SCFA production, metabolism, and distribution within the body [165]. Fecal samples are a conventional choice for evaluating SCFA production within the gastrointestinal tract. However, their SCFA concentrations may not accurately reflect the local rates and levels of SCFA production. This is because as much as 90% of these metabolites are absorbed by colonocytes [166], and the concentration of SCFAs varies along the length of the gut. The highest levels are typically found in the cecum and proximal colon, gradually decreasing towards the distal colon [161,167,168]. Plasma samples are useful for evaluating systemic SCFA, despite their typically low concentrations. SCFAs produced in the gut can be transported via the portal vein to the liver, where they undergo metabolism before being released into the systemic circulation [167]. Tissue samples can provide insights into how SCFA are metabolized and used by specific organs or tissues. For instance, adipose tissue is a significant site for SCFA uptake and metabolism, while the brain can also use SCFAs as an energy source [165,169,170] (Figure 3).

Figure 3. Hallmarks of short-chain fatty acids quantification.

Figure 3.

Figure generated using BioRender.com

To ensure accurate and precise quantification of SCFAs, it is essential to consider their chemical and physical properties, as these factors can significantly impact the accuracy and precision of measurements. Some important properties to take into consideration include [165,171,172]:

  1. Volatility: SCFAs are volatile compounds that can evaporate during sample preparation and analysis.

  2. Solubility: Some solvents have selectivity in extracting specific SCFAs. For instance, diethyl ether is suitable for extracting butyric acid and valeric acid, while hexane is commonly used for the extraction of less polar SCFAs, such as caproic acid and caprylic acid.

  3. Ionization: SCFAs are weak acids that can ionize in solution, with the degree of ionization being dependent on the pH of the solution. The ionization state of SCFAs can influence their behavior in chromatography and their detectability using mass spectrometry.

  4. Stability: SCFAs can be prone to degradation or chemical modifications during sample preparation and storage. It is crucial to employ optimized storage conditions (e.g., low temperature, acidification) and appropriate derivatization or stabilization methods that help overcome these challenges.

  5. Matrix effects: SCFAs are often found in low concentrations within complex matrices like feces or plasma, which contain various other organic acids and metabolites. These matrix effects have the potential to interfere with the accurate quantification of SCFAs [173,174].

For reliable SCFA measurements, it’s essential to consider the properties described above and use appropriate methods. Modern analytical methods, both qualitative and quantitative, are highly effective for analyzing SCFAs in a variety of sample types. These methods include approaches like gas chromatography, high-performance liquid chromatography, and capillary electrophoresis, often paired with detection methods such as mass spectrometry.

In the context of GC-MS analysis, a common procedure for extracting SCFAs from various matrices involves the addition of an acidifying agent, such as phosphoric or sulfuric acid, to the sample. Subsequently, this mixture undergoes liquid-liquid extraction (LLE) with an organic solvent, like ethyl acetate or diethyl ether. These extraction techniques are chosen to specifically address concerns related to solubility and stability, thereby enhancing the accuracy of the quantification process. It’s worth noting that water extraction is discouraged for SCFA GC–MS analysis due to its detrimental impact on MS determination [175,176]. Despite being considered somewhat more prone to errors, the liquid-liquid extraction method (LLE) offers advantages such as straightforwardness and swiftness. Moreover, it obviates the need for specialized extraction filters, cartridges, or columns [48].

In LC-MS analysis, the extraction of SCFAs often employs solid-phase extraction (SPE). This technique uses a solid-phase sorbent, like C18 silica-based material, to selectively adsorb and retain the SCFAs from the sample matrix. Following this adsorption, the sorbent undergoes a washing process, and the SCFAs are subsequently eluted using solvents such as methanol, formic acid or acetonitrile. The resulting extract is then evaporated to dryness and reconstituted in a solvent compatible with downstream chromatography and MS methods, such as water or acetonitrile (Table 2) [174,177180].

Table 2.

Short chain fatty acid quantification methods.

Quantification Method Reference Sample Type Extraction Column Mobile Phase Ionization Method Molecule(s)
GC-MS Zhu et al., 2022 [187] Feces Diethyl ether DB-FFAP Helium, flow rate 1.5 mL/min EI Acetic acid, formic acid, propionic acid, isobutyric acid, butyric acid, isovaleric acid, valeric acid, 2-methylvaleric acid, 4-methylvaleric acid, hexanoic acid, heptanoic acid
Fu et al., 2022 [188] Feces HS-SPME (Headspace Solid-Phase Microextraction). Derivatized with isobutyl chloroformate and isobutanol Rxi-5MS Helium, flow rate 1.0 mL/min EI Formic acid, acetic acid, propanoic acid, isobutyric acid, butyric acid, 2-methylbutyric acid, isovaleric acid, valeric acid, 3-methylvaleric acid, hexanoic acid, heptanoic acid
Kim et al., 2022 [167] Plasma
Serum
Feces
Cecum
Methyl tert-butyl ether DB-FFAP Helium, flow rate 1.0 mL/min EI Acetic acid, propionic acid, butyric acid, valeric acid
Yao et al., 2022 [189] Plasma Methyl tert-butyl ether DB-WAX Helium, flow rate 1.2 mL/min EI Acetic acid, propionic acid, butyric acid
Wang et al., 2020 [190] Serum Ethanol and n-hexane DB-FFAP Helium, flow rate 1.0 mL/min EI Acetic acid, propionic acid, isobutyric acid, pivalic acid, butyric acid, 3-methylbutyric acid, 2-methylbutyric acid, valeric acid, 2-ethylbutyric acid, 2-methylvaleric acid, 3-methylvaleric acid, 4-methylvaleric acid, hexanoic acid, cyclopentanic acid
LC-MS Shafaei et al., 2021 [171] Serum Methyl tert-butyl ether. Derivatized with 3-nitrophenylhydrazine hydrochloride and 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride C18-AR A: Water
B: Acetonitrile
ESI - Negative mode Acetic acid, propionic acid, isobutyric acid, butyric, acid, isovaleric acid, valeric acid, 4-methyl valeric acid, hexanoic acid
Bihan et al., 2022 [172] Cecal contents Water/acetonitrile (1:1. v/v). Derivatized with aniline and N-(3-Dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride C18 A: Water + 0.1% formic acid
B: Methanol + 0.1% formic acid
ESI - Positive mode Acetate, propionate, butyrate
Microbial cultures Water/methanol (1:1. v/v). Derivatized with aniline and N-(3-Dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride
Nagatomo et al., 2018 [173] Feces Methanol with QuEChERS method. Derivatized with 2-picolylamine in 2,2’-dipyridyl disulfide and triphenylphosphine in acetonitrile C18 A: Water + 0.1% formic acid
B: Methanol + 0.1% formic acid
ESI - Positive mode Acetic acid, propionic acid, butyric acid, isobutyric acid, valeric acid, hydroangelic acid, 4-methylvaleric, succinic acid
Liebisch et al., 2019 [174] Feces
Colon content
70% isopropanol. Derivatized with 3-nitrophenylhydrazine hydrochloride and N-(3-dimethylaminopropyl)-N’-ethylcarbodiimide hydrochloride C18 A: Water + 0.1% formic acid
B: Methanol + 0.1% formic acid
ESI - Negative mode Acetate, propionate, butyrate, isobutyrate
Saha et al., 2021 [168] Feces
Liver
Kidney
Brain
Skeletal
Muscle
Spleen
Microbial media
0.5% orthophosphoric acid PGC C18 A: Water + 0.1% formic acid
B: Methanol + 0.1% formic acid
ESI - Positive and negative mode Acetate, butyrate, isobutyrate, isovalerate, valerate, lactate, propionate.

Derivatization involves the chemical modification of a compound’s structure to optimize its suitability for analytical applications. In the context of SCFAs, this often includes introducing an alkyl or silyl group to the carboxylic acid functional group. This chemical modification improves the main challenges properties of the volatility and stability of SCFAs, rendering them more compatible with analysis methods such as GC-MS and LC-MS [172]. Derivatization reagents for SCFAs commonly include pentafluorobenzyl bromide (PFBBr) [165], trimethylsilyl (TMS) derivatization reagents [181], and chloroformate [182,183]. PFBBr derivatization is time-consuming, and silylation requires anhydrous conditions, which is cumbersome [184,185]. In contrast, chloroformate derivatization is widely preferred for SCFAs derivatization due to its convenient operation, mild conditions, and reduced time consumption [182]. However, it’s worth noting that derivatization involves organic solvents and other compounds that may be toxic, volatile, or flammable. Moreover, it extends processing time and introduces another potential source of variability, contamination, and errors [165,185].

GC–MS separation is often carried out using FFAP columns (polar nitroterephthalic-acid-modified polyethylene glycol). It has proven to be a robust method, albeit one that involves time-consuming sample preparation and derivatization processes. In contrast, LC-MS may require less time for sample preparation and analysis, while employing lower temperature conditions can also prevent the degradation of volatile SCFAs [186188]. However, SCFAs pose challenges in terms of their chromatographic and ionization properties when using conventional LC-MS equipment [174]. To address these challenges, for certain types of matrices, derivatizing samples can enhance the chromatographic separation and ionizability of SCFAs. Some effective strategies include derivatization using 3-nitrophenylhydrazine and aniline [189,190]. Both strategies yield favorable results and allow for SCFA analysis on conventional C18 chromatography columns. Typical mobile phases consist of acidified HPLC-grade polar solvents (e.g., water) and non-polar solvents (such as acetonitrile or methanol) for analyte separation through isocratic or gradient elution profiles. These separations are conducted under relatively mild reaction conditions [189,191,192].

b. Bile acids

Bile acids (BAs) occupy a central position in the absorption, excretion and metabolism of lipids within the body [197]. The gut microbiome modifies endogenous primary BAs to produce exogenous secondary BAs [198]. The best-studied secondary BAs are lithocholic acid (LCA) and deoxycholic acid (DOC) [199]. However, recent work has identified more than 140 new secondary BAs in existing datasets, including the novel bile acids phenylalanocholic acid (Phe-chol), tyrosocholic acid (Tyr-chol) and leucocholic acid (Leu-chol) [11]. With credit to public metabolomic databases (GNPS, MassIVE) and the use of synthesis-based reverse metabolomics, even more novel bile conjugates are being discovered [200]. Secondary bile acids at increased levels promote higher chances of developing colorectal carcinomas, gallstone disease, cirrhosis of the liver and Clostridioides difficile infection [201211]. BAs serve as signaling molecules with the ability to bind to receptors such as farnesoid X receptor (FXR), Takeda G protein-coupled receptor 5 (TGR5), constitutive androstane receptor (CAR), and more, with each BA being able to bind with multiple receptor types with varying degrees of effect [212,213]. Further adding to the complexity of BA signaling, bile acids can be conjugated with glycine, taurine, and to a lesser studied extent, other amino acids, which can affect the ability of BAs to act as substrates for specific receptors in the body [11,12,214]. In the context of cancer, the pro- or anticarcinogenic roles of specific BAs have been shown to be reliant on the type of neoplasia and the concentration of the BA, emphasizing the importance of accurate quantification of BAs in order to better understand their role in the body [215].

Regardless of the specific bile acid structure, quantification methods have relied predominantly on LC-MS approaches. Samples most commonly collected for BA analysis are blood, urine, feces, liver, and bile, depending on the BAs of interest. A C18 LC column coupled with MS/MS analysis is most commonly used for BA analysis [216][217] (Table 3). NMR is another common method to analyze BAs and distinguish stereochemical differences (Table 4). Although there is minimal sample preparation, the lower sensitivity of NMR compared to LC-MS make it less common for current BA research [218]. Enzyme-linked immunosorbent assay (ELISA) is another method used to measure BAs but can be limited by antibody cross-reactivity [219221].

Table 3.

MS-based bile acid quantification methods.

Quantification Method Reference Sample Type Extraction Column Mobile Phase Ionization Method Molecule(s)
LC-MS Sarafian et al., 2015 [218] Plasma Ice cold methanol BEH C8 A: Acetonitrile/wate r (1:10. v/v) + 1mM ammonium acetate
B: Acetonitrile/2-propanol (1:1. v/v)
ESI - Negative mode Primary and secondary BAs
Alnouti et al., 2008 [216] Bile Oasis-HLB SPE conditioned with MeOH. Washed twice with water and eluted with MeOH. UPLC C18 A: 95:5 Methanol: acetonitrile
B: Water + 7.5 mM ammonium acetate, pH 4
ESI - Positive and negative mode Positive - Glycine conjugated BAs Negative - Taurine and unconjugated Bas
Lucas et al., 2021 [12] Bacterial culture 100% water BEH C18 A: Water + 10mM ammonium acetate, pH 6
B: Methanol
ESI - Negative mode Secondary BAs
Gómez et al., 2020 [219] Plasma
Serum
Diluted with water. Protein precipitated with 2-propanol BEH C18 A: 95:5 Water: acetonitrile
B: 5:95 Water: acetonitrile
ESI - Positive and negative mode Unconjugated and Conjugated BAs and secondary BAs
Liver Water:chloroform:methanol(1:1:3. v/v/v) BEH C18 A: 95:5 Water: acetonitrile
B: 5:95 Water: acetonitrile
ESI - Positive and negative mode Unconjugated and Conjugated BAs and secondary BAs

Table 4.

NMR-based bile acid quantification methods.

Quantitation Method Reference Sample Type Solutions NMR Molecule(s)
1H-NMR Nagana Gowda et al., 2009 [220] Bile Double distilled water, D2O, Trimethylsilylpropanoic acid (TSP) 800 MHz NMR spectrometer with 5-mm CHN inverse probehead Glycocholic acid, glycodeoxycholic acid, glycochenodeoxycholic acid, taurocholic acid, taurodeoxycholic acid, taurochenodeoxycholic acid
1H-NMR/13C-NMR (1H-1H, 1H-13C) Nagana Gowda et al., 2006 [221] Bile Double distilled water, D2O, Trimethylsilylpropanoic acid (TSP) 800 MHz NMR spectrometer with 5-mm CHN inverse probehead Glycocholic acid
Glycodeoxycholic acid
Glycochenodeoxycholic acid
Taurocholic acid
Taurodeoxycholic acid
Taurochenodeoxycholic acid

Separation and quantification of BAs and their conjugates are challenging because of marked differences in chemical properties, the presence of isomeric forms, and their relatively low concentrations in biological samples [222]. Longer LC gradients allow for adequate separation and annotation of isomers from MS/MS analysis [223]. Current research continues to discover new derivatives of BAs, further complicating efforts to link the health of our gut’s microbiome to the quantity and signaling properties of BAs that have relevant physiological effects.

c. Branched-chain amino acids

Branched-chain amino acids (BCAAs) only comprise three amino acids: isoleucine, leucine, and valine. These molecules are essential, and primarily gained through host diet or synthesized by the microbiome. Many studies have linked the level of BCAAs with insulin resistance, obesity, and microbiome diversity [228][229][230]. There has been conflicting evidence on the benefits of elevated BCAA levels [231,232], and no comprehensive understanding of the relationship between BCAA and microbiome health is currently evident.

Fecal samples can be a non-invasive method to measure the BCAA content in the gut. However, considering BCAAs are often absorbed by intestinal tissues and transported throughout the body, fecal analysis may only give rough quantity estimates and no tissue or function-specific information. For this, BCAAs would need to be extracted from GI tract samples. BCAA levels have also been commonly measured in the blood samples [230][233]. Due to the polar nature of these amino acids, extraction from any sample type would need to be done with a polar solvent like methanol, water, or isopropanol (Table 5).

Table 5.

Branched-chain amino acid quantification methods.

Quantification Method Reference Sample Type Extraction Column Mobile Phase Ionization Method Molecule(s)
LC-MS Fingerhut et al., 2016 [231] Blood Neobase test-kit from PerkinElmer BEH C8 A: Water + 0.1% formic acid + 0.01% heptafluorobutyric acid
B: Acetonitrile + 0.1% formic acid + 0.01% heptafluorobutyric acid
ESI - Positive mode All BCAAs
Le et al., 2018 [46] Serum Acetonitrile Tested multiple, but C18-PFP worked best Tested multiple, but ended up using:
A: Water + 0.1% formic acid
B: Acetonitrile + 0.1% formic acid
ESI - Positive mode All BCAAs
GC-MS Zheng et al., 2013 [232] Feces
Plasma
Urine
Sodium hydroxide solutions HP-5ms capillary column Helium, flow rate 1 mL/min EI Propyl-ester derived BCAAs
CE-MS Piestanksky et al., 2021 [233] Standards
Plasma
Acetonitrile Fused silica capillary 50:50 Methanol:water + 5 mM ammonium acetate ESI - Positive mode All BCAAs

Detection of these three BCAAs is relatively straightforward. They can be separated by LC and are ionized well in positive-mode MS. It is important to note that leucine and isoleucine are structural isomers, with the same precursor m/z. MS2 fragmentation patterns need to be collected to accurately differentiate the two. Additionally, they tend to have very similar retention times, and often cannot be resolved by non-polar stationary phase columns. Using a more polar stationary phase column, like a HILIC column, BEH amide column, or a C18-PFP can help resolve these two molecules [46]. GC-MS methods have also been used for the detection of BCAAs, however they are less common and typically involve first derivatizing the BCAAs to propyl esters [234]. Beyond these MS instruments, other methods of BCAA detection can also be done. BCAAs can be detected with NMR [235], and even more novel methods like a paper strip test have been developed for analysis [236].

d. Tryptophan Derivatives

L-tryptophan is an essential amino acid that can be catabolized through one of three different metabolic pathways: the serotonin/5-hydroxytryptamine pathway, the indole-derivative pathways, or the kynurenine pathway, which makes up approximately 95% of tryptophan metabolism [240242]. The gut microbiome plays a crucial role in L-tryptophan metabolism. The primary direct role of the microbiome in tryptophan catabolism is through the direct and exclusive production of indole derivatives, although some species of bacteria are able to directly produce kynurenine as well [242245]. Mouse studies have shown that the indole derivatives indole-3-propionic acid and indoxyl sulfate are not produced without the presence of gut microbes and that germ-free mice had higher levels of tryptophan (the precursor of indole-3-propionic acid and indoxyl sulfate) and lower levels of kynurenine in their serum compared to the germ free mice [4,246]. Due to the complex variety of molecules produced from tryptophan in the gut, it is important to select sample types, extraction methods, and detection methods that will allow for the detection of metabolites of interest.

The majority of tryptophan derivatives are present in multiple sample types, but urine generally offers the largest number of unique tryptophan derivatives when compared to feces and plasma, with higher overall concentrations of almost all tryptophan derivatives when compared to plasma [45,247,248]. Additionally, some tryptophan derivatives can’t be detected in urine, such as 5-hydroxyindole (present in feces) and 3,4-dihydroxyphenyl acetic acid (present in plasma) [45,248]. For the broadest possible analyses of tryptophan derivatives, collection of both fecal and urine samples is recommended.

For extraction of tryptophan derivatives, biofluids like urine, plasma, and cerebrospinal fluid can be easily extracted using a “dilute-and-shoot” strategy of a 10 fold dilution with water [248,249] or by a simple protein precipitation procedure where ice cold methanol or acetonitrile is added to the biofluid samples and centrifuged, followed by collection of the supernatant [250,251] (Table 6). Tissue and fecal samples tend to follow a similar protein precipitation method, but they are homogenized first.

Table 6.

Tryptophan microbial metabolites quantification methods.

Quantification Method Reference Sample Type Extraction Column Mobile Phase Ionization Method Molecule(s)
LC-MS Marcobal et al., 2013 [45] Bifidobacterium longum NCC2705 Methanol C18 A: Water + 0.1% formic acid
B: Acetonitrile + 0.1% formic acid
ESI - Positive mode Indole-3-lactic acid
Urine Water + 10 mM ammonium formate C18 A: Water + 10 mm ammonium formate
B: Methanol + 10 mm ammonium formate
ESI - Positive and negative mode Indole derivatives
Water + 10 mM ammonium formate and ACN HILIC A: 10:90
Water:acetonitrile + 15 mm ammonium formate
B: 90:10
Water:acetonitrile + 15 mm ammonium formate
Anesi et al., 2019 [242] Plasma Ostro 96 well plate - 3:1 ACN with 0.1% formic acid C18 A: Water + 0.1% formic acid
B: Acetonitrile + 0.1% formic acid
ESI - Positive and negative mode Tryptophan-kynurenine pathway, serotonin pathway, indole derivatives
Urine Water with 0.1% formic acid C18 A: Water + 0.1% formic acid
B: Acetonitrile + 0.1% formic acid
ESI - Positive and negative mode Tryptophan-kynurenine pathway, serotonin pathway, indole derivatives
Lai et al., 2021 [241] Brain Tissue Methanol C18 A: Water + 0.1% formic acid
B: Acetonitrile + 0.1% formic acid
ESI - Positive mode Indole derivatives
Feces Methanol/water (1:1. v/v) C18 A: Water + 0.1% formic acid
B: Acetonitrile + 0.1% formic acid
ESI - Positive mode Indole derivatives
LC-Fluorescence Desbonnet et al., 2008 [249] Plasma 4M perchloric acid C18 97:3
Water:acetonitrile + 50 mM acetic acid + 100 mM Zinc acetate
N/A Tryptophan-Kynurenine pathway
GC-MS Zhao et al., 2017 [51] Serum
Urine
Methanol - Derivatized with alkyl chloroformate DB-5 MS Helium, flow rate 1.0 mL/min EI Indole derivatives

Tryptophan and its derivatives are primarily quantified using LC-MS. C18 columns tend to be favored since tryptophan derivatives have large non-polar indole groups or rings; in contrast, tryptophan derivatives were not retained well on a BEH amide HILIC column and HILIC columns did not provide reliable separation and peak shape for indole tryptophan derivatives [248,252]. For low volume samples or samples with high levels of matrix effects, tryptophan and its metabolites can be derivatized to increase sensitivity and improve ionization, but this naturally adds extra steps, which can be time-consuming for large sample batches or add additional variation [253,254]. Chromatography is most commonly performed with water and acetonitrile as mobile phases, both of which contain 0.1% formic acid to aid in the formation of ions. Tryptophan derivatives are almost universally ionized in positive mode; however, indoxyl sulfate ionizes better in negative mode [248,249]. Instead of using MS for detection, some studies have elected to use a fluorometer for detection after LC separation due to the native fluorescence of tryptophan [255258]. This provides increased sensitivity for tryptophan derivatives but does limit the ability to investigate non-fluorescent molecules without some form of derivatization. GC-MS has been used for the quantification of tryptophan derivatives but is traditionally more challenging due to their need for chemical derivatization [5153].

Due to the broad range of molecules that can be derived from tryptophan, quantifying several types of tryptophan derivatives simultaneously can be difficult. Tryptophan derivatives concentrations can cover 4 magnitudes of concentrations, each with their own ranges of linearity, making it challenging to find a sample concentration that will work for all potential metabolites of interest [247,248,252]. Additionally, tryptophan derivatives can have a variety of functional groups each with their own pKas, impacting the polarity of the molecule based on the pH of the solution it is in [247,252].

e. Trimethylamine N-oxide (TMAO)

Trimethylamine N-oxide (TMAO) is generated through oxidation of trimethylamine (TMA), a metabolite generated in the gut by the microbiome from carnitine, choline, and betaine that is transferred to the liver. In the liver, host flavin-dependent monooxygenase (FMO) isoforms 1 and 3 convert TMA into TMAO. The level of TMAO in the body is dependent upon factors such as diet, gender, natural flora in the gut, age, and medications [259]. High levels of TMAO are strongly correlated to the development of cardiovascular diseases, such as atherosclerosis, stroke, heart failure, and kidney disease [260].

The types of samples analyzed for TMAO quantification include plasma or urine [261][262]. A key consideration when quantifying TMAO is that levels differ between males and females, with males generally having lower TMAO levels than females [263].

TMAO is typically analyzed using LC-MS, with GC-MS and NMR methods used less commonly (Table 7, Table 8) [46][264]. One could even use colorimetric array-based sensors, which is newer and not as explored as other methods [265]. For LC-MS, the type of column used to quantify TMAO is typically a C18 column, though Boyce et al (2019) suggested higher resolution using a C18-PFP (pentafluorophenyl) column [46][266][267]. Another column commonly used is a HILIC column [262].

Table 7.

MS-based trimethylamine N-oxide quantification methods.

Quantification Method Reference Sample Type Extraction Column Mobile Phase Ionization Method Molecule(s)
LC-MS Le et al., 2018 [46] Serum Acetonitrile C18 C18-PFP HILIC A: Water + 0.1% formic acid
B: Acetonitrile + 0.1% formic acid
ESI - Positive mode TMAO
Hefni et al., 2021 [256] Urine Water. Derivatizatized with iodoacetonitrile in acetonitrile:methanol (9:1. v/v) HILIC 30:70
Water:acetonitrile + 25 mM ammonium formate
ESI - Positive mode TMAO
Plasma Not diluted. Derivatizatized with iodoacetonitrile in acetonitrile:methanol (9:1. v/v)
GC-MS Bain et al., 2004 [262] Plasma Acidification with 0.01 M HCl before adding water. Addition of 5M KOH to improve volatilization SPB-1 Helium, flow rate 60 mL/min EI TMAO

Table 8.

NMR-based trimethylamine N-oxide quantification methods.

Quantitation Method Reference Sample Type Solutions NMR Molecule(s)
1H-NMR Garcia et al., 2017 [258] Plasma Aqueous citrate buffer (pH=4.4) 400 MHz with a 4-mm indirect detection probe TMAO

f. N-acyl-amides

The microbiome creates N-acyl amides (NAAs) that closely mimic the function of endogenously produced signaling molecules, particularly endocannabinoids. The microbiome is capable of producing 6 families of N-acyl amides: N-acyl glycine, N-acyloxyacyl lysine, N-acyloxyacyl glutamine, N-acyl lysine/ornithine, N-acyl alanine, and N-acyl serinol [269]. These NAA mimics can serve as agonists or antagonists of G-protein-coupled receptors (GPCR). For instance, Neisseria meningitidis, the bacteria responsible for meningococcal disease, produces N-3-hydroxypalmitoyl-ornithine which serves selectively as an agonist for sphingosine-1-phosphate receptor 4 (S1PR4), a GPCR that has a role in human inflammatory responses [269,270].

Sample type can impact the NAAs detected in a study. While the majority of detected N-acyl homoserine molecules are present in serum, cecal content, and liver samples, some N-acyl homoserines are detected exclusively in one sample type, such as 3OH-C10-HSL only being detected in serum and C12-HSL only being detected in liver and cecal content [47]. As a result, when collecting in vivo samples, it may be beneficial to collect at least one biofluid and one tissue sample based on the NAA of interest.

NAAs can be detected from bacterial cultures or from tissue samples. For in vitro studies, a simple extraction can be performed using equal volumes of ethyl acetate followed by aspiration of ethyl acetate upon separation [269,271] or using n-butanol [272] (Table 9). For serum and tissues, samples are typically extracted and homogenized using cold methanol or acetonitrile and then purified using a C18 solid phase extraction column to reduce matrix effects [47,273,274]. After extraction and vacuum drying, NAA samples should be resuspended with 50% to 100% methanol. 100% MeOH was the most commonly used extraction solvent, but different NAAs favor different ratios of MeOH to water [275]. Additionally, some researchers elect to acidify their resuspension solvent [276,277]. Acidification can help with the dissolution and ionization of charged NAAs as well as with sample stability, but can cause hydrolysis of any ester bonds present in molecules of interest if left at acidic conditions for prolonged periods [278].

Table 9.

N-acyl amide quantification methods.

Quantification Method Reference Sample Type Extraction Column Mobile Phase Ionization Method Molecule(s)
LC-S Cohen et al., 2017 [263] Bacterial cultures Ethyl acetate C18 A: Water + 0.1% formic acid
B: Acetonitrile + 0.1% formic acid
ESI - Positive and negative mode N-acyl glycines, N-acyloxyacyl lysines, N-acyloxyacyl glutamines, N-acyl lysines/ornithines, N-acyl alanines, N-acyl serinols
Tørring et al., 2017 [266] Bacterial cultures n-butanol C18 A: Water + 0.1% formic acid
B: Acetonitrile + 0.1% formic acid
ESI - Positive mode N-acyl histidines
Xue et al., 2021 [47] Serum Methanol + 0.1% formic acid. Mixture preconditioned with a C18 solid phase extraction column. C18 A: Water + 0.1% formic acid
B: Acetonitrile + 0.1% formic acid
ESI - Positive mode N-acyl homoserines
Liver
Cecum
Methanol, followed by dichloromethane:methanol (1:1. v/v). Mixture preconditioned with a C18 solid phase extraction column. C18 A: Water + 0.1% formic acid
B: Acetonitrile + 0.1% formic acid
ESI - Positive mode N-acyl homoserines
Tan et al., 2009 [267] Brain Methanol followed by water. Preconditioned with a C18 solid phase extraction column. C18 A: 80:20
Water:methanol + 1 mM ammonium acetate
B: 100% methanol + 1 mM ammonium acetate
ESI - Positive and negative mode N-acyl amino acids

NAAs have long lipid chains, so they have historically been analyzed using methods that are suitable for the detection of lipids, particularly LC-MS. For the LC separation, NAAs are analyzed using a C18 column due to the long acyl chains that are characteristic of the class. To correct for the effects of extraction, resuspension, and ESI ionization on quantification, labeled internal standards for the NAAs of interest should be suspended in the earliest extraction solvent (ng/mL scale [277,279,280]). Due to the large variety of NAAs, a labeled version of the NAA of interest might not always be available. In that case, the extraction solvent should contain at least one labeled NAA from each of the 6 families mentioned previously.

NAAs can be challenging to quantify. Some NAAs have shown clear signs of matrix suppression, making solid phase extraction a crucial step in the preparation of many NAAs [47]. Additionally, NAA concentration can be as low as pg/g of sample, making sensitive techniques or purification required in order to detect them [273,281]. NAAs are a broad class of molecules, as there are a large variety of acyl chains that each molecule can use as their acyl group. Some acyl groups may be unsaturated. While MS can demonstrate the presence of a double bond, elucidating the location of the double bond is challenging and better suited for a method like NMR. No extraction method will universally extract all NAAs well, so it is important to carefully consider which NAAs are of primary interest before selecting an extraction method.

6. Conclusion

The microbiome has a vast role in host metabolism through its production and modification of small molecules. Growing knowledge of this role has led to increased needs for quantification of microbial metabolism, though challenges remain (Box 1). Samples should be acquired using a method that minimizes contaminants and stored to prevent their degradation, but the chosen methods may depend on the specific molecules being investigated. Many instruments and types of analysis are available for quantification, each with its own use cases and limitations. SCFAs and bile acids are some of the most commonly analyzed molecules, but other classes have biological relevance of their own. Furthermore, increasing implementation of untargeted approaches is leading to the discovery of novel microbial metabolites.

Box 1 -. Challenges and opportunities.

  1. One-size-fits-all approach may not be suitable. Different metabolite classes often necessitate different approaches. Thus, a single method is unlikely to enable quantification of all microbial metabolites that may be of interest in a given study. Untargeted analysis to discover novel microbial metabolites in particular usually necessitates choices with regards to extraction solvents, chromatography, ionization, etc., that shape the properties of the molecules that can be detected.

  2. Coverage. Even though ‘omics” pipelines aim to cover all analytes, this is not achievable in metabolomics. Combining approaches, including extraction and instrumental methods, will increase coverage but also cost and complexity of acquisition and processing.

  3. Lack of consensus about methods. In parallel with the necessary requirement for diverse methods for different metabolite classes and for broad metabolite coverage, there is also sometimes considerable variation in methods implemented between research groups, even for the same metabolite class (see the multiple approaches described in this review’s tables). This can make comparisons between studies and between research groups challenging, especially for untargeted studies or studies that implement only relative quantification in a targeted workflow.

  4. Improved automation for sample preparation and metabolite extraction. Sample preparation for metabolomics experiments lags behind sequencing preparation workflows as well as metabolomics data acquisition steps. Improved automation at this step would increase reproducibility. This is a particular need with regards to tissue analysis.

  5. Lack of consensus about data processing pipelines. In addition to variability in sample preparation, there are often considerable differences in data processing between groups.

  6. Spatial distribution. Microbiome composition will differ between organs, sites, and even on the microscopic level. Likewise, microbial metabolism is context-dependent and will differ from the microscopic to the macroscopic scale. This represents a challenge, in that spatial analysis is required to obtain comprehensive information about microbiota metabolism. However, this spatiality is also an opportunity to better understand regulators of metabolism and how they affect health.

  7. Annotation challenges. The discovery of novel microbial metabolites is hampered by database biases, which may favor mammalian metabolites to the detriment of microbial metabolites. However, there is currently considerable effort being expended to address this issue. Structure elucidation is often labor-intensive and costly, but this is critical to go beyond known molecules and increase biological insight.

  8. Challenges of confirming a microbial source for a novel metabolite. Microbial metabolite production may be context-dependent. Thus, production may not always be observed in the artificial monoculture settings that are often used to confirm the producing organism. New culture techniques are helping address this issue.

  9. Better integration between microbiome metabolomics and the natural product field, especially with regards to workflows for the discovery of novel microbial metabolites. A major strength of natural product chemistry is its focus on de novo elucidation of novel structures, including those produced by microorganisms. However, there is sometimes a lack of interaction between metabolome researchers and natural product chemists.

  10. Lack of commercially available standards. This can hamper confirmation of structural annotations, as well as data normalization and cross-study comparisons.

  11. Method cost. Most microbial metabolite quantification techniques are costly and may be labor-intensive. Cheaper methods including point of care approaches, test strips, and at-home options will be critical to ensure personalized medicine applications.

This review focused on molecular classes produced by microbiome bacteria, but more is being discovered about metabolites produced by other microbiome members. For instance, the presence of the protozoa Blastocystis has been associated with higher abundance of archaea like Methanobrevibacter smithii; M. smithii, in turn, supports human health by removing end products of bacterial fermentation which supports glycan digestion and helps to increase butyrate production [282].

Microbial metabolism is an expansive topic given the complexities of its composition and methods of analysis. Indeed, there are many unique specialized metabolites that we were regretfully unable to cover here due to a lack of space, which include, but are not limited to piericidins [283,284], p-cresol [285,286], and exopolysaccharides [287,288]. While methods for quantifying these classes and others are not explicitly mentioned here, quantitative methods can be inferred from this review based on structural similarities. For example, in the case of p-cresol, methods highlighted for kynurenine and its derivatives would likely work well.

Tools at each step of the process are continuing to be developed, which will lead to improved application to medicine, especially with the growing need to tailor health approaches to the individual. As this area of research expands, more light will be shed on the best practices for microbiome metabolism quantification and its applicability to human health. A key ongoing and future step will be to go from quantification to functional insight, and to the development of new interventions.

Highlights.

  • Major role of microbial metabolites in human health

  • Critical impact of analytical choices in microbial metabolite quantification

  • General vs class-specific analytical approaches

  • Opportunities to identify novel microbial metabolites

  • Analytical advances addressing challenges in microbial metabolite quantification

Acknowledgements

Research on microbial metabolites in the McCall laboratory is supported by the National Institute of General Medical Sciences of the National Institutes of Health under Award Number R01GM145649. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • [1].Martin F-PJ, Dumas M-E, Wang Y, Legido-Quigley C, Yap IKS, Tang H, Zirah S, Murphy GM, Cloarec O, Lindon JC, Sprenger N, Fay LB, Kochhar S, van Bladeren P, Holmes E, Nicholson JK, A top-down systems biology view of microbiome-mammalian metabolic interactions in a mouse model, Mol. Syst. Biol 3 (2007) 112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Vemuri R, Shankar EM, Chieppa M, Eri R, Kavanagh K, Beyond Just Bacteria: Functional Biomes in the Gut Ecosystem Including Virome, Mycobiome, Archaeome and Helminths, Microorganisms. 8 (2020). 10.3390/microorganisms8040483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Koppel N, Maini Rekdal V, Balskus EP, Chemical transformation of xenobiotics by the human gut microbiota, Science. 356 (2017). 10.1126/science.aag2770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Wikoff WR, Anfora AT, Liu J, Schultz PG, Lesley SA, Peters EC, Siuzdak G, Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites, Proc. Natl. Acad. Sci. U. S. A 106 (2009) 3698–3703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Ueyama J, Nadai M, Kanazawa H, Iwase M, Nakayama H, Hashimoto K, Yokoi T, Baba K, Takagi K, Takagi K, Hasegawa T, Endotoxin from various gram-negative bacteria has differential effects on function of hepatic cytochrome P450 and drug transporters, Eur. J. Pharmacol 510 (2005) 127–134. [DOI] [PubMed] [Google Scholar]
  • [6].Cryan JF, O’Mahony SM, The microbiome-gut-brain axis: from bowel to behavior, Neurogastroenterol. Motil 23 (2011) 187–192. [DOI] [PubMed] [Google Scholar]
  • [7].Zhang L, Wang Y, Xiayu X, Shi C, Chen W, Song N, Fu X, Zhou R, Xu Y-F, Huang L, Zhu H, Han Y, Qin C, Altered Gut Microbiota in a Mouse Model of Alzheimer’s Disease, J. Alzheimers. Dis 60 (2017) 1241–1257. [DOI] [PubMed] [Google Scholar]
  • [8].Weir TL, Manter DK, Sheflin AM, Barnett BA, Heuberger AL, Ryan EP, Stool microbiome and metabolome differences between colorectal cancer patients and healthy adults, PLoS One. 8 (2013) e70803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Uchimura Y, Fuhrer T, Li H, Lawson MA, Zimmermann M, Yilmaz B, Zindel J, Ronchi F, Sorribas M, Hapfelmeier S, Ganal-Vonarburg SC, Gomez de Agüero M, McCoy KD, Sauer U, Macpherson AJ, Antibodies Set Boundaries Limiting Microbial Metabolite Penetration and the Resultant Mammalian Host Response, Immunity. 49 (2018) 545–559.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Wishart DS, Oler E, Peters H, Guo A, Girod S, Han S, Saha S, Lui VW, LeVatte M, Gautam V, Kaddurah-Daouk R, Karu N, MiMeDB: the Human Microbial Metabolome Database, Nucleic Acids Res. 51 (2023) D611–D620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Quinn RA, Melnik AV, Vrbanac A, Fu T, Patras KA, Christy MP, Bodai Z, Belda-Ferre P, Tripathi A, Chung LK, Downes M, Welch RD, Quinn M, Humphrey G, Panitchpakdi M, Weldon KC, Aksenov A, da Silva R, Avila-Pacheco J, Clish C, Bae S, Mallick H, Franzosa EA, Lloyd-Price J, Bussell R, Thron T, Nelson AT, Wang M, Leszczynski E, Vargas F, Gauglitz JM, Meehan MJ, Gentry E, Arthur TD, Komor AC, Poulsen O, Boland BS, Chang JT, Sandborn WJ, Lim M, Garg N, Lumeng JC, Xavier RJ, Kazmierczak BI, Jain R, Egan M, Rhee KE, Ferguson D, Raffatellu M, Vlamakis H, Haddad GG, Siegel D, Huttenhower C, Mazmanian SK, Evans RM, Nizet V, Knight R, Dorrestein PC, Global chemical effects of the microbiome include new bile-acid conjugations, Nature. 579 (2020) 123–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Lucas LN, Barrett K, Kerby RL, Zhang Q, Cattaneo LE, Stevenson D, Rey FE, Amador-Noguez D, Dominant Bacterial Phyla from the Human Gut Show Widespread Ability To Transform and Conjugate Bile Acids, mSystems. (2021) e0080521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Wang M, Jarmusch AK, Vargas F, Aksenov AA, Gauglitz JM, Weldon K, Petras D, da Silva R, Quinn R, Melnik AV, van der Hooft JJJ, Caraballo-Rodríguez AM, Nothias LF, Aceves CM, Panitchpakdi M, Brown E, Di Ottavio F, Sikora N, Elijah EO, Labarta-Bajo L, Gentry EC, Shalapour S, Kyle KE, Puckett SP, Watrous JD, Carpenter CS, Bouslimani A, Ernst M, Swafford AD, Zúñiga EI, Balunas MJ, Klassen JL, Loomba R, Knight R, Bandeira N, Dorrestein PC, Mass spectrometry searches using MASST, Nat. Biotechnol 38 (2020) 23–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt EB, Fu X, Chung Y-M, Wu Y, Schauer P, Smith JD, Allayee H, Tang WHW, DiDonato JA, Lusis AJ, Hazen SL, Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease, Nature. 472 (2011) 57–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Cason CA, Dolan KT, Sharma G, Tao M, Kulkarni R, Helenowski IB, Doane BM, Avram MJ, McDermott MM, Chang EB, Ozaki CK, Ho KJ, Plasma microbiome modulated indole- and phenyl-derived metabolites associate with advanced atherosclerosis and postoperative outcomes, J. Vasc. Surg 68 (2018) 1552–1562.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Cussotto S, Delgado I, Anesi A, Dexpert S, Aubert A, Beau C, Forestier D, Ledaguenel P, Magne E, Mattivi F, Capuron L, Tryptophan Metabolic Pathways Are Altered in Obesity and Are Associated With Systemic Inflammation, Front. Immunol 11 (2020) 557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Wilson MR, Jiang Y, Villalta PW, Stornetta A, Boudreau PD, Carrá A, Brennan CA, Chun E, Ngo L, Samson LD, Engelward BP, Garrett WS, Balbo S, Balskus EP, The human gut bacterial genotoxin colibactin alkylates DNA, Science. 363 (2019). 10.1126/science.aar7785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Mehta RS, Mayers JR, Zhang Y, Bhosle A, Glasser NR, Nguyen LH, Ma W, Bae S, Branck T, Song K, Sebastian L, Pacheco JA, Seo H-S, Clish C, Dhe-Paganon S, Ananthakrishnan AN, Franzosa EA, Balskus EP, Chan AT, Huttenhower C, Gut microbial metabolism of 5-ASA diminishes its clinical efficacy in inflammatory bowel disease, Nat. Med 29 (2023) 700–709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Spanogiannopoulos P, Kyaw TS, Guthrie BGH, Bradley PH, Lee JV, Melamed J, Malig YNA, Lam KN, Gempis D, Sandy M, Kidder W, Van Blarigan EL, Atreya CE, Venook A, Gerona RR, Goga A, Pollard KS, Turnbaugh PJ, Host and gut bacteria share metabolic pathways for anti-cancer drug metabolism, Nat Microbiol. 7 (2022) 1605–1620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].He Z, Chen L, Catalan-Dibene J, Bongers G, Faith JJ, Suebsuwong C, DeVita RJ, Shen Z, Fox JG, Lafaille JJ, Furtado GC, Lira SA, Food colorants metabolized by commensal bacteria promote colitis in mice with dysregulated expression of interleukin-23, Cell Metab. 33 (2021) 1358–1371.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Zimmermann M, Zimmermann-Kogadeeva M, Wegmann R, Goodman AL, Separating host and microbiome contributions to drug pharmacokinetics and toxicity, Science. 363 (2019). 10.1126/science.aat9931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Gratton J, Phetcharaburanin J, Mullish BH, Williams HRT, Thursz M, Nicholson JK, Holmes E, Marchesi JR, Li JV, Optimized Sample Handling Strategy for Metabolic Profiling of Human Feces, Anal. Chem 88 (2016) 4661–4668. [DOI] [PubMed] [Google Scholar]
  • [23].Phua LC, Koh PK, Cheah PY, Ho HK, Chan ECY, Global gas chromatography/time-of-flight mass spectrometry (GC/TOFMS)-based metabonomic profiling of lyophilized human feces, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci 937 (2013) 103–113. [DOI] [PubMed] [Google Scholar]
  • [24].Rotter M, Brandmaier S, Prehn C, Adam J, Rabstein S, Gawrych K, Brüning T, Illig T, Lickert H, Adamski J, Wang-Sattler R, Stability of targeted metabolite profiles of urine samples under different storage conditions, Metabolomics. 13 (2017) 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Anton G, Wilson R, Yu Z-H, Prehn C, Zukunft S, Adamski J, Heier M, Meisinger C, Römisch-Margl W, Wang-Sattler R, Hveem K, Wolfenbuttel B, Peters A, Kastenmüller G, Waldenberger M, Pre-analytical sample quality: metabolite ratios as an intrinsic marker for prolonged room temperature exposure of serum samples, PLoS One. 10 (2015) e0121495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Bolten CJ, Kiefer P, Letisse F, Portais J-C, Wittmann C, Sampling for Metabolome Analysis of Microorganisms, Analytical Chemistry. 79 (2007) 3843–3849. 10.1021/ac0623888. [DOI] [PubMed] [Google Scholar]
  • [27].Fuhrer T, Heer D, Begemann B, Zamboni N, High-Throughput, Accurate Mass Metabolome Profiling of Cellular Extracts by Flow Injection–Time-of-Flight Mass Spectrometry, Analytical Chemistry. 83 (2011) 7074–7080. 10.1021/ac201267k. [DOI] [PubMed] [Google Scholar]
  • [28].Ewald JC, Heux S, Zamboni N, High-Throughput Quantitative Metabolomics: Workflow for Cultivation, Quenching, and Analysis of Yeast in a Multiwell Format, Analytical Chemistry. 81 (2009) 3623–3629. 10.1021/ac900002u. [DOI] [PubMed] [Google Scholar]
  • [29].Yin P, Peter A, Franken H, Zhao X, Neukamm SS, Rosenbaum L, Lucio M, Zell A, Häring HU, Xu G, Lehmann R, Preanalytical aspects and sample quality assessment in metabolomics studies of human blood, Clin. Chem 59 (2013) 833–845. [DOI] [PubMed] [Google Scholar]
  • [30].Loftfield E, Vogtmann E, Sampson JN, Moore SC, Nelson H, Knight R, Chia N, Sinha R, Comparison of Collection Methods for Fecal Samples for Discovery Metabolomics in Epidemiologic Studies, Cancer Epidemiol. Biomarkers Prev 25 (2016) 1483–1490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Wang Z, Zolnik CP, Qiu Y, Usyk M, Wang T, Strickler HD, Isasi CR, Kaplan RC, Kurland IJ, Qi Q, Burk RD, Comparison of Fecal Collection Methods for Microbiome and Metabolomics Studies, Front. Cell. Infect. Microbiol 8 (2018) 301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Sinha R, Chen J, Amir A, Vogtmann E, Shi J, Inman KS, Flores R, Sampson J, Knight R, Chia N, Collecting Fecal Samples for Microbiome Analyses in Epidemiology Studies, Cancer Epidemiol. Biomarkers Prev 25 (2016) 407–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Kelly AE, Ou HD, Withers R, Dötsch V, Low-conductivity buffers for high-sensitivity NMR measurements, J. Am. Chem. Soc 124 (2002) 12013–12019. [DOI] [PubMed] [Google Scholar]
  • [34].Ramamoorthy S, Levy S, Mohamed M, Abdelghani A, Evans AM, Miller LAD, Mehta L, Moore S, Freinkman E, Hourigan SK, An ambient-temperature storage and stabilization device performs comparably to flash-frozen collection for stool metabolomics in infants, BMC Microbiol. 21 (2021) 59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Denniff P, Spooner N, Volumetric absorptive microsampling: a dried sample collection technique for quantitative bioanalysis, Anal. Chem 86 (2014) 8489–8495. [DOI] [PubMed] [Google Scholar]
  • [36].Qu Y, Brady K, Apilado R, O’Malley T, Reddy S, Chitkara P, Ibarra C, Alexander RV, Dervieux T, Capillary blood collected on volumetric absorptive microsampling (VAMS) device for monitoring hydroxychloroquine in rheumatoid arthritis patients, J. Pharm. Biomed. Anal 140 (2017) 334–341. [DOI] [PubMed] [Google Scholar]
  • [37].Volani C, Caprioli G, Calderisi G, Sigurdsson BB, Rainer J, Gentilini I, Hicks AA, Pramstaller PP, Weiss G, Smarason SV, Paglia G, Pre-analytic evaluation of volumetric absorptive microsampling and integration in a mass spectrometry-based metabolomics workflow, Anal. Bioanal. Chem 409 (2017) 6263–6276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Kok MGM, Nix C, Nys G, Fillet M, Targeted metabolomics of whole blood using volumetric absorptive microsampling, Talanta. 197 (2019) 49–58. [DOI] [PubMed] [Google Scholar]
  • [39].Protti M, Mandrioli R, Mercolini L, Tutorial: Volumetric absorptive microsampling (VAMS), Anal. Chim. Acta 1046 (2019) 32–47. [DOI] [PubMed] [Google Scholar]
  • [40].Protti M, Catapano MC, Samolsky Dekel BG, Rudge J, Gerra G, Somaini L, Mandrioli R, Mercolini L, Determination of oxycodone and its major metabolites in haematic and urinary matrices: Comparison of traditional and miniaturised sampling approaches, J. Pharm. Biomed. Anal 152 (2018) 204–214. [DOI] [PubMed] [Google Scholar]
  • [41].Saric J, Wang Y, Li J, Coen M, Utzinger J, Marchesi JR, Keiser J, Veselkov K, Lindon JC, Nicholson JK, Holmes E, Species variation in the fecal metabolome gives insight into differential gastrointestinal function, J. Proteome Res 7 (2008) 352–360. [DOI] [PubMed] [Google Scholar]
  • [42].Stevens VL, Hoover E, Wang Y, Zanetti KA, Pre-Analytical Factors that Affect Metabolite Stability in Human Urine, Plasma, and Serum: A Review, Metabolites. 9 (2019). 10.3390/metabo9080156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Ottosson F, Russo F, Abrahamsson A, MacSween N, Courraud J, Nielsen ZK, Hougaard DM, Cohen AS, Ernst M, Effects of Long-Term Storage on the Biobanked Neonatal Dried Blood Spot Metabolome, J. Am. Soc. Mass Spectrom 34 (2023) 685–694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Ulmer CZ, Koelmel JP, Jones CM, Garrett TJ, Aristizabal-Henao JJ, Vesper HW, Bowden JA, A Review of Efforts to Improve Lipid Stability during Sample Preparation and Standardization Efforts to Ensure Accuracy in the Reporting of Lipid Measurements, Lipids. 56 (2021) 3–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Marcobal A, Kashyap PC, Nelson TA, Aronov PA, Donia MS, Spormann A, Fischbach MA, Sonnenburg JL, A metabolomic view of how the human gut microbiota impacts the host metabolome using humanized and gnotobiotic mice, ISME J. 7 (2013) 1933–1943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Le TT, Shafaei A, Genoni A, Christophersen C, Devine A, Lo J, Wall PL, Boyce MC, 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 411 (2018) 1019–1028. [DOI] [PubMed] [Google Scholar]
  • [47].Xue J, Chi L, Tu P, Lai Y, Liu C-W, Ru H, Lu K, Detection of gut microbiota and pathogen produced N-acyl homoserine in host circulation and tissues, NPJ Biofilms Microbiomes. 7 (2021) 53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].De Baere S, Eeckhaut V, Steppe M, De Maesschalck C, De Backer P, Van Immerseel F, Croubels S, Development of a HPLC–UV method for the quantitative determination of four short-chain fatty acids and lactic acid produced by intestinal bacteria during in vitro fermentation, J. Pharm. Biomed. Anal 80 (2013) 107–115. [DOI] [PubMed] [Google Scholar]
  • [49].Zhang C, Tang P, Xu H, Weng Y, Tang Q, Zhao H, Analysis of Short-Chain Fatty Acids in Fecal Samples by Headspace-Gas Chromatography, Chromatographia. 81 (2018) 1317–1323. [Google Scholar]
  • [50].Zhao R, Chu L, Wang Y, Song Y, Liu P, Li C, Huang J, Kang X, Application of packed-fiber solid-phase extraction coupled with GC–MS for the determination of short-chain fatty acids in children’s urine, Clin. Chim. Acta 468 (2017) 120–125. [DOI] [PubMed] [Google Scholar]
  • [51].Zhao L, Ni Y, Su M, Li H, Dong F, Chen W, Wei R, Zhang L, Guiraud SP, Martin F-P, Rajani C, Xie G, Jia W, High Throughput and Quantitative Measurement of Microbial Metabolome by Gas Chromatography/Mass Spectrometry Using Automated Alkyl Chloroformate Derivatization, Anal. Chem 89 (2017) 5565–5577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Dong F, Hao F, Murray IA, Smith PB, Koo I, Tindall AM, Kris-Etherton PM, Gowda K, Amin SG, Patterson AD, Perdew GH, Intestinal microbiota-derived tryptophan metabolites are predictive of Ah receptor activity, Gut Microbes. 12 (2020) 1–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Sadok I, Gamian A, Staniszewska MM, Chromatographic analysis of tryptophan metabolites, J. Sep. Sci 40 (2017) 3020–3045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Beale DJ, Pinu FR, Kouremenos KA, Poojary MM, Narayana VK, Boughton BA, Kanojia K, Dayalan S, Jones OAH, Dias DA, Review of recent developments in GC-MS approaches to metabolomics-based research, Metabolomics. 14 (2018) 152. [DOI] [PubMed] [Google Scholar]
  • [55].Xu K, Guo Y, Wang Y, Ren Y, Low V, Cho S, Ping L, Peng K, Li X, Qiu Y, Liu Q, Li Z, Wang Z, Decreased Enterobacteriaceae translocation due to gut microbiota remodeling mediates the alleviation of premature aging by a high-fat diet, Aging Cell. 22 (2023) e13760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Gerothanassis IP, Troganis A, Exarchou V, Barbarossou K, NUCLEAR MAGNETIC RESONANCE (NMR) SPECTROSCOPY: BASIC PRINCIPLES AND PHENOMENA, AND THEIR APPLICATIONS TO CHEMISTRY, BIOLOGY AND MEDICINE, Chem. Educ. Res. Pract 3 (2002) 229–252. [Google Scholar]
  • [57].Bervoets L, Ippel JH, Smolinska A, van Best N, Savelkoul PHM, Mommers MAH, Penders J, Practical and Robust NMR-Based Metabolic Phenotyping of Gut Health in Early Life, J. Proteome Res 20 (2021) 5079–5087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Lamichhane S, Yde CC, Schmedes MS, Jensen HM, Meier S, Bertram HC, Strategy for Nuclear-Magnetic-Resonance-Based Metabolomics of Human Feces, Anal. Chem 87 (2015) 5930–5937. [DOI] [PubMed] [Google Scholar]
  • [59].Iwasaki Y, Sawada T, Hatayama K, Ohyagi A, Tsukuda Y, Namekawa K, Ito R, Saito K, Nakazawa H, Separation technique for the determination of highly polar metabolites in biological samples, Metabolites. 2 (2012) 496–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Nagana Gowda GA, Gowda YN, Raftery D, Expanding the limits of human blood metabolite quantitation using NMR spectroscopy, Anal. Chem 87 (2015) 706–715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Mora-Ortiz M, Nuñez Ramos P, Oregioni A, Claus SP, NMR metabolomics identifies over 60 biomarkers associated with Type II Diabetes impairment in db/db mice, Metabolomics. 15 (2019) 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Bharti SK, Roy R, Quantitative 1H NMR spectroscopy, Trends Analyt. Chem 35 (2012) 5–26. [Google Scholar]
  • [63].Kostidis S, Addie RD, Morreau H, Mayboroda OA, Giera M, Quantitative NMR analysis of intra- and extracellular metabolism of mammalian cells: A tutorial, Anal. Chim. Acta 980 (2017) 1–24. [DOI] [PubMed] [Google Scholar]
  • [64].Emwas A-HM, The strengths and weaknesses of NMR spectroscopy and mass spectrometry with particular focus on metabolomics research, Methods Mol. Biol 1277 (2015) 161–193. [DOI] [PubMed] [Google Scholar]
  • [65].Gruetter R, Weisdorf SA, Rajanayagan V, Terpstra M, Merkle H, Truwit CL, Garwood M, Nyberg SL, Ugurbil K, Resolution Improvements in in Vivo 1H NMR Spectra with Increased Magnetic Field Strength, J. Magn. Reson 135 (1998) 260–264. [DOI] [PubMed] [Google Scholar]
  • [66].Keun HC, Beckonert O, Griffin JL, Richter C, Moskau D, Lindon JC, Nicholson JK, Cryogenic probe 13C NMR spectroscopy of urine for metabonomic studies, Anal. Chem 74 (2002) 4588–4593. [DOI] [PubMed] [Google Scholar]
  • [67].Grimes JH, O’Connell TM, The application of micro-coil NMR probe technology to metabolomics of urine and serum, J. Biomol. NMR 49 (2011) 297–305. [DOI] [PubMed] [Google Scholar]
  • [68].Ferrer M, Raczkowska BA, Martínez-Martínez M, Barbas C, Rojo D, Phenotyping of gut microbiota: Focus on capillary electrophoresis, Electrophoresis. 38 (2017) 2275–2286. [DOI] [PubMed] [Google Scholar]
  • [69].Mishima E, Fukuda S, Mukawa C, Yuri A, Kanemitsu Y, Matsumoto Y, Akiyama Y, Fukuda NN, Tsukamoto H, Asaji K, Shima H, Kikuchi K, Suzuki C, Suzuki T, Tomioka Y, Soga T, Ito S, Abe T, Evaluation of the impact of gut microbiota on uremic solute accumulation by a CE-TOFMS-based metabolomics approach, Kidney Int. 92 (2017) 634–645. [DOI] [PubMed] [Google Scholar]
  • [70].Pruski P, Correia GDS, Lewis HV, Capuccini K, Inglese P, Chan D, Brown RG, Kindinger L, Lee YS, Smith A, Marchesi J, McDonald JAK, Cameron S, Alexander-Hardiman K, David AL, Stock SJ, Norman JE, Terzidou V, Teoh TG, Sykes L, Bennett PR, Takats Z, MacIntyre DA, Direct on-swab metabolic profiling of vaginal microbiome host interactions during pregnancy and preterm birth, Nat. Commun 12 (2021) 5967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Ayala-Ortiz C, Graf-Grachet N, Freire-Zapata V, Fudyma J, Hildebrand G, AminiTabrizi R, Howard-Varona C, Corilo YE, Hess N, Duhaime MB, Sullivan MB, Tfaily MM, MetaboDirect: an analytical pipeline for the processing of FT-ICR MS-based metabolomic data, Microbiome. 11 (2023) 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Moree WJ, Phelan VV, Wu C-H, Bandeira N, Cornett DS, Duggan BM, Dorrestein PC, Interkingdom metabolic transformations captured by microbial imaging mass spectrometry, Proc. Natl. Acad. Sci. U. S. A 109 (2012) 13811–13816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Sarmad S, Viant MR, Dunn WB, Goodacre R, Wilson ID, Chappell KE, Griffin JL, O’Donnell VB, Naicker B, Lewis MR, Suzuki T, A proposed framework to evaluate the quality and reliability of targeted metabolomics assays from the UK Consortium on Metabolic Phenotyping (MAP/UK), Nat. Protoc 18 (2023) 1017–1027. [DOI] [PubMed] [Google Scholar]
  • [74].Kirwan JA, Gika H, Beger RD, Bearden D, Dunn WB, Goodacre R, Theodoridis G, Witting M, Yu L-R, Wilson ID, metabolomics Quality Assurance and Quality Control Consortium (mQACC), Quality assurance and quality control reporting in untargeted metabolic phenotyping: mQACC recommendations for analytical quality management, Metabolomics. 18 (2022) 70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Bauermeister A, Mannochio-Russo H, Costa-Lotufo LV, Jarmusch AK, Dorrestein PC, Mass spectrometry-based metabolomics in microbiome investigations, Nat. Rev. Microbiol 20 (2022) 143–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Debik J, Sangermani M, Wang F, Madssen TS, Giskeødegård GF, Multivariate analysis of NMR-based metabolomic data, NMR Biomed. 35 (2022) e4638. [DOI] [PubMed] [Google Scholar]
  • [77].Alseekh S, Aharoni A, Brotman Y, Contrepois K, D’Auria J, Ewald JC, Ewald J, Fraser PD, Giavalisco P, Hall RD, Heinemann M, Link H, Luo J, Neumann S, Nielsen J, Perez de Souza L, Saito K, Sauer U, Schroeder FC, Schuster S, Siuzdak G, Skirycz A, Sumner LW, Snyder MP, Tang H, Tohge T, Wang Y, Wen W, Wu S, Xu G, Zamboni N, Fernie AR, Mass spectrometry-based metabolomics: a guide for annotation, quantification and best reporting practices, Nat. Methods 18 (2021) 747–756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Van Der Hooft J, Hanhineva K, Metabolomics Data Processing and Data Analysis-Current Best Practices, Mdpi AG, 2021. [Google Scholar]
  • [79].Pang Z, Zhou G, Ewald J, Chang L, Hacariz O, Basu N, Xia J, Using MetaboAnalyst 5.0 for LC-HRMS spectra processing, multi-omics integration and covariate adjustment of global metabolomics data, Nat. Protoc 17 (2022) 1735–1761. [DOI] [PubMed] [Google Scholar]
  • [80].Poland JC, Leaptrot KL, Sherrod SD, Flynn CR, McLean JA, Collision Cross Section Conformational Analyses of Bile Acids via Ion Mobility-Mass Spectrometry, J. Am. Soc. Mass Spectrom (2020). 10.1021/jasms.0c00015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Schmid R, Heuckeroth S, Korf A, Smirnov A, Myers O, Dyrlund TS, Bushuiev R, Murray KJ, Hoffmann N, Lu M, Sarvepalli A, Zhang Z, Fleischauer M, Dührkop K, Wesner M, Hoogstra SJ, Rudt E, Mokshyna O, Brungs C, Ponomarov K, Mutabdžija L, Damiani T, Pudney CJ, Earll M, Helmer PO, Fallon TR, Schulze T, Rivas-Ubach A, Bilbao A, Richter H, Nothias L-F, Wang M, Orešič M, Weng J-K, Böcker S, Jeibmann A, Hayen H, Karst U, Dorrestein PC, Petras D, Du X, Pluskal T, Integrative analysis of multimodal mass spectrometry data in MZmine 3, Nat. Biotechnol (2023). 10.1038/s41587-023-01690-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Tautenhahn R, Böttcher C, Neumann S, Highly sensitive feature detection for high resolution LC/MS, BMC Bioinformatics. 9 (2008) 504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Röst HL, Sachsenberg T, Aiche S, Bielow C, Weisser H, Aicheler F, Andreotti S, Ehrlich H-C, Gutenbrunner P, Kenar E, Liang X, Nahnsen S, Nilse L, Pfeuffer J, Rosenberger G, Rurik M, Schmitt U, Veit J, Walzer M, Wojnar D, Wolski WE, Schilling O, Choudhary JS, Malmström L, Aebersold R, Reinert K, Kohlbacher O, OpenMS: a flexible open-source software platform for mass spectrometry data analysis, Nat. Methods 13 (2016) 741–748. [DOI] [PubMed] [Google Scholar]
  • [84].Tsugawa H, Cajka T, Kind T, Ma Y, Higgins B, Ikeda K, Kanazawa M, VanderGheynst J, Fiehn O, Arita M, MS-DIAL: data-independent MS/MS deconvolution for comprehensive metabolome analysis, Nat. Methods 12 (2015) 523–526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Pino LK, Searle BC, Bollinger JG, Nunn B, MacLean B, MacCoss MJ, The Skyline ecosystem: Informatics for quantitative mass spectrometry proteomics, Mass Spectrom. Rev 39 (2020) 229–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Teo G, Chew WS, Burla BJ, Herr D, Tai ES, Wenk MR, Torta F, Choi H, MRMkit: Automated Data Processing for Large-Scale Targeted Metabolomics Analysis, Anal. Chem 92 (2020) 13677–13682. [DOI] [PubMed] [Google Scholar]
  • [87].Tautenhahn R, Patti GJ, Rinehart D, Siuzdak G, XCMS Online: a web-based platform to process untargeted metabolomic data, Anal. Chem 84 (2012) 5035–5039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Adams KJ, Pratt B, Bose N, Dubois LG, St John-Williams L, Perrott KM, Ky K, Kapahi P, Sharma V, MacCoss MJ, Moseley MA, Colton CA, MacLean BX, Schilling B, Thompson JW, Alzheimer’s Disease Metabolomics Consortium, Skyline for Small Molecules: A Unifying Software Package for Quantitative Metabolomics, J. Proteome Res 19 (2020) 1447–1458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Colby BN, Spectral deconvolution for overlapping GC/MS components, J. Am. Soc. Mass Spectrom 3 (1992) 558–562. [DOI] [PubMed] [Google Scholar]
  • [90].Dromey RG, Stefik MJ, Rindfleisch TC, Duffield AM, Extraction of Mass Spectra Free of Background and Neighboring Component Contributions from Gas Chromatography/mass Spectrometry Data, Anal. Chem 48 (1976) 1368–1375. [Google Scholar]
  • [91].Aksenov AA, Laponogov I, Zhang Z, Doran SLF, Belluomo I, Veselkov D, Bittremieux W, Nothias LF, Nothias-Esposito M, Maloney KN, Misra BB, Melnik AV, Smirnov A, Du X, Jones KL 2nd, Dorrestein K, Panitchpakdi M, Ernst M, van der Hooft JJJ, Gonzalez M, Carazzone C, Amézquita A, Callewaert C, Morton JT, Quinn RA, Bouslimani A, Orio AA, Petras D, Smania AM, Couvillion SP, Burnet MC, Nicora CD, Zink E, Metz TO, Artaev V, Humston-Fulmer E, Gregor R, Meijler MM, Mizrahi I, Eyal S, Anderson B, Dutton R, Lugan R, Boulch PL, Guitton Y, Prevost S, Poirier A, Dervilly G, Le Bizec B, Fait A, Persi NS, Song C, Gashu K, Coras R, Guma M, Manasson J, Scher JU, Barupal DK, Alseekh S, Fernie AR, Mirnezami R, Vasiliou V, Schmid R, Borisov RS, Kulikova LN, Knight R, Wang M, Hanna GB, Dorrestein PC, Veselkov K, Auto-deconvolution and molecular networking of gas chromatography-mass spectrometry data, Nat. Biotechnol 39 (2021) 169–173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Stancliffe E, Schwaiger-Haber M, Sindelar M, Patti GJ, DecoID improves identification rates in metabolomics through database-assisted MS/MS deconvolution, Nat. Methods 18 (2021) 779–787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Wu C-T, Wang Y, Wang Y, Ebbels T, Karaman I, Graça G, Pinto R, Herrington DM, Wang Y, Yu G, Targeted realignment of LC-MS profiles by neighbor-wise compound-specific graphical time warping with misalignment detection, Bioinformatics. 36 (2020) 2862–2871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Välikangas T, Suomi T, Elo LL, A systematic evaluation of normalization methods in quantitative label-free proteomics, Brief. Bioinform 19 (2018) 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Sysi-Aho M, Katajamaa M, Yetukuri L, Oresic M, Normalization method for metabolomics data using optimal selection of multiple internal standards, BMC Bioinformatics. 8 (2007) 93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Deininger S-O, Cornett DS, Paape R, Becker M, Pineau C, Rauser S, Walch A, Wolski E, Normalization in MALDI-TOF imaging datasets of proteins: practical considerations, Anal. Bioanal. Chem 401 (2011) 167–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Dieterle F, Ross A, Schlotterbeck G, Senn H, Probabilistic quotient normalization as robust method to account for dilution of complex biological mixtures. Application in 1H NMR metabonomics, Anal. Chem 78 (2006) 4281–4290. [DOI] [PubMed] [Google Scholar]
  • [98].Deng K, Zhao F, Rong Z, Cao L, Zhang L, Li K, Hou Y, Zhu Z-J, WaveICA 2.0: a novel batch effect removal method for untargeted metabolomics data without using batch information, Metabolomics. 17 (2021) 87. [DOI] [PubMed] [Google Scholar]
  • [99].Kim T, Tang O, Vernon ST, Kott KA, Koay YC, Park J, James DE, Grieve SM, Speed TP, Yang P, Figtree GA, O’Sullivan JF, Yang JYH, A hierarchical approach to removal of unwanted variation for large-scale metabolomics data, Nat. Commun 12 (2021) 4992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Schiffman C, Petrick L, Perttula K, Yano Y, Carlsson H, Whitehead T, Metayer C, Hayes J, Rappaport S, Dudoit S, Filtering procedures for untargeted LC-MS metabolomics data, BMC Bioinformatics. 20 (2019) 334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Kouřil Š, de Sousa J, Václavík J, Friedecký D, Adam T, CROP: correlation-based reduction of feature multiplicities in untargeted metabolomic data, Bioinformatics. 36 (2020) 2941–2942. [DOI] [PubMed] [Google Scholar]
  • [102].Chetnik K, Petrick L, Pandey G, MetaClean: a machine learning-based classifier for reduced false positive peak detection in untargeted LC-MS metabolomics data, Metabolomics. 16 (2020) 117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Gloaguen Y, Kirwan JA, Beule D, Deep Learning-Assisted Peak Curation for Large-Scale LC-MS Metabolomics, Anal. Chem 94 (2022) 4930–4937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Loulou Peisl BY, Schymanski EL, Wilmes P, Dark matter in host-microbiome metabolomics: Tackling the unknowns–A review, Anal. Chim. Acta 1037 (2018) 13–27. [DOI] [PubMed] [Google Scholar]
  • [105].da Silva RR, Dorrestein PC, Quinn RA, Illuminating the dark matter in metabolomics, Proc. Natl. Acad. Sci. U. S. A 112 (2015) 12549–12550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Liu R, Hong J, Xu X, Feng Q, Zhang D, Gu Y, Shi J, Zhao S, Liu W, Wang X, Xia H, Liu Z, Cui B, Liang P, Xi L, Jin J, Ying X, Wang X, Zhao X, Li W, Jia H, Lan Z, Li F, Wang R, Sun Y, Yang M, Shen Y, Jie Z, Li J, Chen X, Zhong H, Xie H, Zhang Y, Gu W, Deng X, Shen B, Xu X, Yang H, Xu G, Bi Y, Lai S, Wang J, Qi L, Madsen L, Wang J, Ning G, Kristiansen K, Wang W, Gut microbiome and serum metabolome alterations in obesity and after weight-loss intervention, Nat. Med 23 (2017) 859–868. [DOI] [PubMed] [Google Scholar]
  • [107].Santoru ML, Piras C, Murgia A, Palmas V, Camboni T, Liggi S, Ibba I, Lai MA, Orrù S, Blois S, Loizedda AL, Griffin JL, Usai P, Caboni P, Atzori L, Manzin A, Cross sectional evaluation of the gut-microbiome metabolome axis in an Italian cohort of IBD patients, Sci. Rep 7 (2017) 9523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Sun Y, Su Y, Zhu W, Microbiome-Metabolome Responses in the Cecum and Colon of Pig to a High Resistant Starch Diet, Front. Microbiol 7 (2016) 779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Zhang F, Zheng W, Xue Y, Yao W, Suhuai suckling piglet hindgut microbiome-metabolome responses to different dietary copper levels, Appl. Microbiol. Biotechnol 103 (2019) 853–868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Chong J, Xia J, Computational Approaches for Integrative Analysis of the Metabolome and Microbiome, Metabolites. 7 (2017). 10.3390/metabo7040062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Gloor GB, Macklaim JM, Pawlowsky-Glahn V, Egozcue JJ, Microbiome Datasets Are Compositional: And This Is Not Optional, Front. Microbiol 8 (2017) 2224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].Kalivodová A, Hron K, Filzmoser P, Najdekr L, Janečková H, Adam T, PLS-DA for compositional data with application to metabolomics, J. Chemom 29 (2015) 21–28. [Google Scholar]
  • [113].Zhang B, Horvath S, A general framework for weighted gene co-expression network analysis, Stat. Appl. Genet. Mol. Biol 4 (2005). 10.2202/1544-6115.1128. [DOI] [PubMed] [Google Scholar]
  • [114].Li J, Zhou D, Qiu W, Shi Y, Yang J-J, Chen S, Wang Q, Pan H, Application of Weighted Gene Co-expression Network Analysis for Data from Paired Design, Sci. Rep 8 (2018) 622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].McCall L-I, Tripathi A, Vargas F, Knight R, Dorrestein PC, Siqueira-Neto JL, Experimental Chagas disease-induced perturbations of the fecal microbiome and metabolome, PLoS Negl. Trop. Dis 12 (2018) e0006344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Morton JT, Aksenov AA, Nothias LF, Foulds JR, Quinn RA, Badri MH, Swenson TL, Van Goethem MW, Northen TR, Vazquez-Baeza Y, Wang M, Bokulich NA, Watters A, Song SJ, Bonneau R, Dorrestein PC, Knight R, Learning representations of microbe–metabolite interactions, Nature Methods. 16 (2019) 1306–1314. 10.1038/s41592-019-0616-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Jasbi P, Mohr AE, Shi X, Mahmood T, Zhu Q, Bruening M, Gu H, Whisner C, Microbiome and metabolome profiles of high screen time in a cohort of healthy college students, Sci. Rep 12 (2022) 3452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Bokulich NA, Łaniewski P, Adamov A, Chase DM, Caporaso JG, Herbst-Kralovetz MM, Multi-omics data integration reveals metabolome as the top predictor of the cervicovaginal microenvironment, PLoS Comput. Biol 18 (2022) e1009876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Taylor BC, Lejzerowicz F, Poirel M, Shaffer JP, Jiang L, Aksenov A, Litwin N, Humphrey G, Martino C, Miller-Montgomery S, Dorrestein PC, Veiga P, Song SJ, McDonald D, Derrien M, Knight R, Consumption of Fermented Foods Is Associated with Systematic Differences in the Gut Microbiome and Metabolome, mSystems. 5 (2020). 10.1128/mSystems.00901-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Haffner JJ, Katemauswa M, Kagone TS, Hossain E, Jacobson D, Flores K, Parab AR, Obregon-Tito AJ, Tito RY, Reyes LM, Troncoso-Corzo L, Guija-Poma E, Meda N, Carabin H, Honap TP, Sankaranarayanan K, Lewis CM Jr, McCall L-I, Untargeted Fecal Metabolomic Analyses across an Industrialization Gradient Reveal Shared Metabolites and Impact of Industrialization on Fecal Microbiome-Metabolome Interactions, mSystems. 7 (2022) e0071022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Gauglitz JM, West KA, Bittremieux W, Williams CL, Weldon KC, Panitchpakdi M, Di Ottavio F, Aceves CM, Brown E, Sikora NC, Jarmusch AK, Martino C, Tripathi A, Meehan MJ, Dorrestein K, Shaffer JP, Coras R, Vargas F, Goldasich LD, Schwartz T, Bryant M, Humphrey G, Johnson AJ, Spengler K, Belda-Ferre P, Diaz E, McDonald D, Zhu Q, Elijah EO, Wang M, Marotz C, Sprecher KE, Vargas-Robles D, Withrow D, Ackermann G, Herrera L, Bradford BJ, Marques LMM, Amaral JG, Silva RM, Veras FP, Cunha TM, Oliveira RDR, Louzada-Junior P, Mills RH, Piotrowski PK, Servetas SL, Da Silva SM, Jones CM, Lin NJ, Lippa KA, Jackson SA, Daouk RK, Galasko D, Dulai PS, Kalashnikova TI, Wittenberg C, Terkeltaub R, Doty MM, Kim JH, Rhee KE, Beauchamp-Walters J, Wright KP Jr, Dominguez-Bello MG, Manary M, Oliveira MF, Boland BS, Lopes NP, Guma M, Swafford AD, Dutton RJ, Knight R, Dorrestein PC, Enhancing untargeted metabolomics using metadata-based source annotation, Nat. Biotechnol 40 (2022) 1774–1779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Noecker C, Sanchez J, Bisanz JE, Escalante V, Alexander M, Trepka K, Heinken A, Liu Y, Dodd D, Thiele I, DeFelice B, Turnbaugh PJ, Systems biology illuminates alternative metabolic niches in the human gut microbiome, bioRxiv. (2022) 2022.09.19.508335. 10.1101/2022.09.19.508335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Maansson M, Vynne NG, Klitgaard A, Nybo JL, Melchiorsen J, Nguyen DD, Sanchez LM, Ziemert N, Dorrestein PC, Andersen MR, Gram L, An Integrated Metabolomic and Genomic Mining Workflow To Uncover the Biosynthetic Potential of Bacteria, mSystems. 1 (2016). 10.1128/mSystems.00028-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Elsden SR, Hilton MG, Waller JM, The end products of the metabolism of aromatic amino acids by Clostridia, Arch. Microbiol 107 (1976) 283–288. [DOI] [PubMed] [Google Scholar]
  • [125].Smith EA, Macfarlane GT, Formation of Phenolic and Indolic Compounds by Anaerobic Bacteria in the Human Large Intestine, Microb. Ecol 33 (1997) 180–188. [DOI] [PubMed] [Google Scholar]
  • [126].Han S, Van Treuren W, Fischer CR, Merrill BD, DeFelice BC, Sanchez JM, Higginbottom SK, Guthrie L, Fall LA, Dodd D, Fischbach MA, Sonnenburg JL, A metabolomics pipeline for the mechanistic interrogation of the gut microbiome, Nature. 595 (2021) 415–420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Aronov PA, Luo FJ-G, Plummer NS, Quan Z, Holmes S, Hostetter TH, Meyer TW, Colonic contribution to uremic solutes, J. Am. Soc. Nephrol 22 (2011) 1769–1776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Nemet I, Li XS, Haghikia A, Li L, Wilcox J, Romano KA, Buffa JA, Witkowski M, Demuth I, König M, Steinhagen-Thiessen E, Bäckhed F, Fischbach MA, Tang WHW, Landmesser U, Hazen SL, Atlas of gut microbe-derived products from aromatic amino acids and risk of cardiovascular morbidity and mortality, Eur. Heart J 44 (2023) 3085–3096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X, Wu Y, Li L, Smith JD, DiDonato JA, Chen J, Li H, Wu GD, Lewis JD, Warrier M, Brown JM, Krauss RM, Tang WHW, Bushman FD, Lusis AJ, Hazen SL, Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis, Nat. Med 19 (2013) 576–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Tang WHW, Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, Wu Y, Hazen SL, Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk, N. Engl. J. Med 368 (2013) 1575–1584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Dodd D, Spitzer MH, Van Treuren W, Merrill BD, Hryckowian AJ, Higginbottom SK, Le A, Cowan TM, Nolan GP, Fischbach MA, Sonnenburg JL, A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites, Nature. 551 (2017) 648–652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Marcobal A, Yusufaly T, Higginbottom S, Snyder M, Sonnenburg JL, Mias GI, Metabolome progression during early gut microbial colonization of gnotobiotic mice, Sci. Rep 5 (2015) 11589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Velagapudi VR, Hezaveh R, Reigstad CS, Gopalacharyulu P, Yetukuri L, Islam S, Felin J, Perkins R, Borén J, Oresic M, Bäckhed F, The gut microbiota modulates host energy and lipid metabolism in mice, J. Lipid Res 51 (2010) 1101–1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Subramanian I, Verma S, Kumar S, Jere A, Anamika K, Multi-omics Data Integration, Interpretation, and Its Application, Bioinform. Biol. Insights 14 (2020) 1177932219899051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Park J, Kerner A, Burns MA, Lin XN, Microdroplet-enabled highly parallel co-cultivation of microbial communities, PLoS One. 6 (2011) e17019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Engineering Robust Production Microbes for Large-Scale Cultivation, Trends Microbiol. 27 (2019) 524–537. [DOI] [PubMed] [Google Scholar]
  • [137].Clavel T, Gomes-Neto JC, Lagkouvardos I, Ramer-Tait AE, Deciphering interactions between the gut microbiota and the immune system via microbial cultivation and minimal microbiomes, Immunol. Rev 279 (2017) 8–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Nichols D, Cahoon N, Trakhtenberg EM, Pham L, Mehta A, Belanger A, Kanigan T, Lewis K, Epstein SS, Use of ichip for high-throughput in situ cultivation of “uncultivable” microbial species, Appl. Environ. Microbiol 76 (2010) 2445–2450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Berdy B, Spoering AL, Ling LL, Epstein SS, In situ cultivation of previously uncultivable microorganisms using the ichip, Nat. Protoc 12 (2017) 2232–2242. [DOI] [PubMed] [Google Scholar]
  • [140].Sizova MV, Hohmann T, Hazen A, Paster BJ, Halem SR, Murphy CM, Panikov NS, Epstein SS, New approaches for isolation of previously uncultivated oral bacteria, Appl. Environ. Microbiol 78 (2012) 194–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [141].Xiang Y, Wen H, Yu Y, Li M, Fu X, Huang S, Gut-on-chip: Recreating human intestine in vitro, J. Tissue Eng 11 (2020) 2041731420965318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].Higgs RE, Zahn JA, Gygi JD, Hilton MD, Rapid Method To Estimate the Presence of Secondary Metabolites in Microbial Extracts, Appl. Environ. Microbiol 67 (2001). 10.1128/AEM.67.1.371-376.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Gaudêncio SP, Pereira F, Dereplication: racing to speed up the natural products discovery process, Nat. Prod. Rep 32 (2015) 779–810. [DOI] [PubMed] [Google Scholar]
  • [144].Wang M, Carver JJ, Phelan VV, Sanchez LM, Garg N, Peng Y, Nguyen DD, Watrous J, Kapono CA, Luzzatto-Knaan T, Porto C, Bouslimani A, Melnik AV, Meehan MJ, Liu W-T, Crüsemann M, Boudreau PD, Esquenazi E, Sandoval-Calderón M, Kersten RD, Pace LA, Quinn RA, Duncan KR, Hsu C-C, Floros DJ, Gavilan RG, Kleigrewe K, Northen T, Dutton RJ, Parrot D, Carlson EE, Aigle B, Michelsen CF, Jelsbak L, Sohlenkamp C, Pevzner P, Edlund A, McLean J, Piel J, Murphy BT, Gerwick L, Liaw C-C, Yang Y-L, Humpf H-U, Maansson M, Keyzers RA, Sims AC, Johnson AR, Sidebottom AM, Sedio BE, Klitgaard A, Larson CB, P CAB, Torres-Mendoza D, Gonzalez DJ, Silva DB, Marques LM, Demarque DP, Pociute E, O’Neill EC, Briand E, Helfrich EJN, Granatosky EA, Glukhov E, Ryffel F, Houson H, Mohimani H, Kharbush JJ, Zeng Y, Vorholt JA, Kurita KL, Charusanti P, McPhail KL, Nielsen KF, Vuong L, Elfeki M, Traxler MF, Engene N, Koyama N, Vining OB, Baric R, Silva RR, Mascuch SJ, Tomasi S, Jenkins S, Macherla V, Hoffman T, Agarwal V, Williams PG, Dai J, Neupane R, Gurr J, Rodríguez AMC, Lamsa A, Zhang C, Dorrestein K, Duggan BM, Almaliti J, Allard P-M, Phapale P, Nothias L-F, Alexandrov T, Litaudon M, Wolfender J-L, Kyle JE, Metz TO, Peryea T, Nguyen D-T, VanLeer D, Shinn P, Jadhav A, Müller R, Waters KM, Shi W, Liu X, Zhang L, Knight R, Jensen PR, Palsson BO, Pogliano K, Linington RG, Gutiérrez M, Lopes NP, Gerwick WH, Moore BS, Dorrestein PC, Bandeira N, Sharing and community curation of mass spectrometry data with Global Natural Products Social Molecular Networking, Nat. Biotechnol 34 (2016) 828–837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Smith CA, O’Maille G, Want EJ, Qin C, Trauger SA, Brandon TR, Custodio DE, Abagyan R, Siuzdak G, METLIN: a metabolite mass spectral database, Ther. Drug Monit 27 (2005) 747–751. [DOI] [PubMed] [Google Scholar]
  • [146].Dictionary of Natural Products, (n.d.). https://dnp.chemnetbase.com/chemical/ChemicalSearch.xhtml?dswid=-6075 (accessed May 26, 2023).
  • [147].NIST Standard Reference Database, (n.d.). https://www.nist.gov/srd (accessed May 26, 2023).
  • [148].Dias DA, Jones OAH, Beale DJ, Boughton BA, Benheim D, Kouremenos KA, Wolfender J-L, Wishart DS, Current and Future Perspectives on the Structural Identification of Small Molecules in Biological Systems, Metabolites. 6 (2016). 10.3390/metabo6040046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Elyashberg M, Identification and structure elucidation by NMR spectroscopy, Trends Analyt. Chem 69 (2015) 88–97. [Google Scholar]
  • [150].Huang Z, Chen MS, Woroch CP, Markland TE, Kanan MW, A framework for automated structure elucidation from routine NMR spectra, Chem. Sci 12 (2021) 15329–15338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Shen S-M, Appendino G, Guo Y-W, Pitfalls in the structural elucidation of small molecules. A critical analysis of a decade of structural misassignments of marine natural products, Nat. Prod. Rep 39 (2022) 1803–1832. [DOI] [PubMed] [Google Scholar]
  • [152].Kreis R, Issues of spectral quality in clinical 1H-magnetic resonance spectroscopy and a gallery of artifacts, NMR Biomed. 17 (2004) 361–381. [DOI] [PubMed] [Google Scholar]
  • [153].Nothias L-F, Petras D, Schmid R, Dührkop K, Rainer J, Sarvepalli A, Protsyuk I, Ernst M, Tsugawa H, Fleischauer M, Aicheler F, Aksenov AA, Alka O, Allard P-M, Barsch A, Cachet X, Caraballo-Rodriguez AM, Da Silva RR, Dang T, Garg N, Gauglitz JM, Gurevich A, Isaac G, Jarmusch AK, Kameník Z, Kang KB, Kessler N, Koester I, Korf A, Le Gouellec A, Ludwig M, Martin H C, McCall L-I, McSayles J, Meyer SW, Mohimani H, Morsy M, Moyne O, Neumann S, Neuweger H, Nguyen NH, Nothias-Esposito M, Paolini J, Phelan VV, Pluskal T, Quinn RA, Rogers S, Shrestha B, Tripathi A, van der Hooft JJJ, Vargas F, Weldon KC, Witting M, Yang H, Zhang Z, Zubeil F, Kohlbacher O, Böcker S, Alexandrov T, Bandeira N, Wang M, Dorrestein PC, Feature-based molecular networking in the GNPS analysis environment, Nat. Methods 17 (2020) 905–908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Liu Y, Mrzic A, Meysman P, De Vijlder T, Romijn EP, Valkenborg D, Bittremieux W, Laukens K, MESSAR: Automated recommendation of metabolite substructures from tandem mass spectra, PLoS One. 15 (2020) e0226770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].Zhang C, Idelbayev Y, Roberts N, Tao Y, Nannapaneni Y, Duggan BM, Min J, Lin EC, Gerwick EC, Cottrell GW, Gerwick WH, Author Correction: Small Molecule Accurate Recognition Technology (SMART) to Enhance Natural Products Research, Sci. Rep 10 (2020) 4724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].Cao L, Guler M, Tagirdzhanov A, Lee Y-Y, Gurevich A, Mohimani H, MolDiscovery: learning mass spectrometry fragmentation of small molecules, Nat. Commun 12 (2021) 3718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Schött H-F, Krautbauer S, Höring M, Liebisch G, Matysik S, A Validated, Fast Method for Quantification of Sterols and Gut Microbiome Derived 5α/β-Stanols in Human Feces by Isotope Dilution LC-High-Resolution MS, Anal. Chem 90 (2018) 8487–8494. [DOI] [PubMed] [Google Scholar]
  • [158].den Besten G, van Eunen K, Groen AK, Venema K, Reijngoud D-J, Bakker BM, The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism, J. Lipid Res 54 (2013) 2325–2340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [159].Marques LA, Cazarin CBB, Bicas J, Maróstica MR, Carrilho E, Bogusz S, Determination of Short Chain Fatty Acids in Mice Feces by Capillary Electrophoresis, J. Braz. Chem. Soc 30 (2019) 1326–1334. [Google Scholar]
  • [160].den Besten G, Bleeker A, Gerding A, van Eunen K, Havinga R, van Dijk TH, Oosterveer MH, Jonker JW, Groen AK, Reijngoud D-J, Bakker BM, Short-Chain Fatty Acids Protect Against High-Fat Diet–Induced Obesity via a PPARγ-Dependent Switch From Lipogenesis to Fat Oxidation, Diabetes. 64 (2015) 2398–2408. [DOI] [PubMed] [Google Scholar]
  • [161].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. 165 (2016) 1332–1345. [DOI] [PubMed] [Google Scholar]
  • [162].Liu P, Wang Y, Yang G, Zhang Q, Meng L, Xin Y, Jiang X, The role of short-chain fatty acids in intestinal barrier function, inflammation, oxidative stress, and colonic carcinogenesis, Pharmacol. Res 165 (2021) 105420. [DOI] [PubMed] [Google Scholar]
  • [163].Bergman EN, Energy contributions of volatile fatty acids from the gastrointestinal tract in various species, Physiol. Rev 70 (1990) 567–590. [DOI] [PubMed] [Google Scholar]
  • [164].Silva YP, Bernardi A, Frozza RL, The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication, Front. Endocrinol . 11 (2020) 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].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, in: Giera M (Ed.), Clinical Metabolomics: Methods and Protocols, Springer New York, New York, NY, 2018: pp. 247–256. [DOI] [PubMed] [Google Scholar]
  • [166].de la Cuesta-Zuluaga J, Mueller NT, Álvarez-Quintero R, Velásquez-Mejía EP, Sierra JA, Corrales-Agudelo V, Carmona JA, Abad JM, Escobar JS, Higher Fecal Short-Chain Fatty Acid Levels Are Associated with Gut Microbiome Dysbiosis, Obesity, Hypertension and Cardiometabolic Disease Risk Factors, Nutrients. 11 (2018). 10.3390/nu11010051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Cummings JH, Pomare EW, Branch WJ, Naylor CP, Macfarlane GT, Short chain fatty acids in human large intestine, portal, hepatic and venous blood, Gut. 28 (1987) 1221–1227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [168].Parada Venegas D, De la Fuente MK, Landskron G, González MJ, Quera R, Dijkstra G, Harmsen HJM, Faber KN, Hermoso MA, Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases, Front. Immunol 10 (2019) 277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Topping DL, Clifton PM, Short-chain fatty acids and human colonic function: roles of resistant starch and nonstarch polysaccharides, Physiol. Rev 81 (2001) 1031–1064. [DOI] [PubMed] [Google Scholar]
  • [170].Haghikia A, Jörg S, Duscha A, Berg J, Manzel A, Waschbisch A, Hammer A, Lee D-H, May C, Wilck N, Balogh A, Ostermann AI, Schebb NH, Akkad DA, Grohme DA, Kleinewietfeld M, Kempa S, Thöne J, Demir S, Müller DN, Gold R, Linker RA, Dietary Fatty Acids Directly Impact Central Nervous System Autoimmunity via the Small Intestine, Immunity. 43 (2015) 817–829. [DOI] [PubMed] [Google Scholar]
  • [171].Trivedi N, Erickson HE, Bala V, Chhonker YS, Murry DJ, A Concise Review of Liquid Chromatography-Mass Spectrometry-Based Quantification Methods for Short Chain Fatty Acids as Endogenous Biomarkers, Int. J. Mol. Sci 23 (2022). 10.3390/ijms232113486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [172].Bertolo MRV, Bogusz Junior S, Quantification of Short-Chain Fatty Acids in Feces, in: Betim Cazarin CB (Ed.), Basic Protocols in Foods and Nutrition, Springer US, New York, NY, 2022: pp. 73–92. [Google Scholar]
  • [173].Kim K-S, Lee Y, Chae W, Cho J-Y, An Improved Method to Quantify Short-Chain Fatty Acids in Biological Samples Using Gas Chromatography-Mass Spectrometry, Metabolites. 12 (2022). 10.3390/metabo12060525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Saha S, Day-Walsh P, Shehata E, Kroon PA, Development and validation of a LC-MS/MS technique for the analysis of short chain fatty acids in tissues and biological fluids without derivatisation using isotope labelled internal standards, Molecules. 26 (2021) 6444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Han X, Guo J, You Y, Yin M, Ren C, Zhan J, Huang W, A fast and accurate way to determine short chain fatty acids in mouse feces based on GC–MS, Journal of Chromatography B. 1099 (2018) 73–82. [DOI] [PubMed] [Google Scholar]
  • [176].Banel A, Jakimska A, Wasielewska M, Wolska L, Zygmunt B, Determination of SCFAs in water using GC-FID. Selection of the separation system, Anal. Chim. Acta 716 (2012) 24–27. [DOI] [PubMed] [Google Scholar]
  • [177].Shafaei A, Vamathevan V, Pandohee J, Lawler NG, Broadhurst D, Boyce MC, Sensitive and quantitative determination of short-chain fatty acids in human serum using liquid chromatography mass spectrometry, Anal. Bioanal. Chem 413 (2021) 6333–6342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [178].Bihan DG, Rydzak T, Wyss M, Pittman K, McCoy KD, Lewis IA, Method for absolute quantification of short chain fatty acids via reverse phase chromatography mass spectrometry, PLoS One. 17 (2022) e0267093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Nagatomo R, Okada Y, Ichimura M, Tsuneyama K, Inoue K, Application of 2-Picolylamine Derivatized Ultra-high Performance Liquid Chromatography Tandem Mass Spectrometry for the Determination of Short-chain Fatty Acids in Feces Samples, Anal. Sci 34 (2018) 1031–1036. [DOI] [PubMed] [Google Scholar]
  • [180].Liebisch G, Ecker J, Roth S, Schweizer, Sabine, Öttl V, Schött H-F, Yoon H, Haller D, Holler E, Burkhardt R, Matysik S, Quantification of Fecal Short Chain Fatty Acids by Liquid Chromatography Tandem Mass Spectrometry—Investigation of Pre-Analytic Stability, Biomolecules. 9 (2019) 121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [181].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. 196 (2019) 249–254. [DOI] [PubMed] [Google Scholar]
  • [182].Li M, Zhu R, Song X, Wang Z, Weng H, Liang J, A sensitive method for the quantification of short-chain fatty acids by benzyl chloroformate derivatization combined with GC-MS, Analyst. 145 (2020) 2692–2700. [DOI] [PubMed] [Google Scholar]
  • [183].Furuhashi T, Sugitate K, Nakai T, Jikumaru Y, Ishihara G, Rapid profiling method for mammalian feces short chain fatty acids by GC-MS, Anal. Biochem 543 (2018) 51–54. [DOI] [PubMed] [Google Scholar]
  • [184].Tao X, Liu Y, Wang Y, Qiu Y, Lin J, Zhao A, Su M, Jia W, GC-MS with ethyl chloroformate derivatization for comprehensive analysis of metabolites in serum and its application to human uremia, Anal. Bioanal. Chem 391 (2008) 2881–2889. [DOI] [PubMed] [Google Scholar]
  • [185].He L, Prodhan MAI, Yuan F, Yin X, Lorkiewicz PK, Wei X, Feng W, McClain C, Zhang X, Simultaneous quantification of straight-chain and branched-chain short chain fatty acids by gas chromatography mass spectrometry, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci 1092 (2018) 359–367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Dei Cas M, Paroni R, Saccardo A, Casagni E, Arnoldi S, Gambaro V, Saresella M, Mario C, La Rosa F, Marventano I, Piancone F, Roda G, A straightforward LC-MS/MS analysis to study serum profile of short and medium chain fatty acids, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci 1154 (2020) 121982. [DOI] [PubMed] [Google Scholar]
  • [187].Feng X, Tang Z, Chen B, Feng S, Liu Y, Meng Q, A high-efficiency quantitation method for fatty aldehyde based on chemical isotope-tagged derivatisation, J. Food Compost. Anal 107 (2022) 104381. [Google Scholar]
  • [188].Ma S-R, Tong Q, Zhao Z-X, Cong L, Yu J-B, Fu J, Han P, Pan L-B, Gu R, Peng R, Zhang Z-W, Wang Y, Jiang J-D, Determination of berberine-upregulated endogenous short-chain fatty acids through derivatization by 2-bromoacetophenone, Anal. Bioanal. Chem 411 (2019) 3191–3207. [DOI] [PubMed] [Google Scholar]
  • [189].Han J, Lin K, Sequeira C, Borchers CH, An isotope-labeled chemical derivatization method for the quantitation of short-chain fatty acids in human feces by liquid chromatography-tandem mass spectrometry, Anal. Chim. Acta 854 (2015) 86–94. [DOI] [PubMed] [Google Scholar]
  • [190].Chan JCY, Kioh DYQ, Yap GC, Lee BW, Chan ECY, A novel LCMSMS method for quantitative measurement of short-chain fatty acids in human stool derivatized with 12C- and 13C-labelled aniline, J. Pharm. Biomed. Anal 138 (2017) 43–53. [DOI] [PubMed] [Google Scholar]
  • [191].Xu W, Chen D, Wang N, Zhang T, Zhou R, Huan T, Lu Y, Su X, Xie Q, Li L, Li L, Development of high-performance chemical isotope labeling LC-MS for profiling the human fecal metabolome, Anal. Chem 87 (2015) 829–836. [DOI] [PubMed] [Google Scholar]
  • [192].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 Analyt. Technol. Biomed. Life Sci 1083 (2018) 137–145. [DOI] [PubMed] [Google Scholar]
  • [193].Zhu J-H, Mao Q, Wang S-Y, Liu H, Zhou S-S, Zhang W, Kong M, Zhu H, Li S-L, Optimization and validation of direct gas chromatography-mass spectrometry method for simultaneous quantification of ten short-chain fatty acids in rat feces, J. Chromatogr. A 1669 (2022) 462958. [DOI] [PubMed] [Google Scholar]
  • [194].Fu Z, Jia Q, Zhang H, Kang L, Sun X, Zhang M, Wang Y, Hu P, Simultaneous quantification of eleven short-chain fatty acids by derivatization and solid phase microextraction-Gas chromatography tandem mass spectrometry, J. Chromatogr. A 1661 (2022) 462680. [DOI] [PubMed] [Google Scholar]
  • [195].Yao L, Davidson EA, Shaikh MW, Forsyth CB, Prenni JE, Broeckling CD, Quantitative analysis of short-chain fatty acids in human plasma and serum by GC-MS, Anal. Bioanal. Chem 414 (2022) 4391–4399. [DOI] [PubMed] [Google Scholar]
  • [196].Wang R, Fan C, Fan X, Zhao Y, Wang Y, Li P, Tang T, Yao H, Chen S, Chen D, Qi K, A Fast and Accurate Way to Determine Short Chain Fatty Acids in Human Serum by GC–MS and Their Distribution in Children with Digestive Diseases, Chromatographia. 83 (2020) 273–286. [Google Scholar]
  • [197].Northfield TC, Zentler-Munro P, Jazwari R, Bile Acids in Health and Disease: Update on Cholesterol Gallstones and Bile Acid Diarrhoea, Springer Science & Business Media, 2012. [Google Scholar]
  • [198].Zhang J, Gao L-Z, Chen Y-J, Zhu P-P, Yin S-S, Su M-M, Ni Y, Miao J, Wu W-L, Chen H, Brouwer KLR, Liu C-X, Xu L, Jia W, Lan K, Continuum of Host-Gut Microbial Co-metabolism: Host CYP3A4/3A7 are Responsible for Tertiary Oxidations of Deoxycholate Species, Drug Metab. Dispos 47 (2019) 283–294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [199].Ajouz H, Mukherji D, Shamseddine A, Secondary bile acids: an underrecognized cause of colon cancer, World J. Surg. Oncol 12 (2014) 164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [200].Dorrestein P, Gentry E, Collins S, Panitchpakdi M, Belda-Ferre P, Stewart A, Wang M, Jarmusch A, Avila-Pacheco J, Plichta D, Aron A, Vlamakis H, Ananthakrishnan A, Clish C, Xavier R, Baker E, Patterson A, Knight R, Siegel D, A Synthesis-Based Reverse Metabolomics Approach for the Discovery of Chemical Structures from Humans and Animals, (2021). 10.21203/rs.3.rs-820302/v1. [DOI] [Google Scholar]
  • [201].Jia B, Commentary: Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells, Front. Immunol 10 (2019) 282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [202].Yoshimoto S, Loo TM, Atarashi K, Kanda H, Sato S, Oyadomari S, Iwakura Y, Oshima K, Morita H, Hattori M, Honda K, Ishikawa Y, Hara E, Ohtani N, Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome, Nature. 499 (2013) 97–101. [DOI] [PubMed] [Google Scholar]
  • [203].Pai R, Tarnawski AS, Tran T, Deoxycholic acid activates beta-catenin signaling pathway and increases colon cell cancer growth and invasiveness, Mol. Biol. Cell 15 (2004) 2156–2163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [204].Ridlon JM, Wolf PG, Gaskins HR, Taurocholic acid metabolism by gut microbes and colon cancer, Gut Microbes. 7 (2016) 201–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [205].Wells JE, Berr F, Thomas LA, Hermon Dowling R, Hylemon PB, Isolation and characterization of cholic acid 7α-dehydroxylating fecal bacteria from cholesterol gallstone patients, Journal of Hepatology. 32 (2000) 4–10. 10.1016/s0168-8278(00)80183-x. [DOI] [PubMed] [Google Scholar]
  • [206].Berr F, Kullak-Ublick GA, Paumgartner G, Münzing W, Hylemon PB, 7 alpha-dehydroxylating bacteria enhance deoxycholic acid input and cholesterol saturation of bile in patients with gallstones, Gastroenterology. 111 (1996) 1611–1620. [DOI] [PubMed] [Google Scholar]
  • [207].Kakiyama G, Hylemon PB, Zhou H, Pandak WM, Heuman DM, Kang DJ, Takei H, Nittono H, Ridlon JM, Fuchs M, Gurley EC, Wang Y, Liu R, Sanyal AJ, Gillevet PM, Bajaj JS, Colonic inflammation and secondary bile acids in alcoholic cirrhosis, Am. J. Physiol. Gastrointest. Liver Physiol 306 (2014) G929–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [208].Kakiyama G, Pandak WM, Gillevet PM, Hylemon PB, Heuman DM, Daita K, Takei H, Muto A, Nittono H, Ridlon JM, White MB, Noble NA, Monteith P, Fuchs M, Thacker LR, Sikaroodi M, Bajaj JS, Modulation of the fecal bile acid profile by gut microbiota in cirrhosis, J. Hepatol 58 (2013) 949–955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [209].Buffie CG, Bucci V, Stein RR, McKenney PT, Ling L, Gobourne A, No D, Liu H, Kinnebrew M, Viale A, Littmann E, van den Brink MRM, Jenq RR, Taur Y, Sander C, Cross JR, Toussaint NC, Xavier JB, Pamer EG, Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile, Nature. 517 (2015) 205–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [210].Sorg JA, Sonenshein AL, Bile Salts and Glycine as Cogerminants for Clostridium difficile Spores, Journal of Bacteriology. 190 (2008) 2505–2512. 10.1128/jb.01765-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [211].Theriot CM, Bowman AA, Young VB, Antibiotic-Induced Alterations of the Gut Microbiota Alter Secondary Bile Acid Production and Allow for Clostridium difficile Spore Germination and Outgrowth in the Large Intestine, mSphere. 1 (2016). 10.1128/mSphere.00045-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [212].Makishima M, Okamoto AY, Repa JJ, Tu H, Learned RM, Luk A, Hull MV, Lustig KD, Mangelsdorf DJ, Shan B, Identification of a nuclear receptor for bile acids, Science. 284 (1999) 1362–1365. [DOI] [PubMed] [Google Scholar]
  • [213].Keitel V, Donner M, Winandy S, Kubitz R, Häussinger D, Expression and function of the bile acid receptor TGR5 in Kupffer cells, Biochem. Biophys. Res. Commun 372 (2008) 78–84. [DOI] [PubMed] [Google Scholar]
  • [214].Suga T, Yamaguchi H, Sato T, Maekawa M, Goto J, Mano N, Preference of Conjugated Bile Acids over Unconjugated Bile Acids as Substrates for OATP1B1 and OATP1B3, PLoS One. 12 (2017) e0169719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [215].Režen T, Rozman D, Kovács T, Kovács P, Sipos A, Bai P, Mikó E, The role of bile acids in carcinogenesis, Cell. Mol. Life Sci 79 (2022) 243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [216].Peng Z, Zhang Q, Mao Z, Wang J, Liu C, Lin X, Li X, Ji W, Fan J, Wang M, Su C, A rapid quantitative analysis of bile acids, lysophosphatidylcholines and polyunsaturated fatty acids in biofluids based on ultraperformance liquid chromatography coupled with triple quadrupole tandem massspectrometry, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci 1068-1069 (2017) 343–351. [DOI] [PubMed] [Google Scholar]
  • [217].John C, Werner P, Worthmann A, Wegner K, Tödter K, Scheja L, Rohn S, Heeren J, Fischer M, A liquid chromatography-tandem mass spectrometry-based method for the simultaneous determination of hydroxy sterols and bile acids, J. Chromatogr. A 1371 (2014) 184–195. [DOI] [PubMed] [Google Scholar]
  • [218].Dutta M, Cai J, Gui W, Patterson AD, A review of analytical platforms for accurate bile acid measurement, Anal. Bioanal. Chem 411 (2019) 4541 –4549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [219].Liu S, Zhang Y, Qu B, Qin G, Cheng J, Lu F, Qu H, Zhao Y, Detection of total bile acids in biological samples using an indirect competitive ELISA based on four monoclonal antibodies, Anal. Methods 9 (2017) 625–633. [Google Scholar]
  • [220].Baldofski S, Hoffmann H, Lehmann A, Breitfeld S, Garbe L-A, Schneider RJ, Enzyme-linked immunosorbent assay (ELISA) for the anthropogenic marker isolithocholic acid in water, J. Environ. Manage 182 (2016) 612–619. [DOI] [PubMed] [Google Scholar]
  • [221].Cui X, Vasylieva N, Shen D, Barnych B, Yang J, He Q, Jiang Z, Zhao S, Hammock BD, Biotinylated single-chain variable fragment-based enzyme-linked immunosorbent assay for glycocholic acid, Analyst. 143 (2018) 2057–2065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [222].Alnouti Y, Csanaky IL, Klaassen CD, Quantitative-profiling of bile acids and their conjugates in mouse liver, bile, plasma, and urine using LC-MS/MS, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci 873 (2008) 209–217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [223].Humbert L, Maubert MA, Wolf C, Duboc H, Mahé M, Farabos D, Seksik P, Mallet JM, Trugnan G, Masliah J, Rainteau D, Bile acid profiling in human biological samples: comparison of extraction procedures and application to normal and cholestatic patients, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci 899 (2012) 135–145. [DOI] [PubMed] [Google Scholar]
  • [224].Sarafian MH, Lewis MR, Pechlivanis A, Ralphs S, McPhail MJW, Patel VC, Dumas M-E, Holmes E, Nicholson JK, Bile acid profiling and quantification in biofluids using ultra-performance liquid chromatography tandem mass spectrometry, Anal. Chem 87 (2015) 9662–9670. [DOI] [PubMed] [Google Scholar]
  • [225].Gómez C, Stücheli S, Kratschmar DV, Bouitbir J, Odermatt A, Development and Validation of a Highly Sensitive LC-MS/MS Method for the Analysis of Bile Acids in Serum, Plasma, and Liver Tissue Samples, Metabolites. 10 (2020). 10.3390/metabo10070282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [226].Nagana Gowda GA, Shanaiah N, Cooper A, Maluccio M, Raftery D, Bile acids conjugation in human bile is not random: new insights from (1)H-NMR spectroscopy at 800 MHz, Lipids. 44 (2009) 527–535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [227].Gowda GAN, Ijare OB, Somashekar BS, Sharma A, Kapoor VK, Khetrapal CL, Single-step analysis of individual conjugated bile acids in human bile using 1H NMR spectroscopy, Lipids. 41 (2006) 591–603. [DOI] [PubMed] [Google Scholar]
  • [228].Zhou M, Shao J, Wu C-Y, Shu L, Dong W, Liu Y, Chen M, Wynn RM, Wang J, Wang J, Gui W-J, Qi X, Lusis AJ, Li Z, Wang W, Ning G, Yang X, Chuang DT, Wang Y, Sun H, Targeting BCAA Catabolism to Treat Obesity-Associated Insulin Resistance, Diabetes. 68 (2019) 1730–1746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [229].Gojda J, Cahova M, Gut Microbiota as the Link between Elevated BCAA Serum Levels and Insulin Resistance, Biomolecules. 11 (2021). 10.3390/biom11101414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [230].Agus A, Clément K, Sokol H, Gut microbiota-derived metabolites as central regulators in metabolic disorders, Gut. 70 (2021) 1174–1182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [231].Siddik MAB, Shin AC, Recent Progress on Branched-Chain Amino Acids in Obesity, Diabetes, and Beyond, Endocrinol Metab (Seoul). 34 (2019) 234–246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [232].Newgard CB, An J, Bain JR, Muehlbauer MJ, Stevens RD, Lien LF, Haqq AM, Shah SH, Arlotto M, Slentz CA, Rochon J, Gallup D, Ilkayeva O, Wenner BR, Yancy WS Jr, Eisenson H, Musante G, Surwit RS, Millington DS, Butler MD, Svetkey LP, A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance, Cell Metab. 9 (2009) 311–326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [233].Yu L, Zhu Q, Li Y, Song P, Zhang J, Dietary Branched-Chain Amino Acids (BCAAs) and Risk of Dyslipidemia in a Chinese Population, Nutrients. 14 (2022). 10.3390/nu14091824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [234].Ramos-Garcia V, Ten-Doménech I, Moreno-Giménez A, Campos-Berga L, Parra-Llorca A, Solaz-García Á, Lara-Cantón I, Pinilla-Gonzalez A, Gormaz M, Vento M, Kuligowski J, Quintás G, GC-MS analysis of short chain fatty acids and branched chain amino acids in urine and faeces samples from newborns and lactating mothers, Clin. Chim. Acta 532 (2022) 172–180. [DOI] [PubMed] [Google Scholar]
  • [235].Whipp AM, Heinonen-Guzejev M, Pietiläinen KH, van Kamp I, Kaprio J, Branched-chain amino acids linked to depression in young adults, Front. Neurosci 16 (2022) 935858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [236].Teepoo S, Sannok T, Arsawiset S, Sansenya S, A portable device as a paper test strip platform with smartphone application for detection of branched-chain amino acids in edible insects, Food Chem. 405 (2023) 134560. [DOI] [PubMed] [Google Scholar]
  • [237].Fingerhut R, Röschinger W, Heck M, A rapid and sensitive UPLC-MS/MS-method for the separation and quantification of branched-chain amino acids from dried blood samples of patients with maple syrup urine disease (MSUD), Screening. 2 (2016) 2. [Google Scholar]
  • [238].Zheng X, Qiu Y, Zhong W, Baxter S, Su M, Li Q, Xie G, Ore BM, Qiao S, Spencer MD, Zeisel SH, Zhou Z, Zhao A, Jia W, A targeted metabolomic protocol for short-chain fatty acids and branched-chain amino acids, Metabolomics. 9 (2013) 818–827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [239].Piestansky J, Matuskova M, Cizmarova I, Olesova D, Mikus P, Determination of Branched-Chain Amino Acids in Food Supplements and Human Plasma by a CE-MS/MS Method with Enhanced Resolution, Int. J. Mol. Sci 22 (2021). 10.3390/ijms22158261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [240].Haq S, Grondin JA, Khan WI, Tryptophan-derived serotonin-kynurenine balance in immune activation and intestinal inflammation, FASEB J. 35 (2021) e21888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [241].Roager HM, Licht TR, Microbial tryptophan catabolites in health and disease, Nat. Commun 9 (2018) 3294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [242].Gao J, Xu K, Liu H, Liu G, Bai M, Peng C, Li T, Yin Y, Impact of the Gut Microbiota on Intestinal Immunity Mediated by Tryptophan Metabolism, Front. Cell. Infect. Microbiol 8 (2018) 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [243].Gouellec Le, Chaker H, Polack B, Guery B, Toussaint B, Stasia MJ, Genestet C, Scavenging of reactive oxygen species by tryptophan metabolites helps Pseudomonas aeruginosa escape neutrophil killing, Free Radical Biology and Medicine. 73 (2014) 400–410. [DOI] [PubMed] [Google Scholar]
  • [244].Li G, Young KD, Indole production by the tryptophanase TnaA in Escherichia coli is determined by the amount of exogenous tryptophan, Microbiology. 159 (2013) 402–410. [DOI] [PubMed] [Google Scholar]
  • [245].Vujkovic-Cvijin I, Dunham RM, Iwai S, Maher MC, Albright RG, Broadhurst MJ, Hernandez RD, Lederman MM, Huang Y, Somsouk M, Deeks SG, Hunt PW, Lynch SV, McCune JM, Dysbiosis of the gut microbiota is associated with HIV disease progression and tryptophan catabolism, Sci. Transl. Med 5 (2013) 193ra91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [246].Desbonnet L, Clarke G, O’Sullivan O, Cotter PD, Dinan TG, Cryan JF, Re: Gut microbiota depletion from early adolescence in mice: Implications for brain and behaviour, Brain Behav. Immun 50 (2015) 335–336. [DOI] [PubMed] [Google Scholar]
  • [247].Lai Y, Liu C-W, Chi L, Ru H, Lu K, High-Resolution Metabolomics of 50 Neurotransmitters and Tryptophan Metabolites in Feces, Serum, and Brain Tissues Using UHPLC-ESI-Q Exactive Mass Spectrometry, ACS Omega. 6 (2021) 8094–8103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [248].Anesi A, Rubert J, Oluwagbemigun K, Orozco-Ruiz X, Nöthlings U, Breteler MMB, Mattivi F, Metabolic Profiling of Human Plasma and Urine, Targeting Tryptophan, Tyrosine and Branched Chain Amino Acid Pathways, Metabolites. 9 (2019). 10.3390/metabo9110261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [249].Weber D, Oefner PJ, Hiergeist A, Koestler J, Gessner A, Weber M, Hahn J, Wolff D, Stämmler F, Spang R, Herr W, Dettmer K, Holler E, Low urinary indoxyl sulfate levels early after transplantation reflect a disrupted microbiome and are associated with poor outcome, Blood. 126 (2015) 1723–1728. [DOI] [PubMed] [Google Scholar]
  • [250].Zhen J, Zhao P, Li Y, Cai Y, Yu W, Wang W, Zhao L, Wang H, Huang G, Xu A, The Multiomics Analyses of Gut Microbiota, Urine Metabolome and Plasma Proteome Revealed Significant Changes in Allergy Featured with Indole Derivatives of Tryptophan, J. Asthma Allergy 15 (2022) 117–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [251].Esperanza MG, Wrobel K, Ojeda AG, Garay-Sevilla ME, Escobosa ARC, Barrientos EY, Wrobel K, Liquid chromatography-mass spectrometry untargeted metabolomics reveals increased levels of tryptophan indole metabolites in urine of metabolic syndrome patients, Eur. J. Mass Spectrom . 26 (2020) 379–387. [DOI] [PubMed] [Google Scholar]
  • [252].Fuertig R, Ceci A, Camus SM, Bezard E, Luippold AH, Hengerer B, LC-MS/MS-based quantification of kynurenine metabolites, tryptophan, monoamines and neopterin in plasma, cerebrospinal fluid and brain, Bioanalysis. 8 (2016) 1903–1917. [DOI] [PubMed] [Google Scholar]
  • [253].Guo H, Jiao Y, Wang X, Lu T, Zhang Z, Xu F, Twins labeling-liquid chromatography/mass spectrometry based metabolomics for absolute quantification of tryptophan and its key metabolites, J. Chromatogr. A 1504 (2017) 83–90. [DOI] [PubMed] [Google Scholar]
  • [254].Tuka B, Nyári A, Cseh EK, Körtési T, Veréb D, Tömösi F, Kecskeméti G, Janáky T, Tajti J, Vécsei L, Clinical relevance of depressed kynurenine pathway in episodic migraine patients: potential prognostic markers in the peripheral plasma during the interictal period, J. Headache Pain 22 (2021) 1–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [255].Desbonnet L, Garrett L, Clarke G, Bienenstock J, Dinan TG, The probiotic Bifidobacteria infantis: An assessment of potential antidepressant properties in the rat, J. Psychiatr. Res 43 (2008) 164–174. [DOI] [PubMed] [Google Scholar]
  • [256].Hervé C, Beyne P, Jamault H, Delacoux E, Determination of tryptophan and its kynurenine pathway metabolites in human serum by high-performance liquid chromatography with simultaneous ultraviolet and fluorimetric detection, J. Chromatogr. B Biomed. Sci. Appl 675 (1996) 157–161. [DOI] [PubMed] [Google Scholar]
  • [257].Jaglin M, Rhimi M, Philippe C, Pons N, Bruneau A, Goustard B, Daugé V, Maguin E, Naudon L, Rabot S, Indole, a Signaling Molecule Produced by the Gut Microbiota, Negatively Impacts Emotional Behaviors in Rats, Front. Neurosci 12 (2018). 10.3389/fnins.2018.00216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [258].Favre D, Mold J, Hunt PW, Kanwar B, ’ng Loke P, Seu L, Barbour JD, Lowe MM, Jayawardene A, Aweeka F, Huang Y, Douek DC, Brenchley JM, Martin JN, Hecht FM, Deeks SG, McCune JM, Tryptophan catabolism by indoleamine 2,3-dioxygenase 1 alters the balance of TH17 to regulatory T cells in HIV disease, Sci. Transl. Med 2 (2010) 32ra36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [259].Gatarek P, Kaluzna-Czaplinska J, Trimethylamine N-oxide (TMAO) in human health, EXCLI J. 20 (2021) 301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [260].Thomas MS, Fernandez ML, Trimethylamine N-Oxide (TMAO), Diet and Cardiovascular Disease, Curr. Atheroscler. Rep 23 (2021) 1–7. [DOI] [PubMed] [Google Scholar]
  • [261].He M, Yu H, Lei P, Huang S, Ren J, Fan W, Han L, Yu H, Wang Y, Ren M, Jiang M, Determination of Trimethylamine N-oxide and Betaine in Serum and Food by Targeted Metabonomics, Molecules. 26 (2021). 10.3390/molecules26051334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [262].Hefni ME, Bergström M, Lennqvist T, Fagerström C, Witthöft CM, Simultaneous quantification of trimethylamine N-oxide, trimethylamine, choline, betaine, creatinine, and propionyl-, acetyl-, and l-carnitine in clinical and food samples using HILIC-LC-MS, Anal. Bioanal. Chem 413 (2021) 5349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [263].Baranyi A, Meinitzer A, von Lewinski D, Rothenhäusler H-B, Amouzadeh-Ghadikolai O, Harpf H, Harpf L, Traninger H, Hödl R, Harb BM, Obermayer-Pietsch B, Schweinzer M, Braun CK, Enko D, Sex-specific differences in trimethylamine N-oxide (TMAO) concentrations before and after cardiac rehabilitation in acute myocardial infarction patients, EXCLI J. 21 (2022) 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [264].Garcia E, Wolak-Dinsmore J, Wang Z, Li XS, Bennett DW, Connelly MA, Otvos JD, Hazen SL, Jeyarajah EJ, NMR quantification of trimethylamine-N-oxide in human serum and plasma in the clinical laboratory setting, Clin. Biochem 50 (2017) 947–955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [265].Li Z, Suslick KS, Ultrasonic Preparation of Porous Silica-Dye Microspheres: Sensors for Quantification of Urinary Trimethylamine N-Oxide, ACS Appl. Mater. Interfaces (2018). 10.1021/acsami.8b00995. [DOI] [PubMed] [Google Scholar]
  • [266].Kanemitsu Y, Asaji K, Matsumoto Y, Tsukamoto H, Saigusa D, Mukawa C, Tachikawa T, Abe T, Tomioka Y, Simultaneous quantitative analysis of uremic toxins by LC–MS/MS with a reversed-phase/cation-exchange/anion-exchange tri-modal mixed-mode column, Journal of Chromatography B. 1068-1069 (2017) 1–8. [DOI] [PubMed] [Google Scholar]
  • [267].Veeravalli S, Karu K, Phillips IR, Shephard EA, A highly sensitive liquid chromatography electrospray ionization mass spectrometry method for quantification of TMA, TMAO and creatinine in mouse urine, MethodsX. 4 (2017) 310–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [268].Bain MA, Faull R, Fornasini G, Milne RW, Schumann R, Evans AM, Quantifying trimethylamine and trimethylamine-N-oxide in human plasma: interference from endogenous quaternary ammonium compounds, Anal. Biochem 334 (2004) 403–405. [DOI] [PubMed] [Google Scholar]
  • [269].Cohen LJ, Esterhazy D, Kim S-H, Lemetre C, Aguilar RR, Gordon EA, Pickard AJ, Cross JR, Emiliano AB, Han SM, Chu J, Vila-Farres X, Kaplitt J, Rogoz A, Calle PY, Hunter C, Bitok JK, Brady SF, Commensal bacteria make GPCR ligands that mimic human signalling molecules, Nature. 549 (2017) 48–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [270].Cho W, York AG, Wang R, Wyche TP, Piizzi G, Flavell RA, Crawford JM, N-Acyl Amides from Neisseria meningitidis and Their Role in Sphingosine Receptor Signaling, Chembiochem. 23 (2022) e202200490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [271].Cohen LJ, Kang H-S, Chu J, Huang Y-H, Gordon EA, Reddy BVB, Ternei MA, Craig JW, Brady SF, Functional metagenomic discovery of bacterial effectors in the human microbiome and isolation of commendamide, a GPCR G2A/132 agonist, Proc. Natl. Acad. Sci. U. S. A 112 (2015) E4825–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [272].Tørring T, Shames SR, Cho W, Roy CR, Crawford JM, Acyl Histidines: New N-Acyl Amides from Legionella pneumophila, Chembiochem. 18 (2017) 638–646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [273].Tan B, Yu YW, Monn MF, Hughes HV, O’Dell DK, Walker JM, Targeted lipidomics approach for endogenous N-acyl amino acids in rat brain tissue, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci 877 (2009) 2890–2894. [DOI] [PubMed] [Google Scholar]
  • [274].Landman C, Grill J-P, Mallet J-M, Marteau P, Humbert L, Le Balc’h E, Maubert M-A, Perez K, Chaara W, Brot L, Beaugerie L, Sokol H, Thenet S, Rainteau D, Seksik P, Quévrain E, on behalf of the Saint Antoine IBD Network, Inter-kingdom effect on epithelial cells of the N-Acyl homoserine lactone 3-oxo-C12:2, a major quorum-sensing molecule from gut microbiota, PLoS One. 13 (2018) e0202587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [275].Bradshaw HB, Johnson CT, Measuring the Content of Endocannabinoid-Like Compounds in Biological Fluids: A Critical Overview of Sample Preparation Methodologies, Endocannabinoid Signaling. (2023) 21–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [276].Syed SK, Bui HH, Beavers LS, Farb TB, Ficorilli J, Chesterfield AK, Kuo M-S, Bokvist K, Barrett DG, Efanov AM, Regulation of GPR119 receptor activity with endocannabinoid-like lipids, Am. J. Physiol. Endocrinol. Metab 303 (2012) E1469–78. [DOI] [PubMed] [Google Scholar]
  • [277].Balvers MGJ, Verhoeckx KCM, Witkamp RF, Development and validation of a quantitative method for the determination of 12 endocannabinoids and related compounds in human plasma using liquid chromatography–tandem mass spectrometry, Journal of Chromatography B. 877 (2009) 1583–1590. [DOI] [PubMed] [Google Scholar]
  • [278].Saini RK, Prasad P, Shang X, Keum Y-S, Advances in Lipid Extraction Methods-A Review, Int. J. Mol. Sci 22 (2021). 10.3390/ijms222413643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [279].Röhrig W, Achenbach S, Deutsch B, Pischetsrieder M, Quantification of 24 circulating endocannabinoids, endocannabinoid-related compounds, and their phospholipid precursors in human plasma by UHPLC-MS/MS, J. Lipid Res 60 (2019) 1475–1488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [280].Sempio C, Klawitter J, Jackson M, Freni F, Shillingburg R, Hutchison K, Bidwell LC, Christians U, Klawitter J, Analysis of 14 endocannabinoids and endocannabinoid congeners in human plasma using column switching high-performance atmospheric pressure chemical ionization liquid chromatography–mass spectrometry, Anal. Bioanal. Chem 413 (2021) 3381–3392. [DOI] [PubMed] [Google Scholar]
  • [281].Tan B, O’Dell DK, Yu YW, Monn MF, Hughes HV, Burstein S, Walker JM, Identification of endogenous acyl amino acids based on a targeted lipidomics approach, J. Lipid Res 51 (2010) 112–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [282].Dubik M, Pilecki B, Moeller JB, Commensal Intestinal Protozoa—Underestimated Members of the Gut Microbial Community, Biology . 11 (2022) 1742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [283].Li K, Su Z, Gao Y, Lin X, Pang X, Yang B, Tao H, Luo X, Liu Y, Zhou X, Cytotoxic Minor Piericidin Derivatives from the Actinomycete Strain Streptomyces psammoticus SCSIO NS126, Mar. Drugs 19 (2021). 10.3390/md19080428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [284].Shi S, Cui L, Zhang K, Zeng Q, Li Q, Ma L, Long L, Tian X, Streptomyces marincola sp. nov., a Novel Marine Actinomycete, and Its Biosynthetic Potential of Bioactive Natural Products, Front. Microbiol 13 (2022) 860308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [285].Guzmán-Salas S, Weber A, Malci A, Lin X, Herrera-Molina R, Cerpa W, Dorador C, Signorelli J, Zamorano P, The metabolite p-cresol impairs dendritic development, synaptogenesis, and synapse function in hippocampal neurons: Implications for autism spectrum disorder, J. Neurochem 161 (2022) 335–349. [DOI] [PubMed] [Google Scholar]
  • [286].Bermudez-Martin P, Becker JAJ, Caramello N, Fernandez SP, Costa-Campos R, Canaguier J, Barbosa S, Martinez-Gili L, Myridakis A, Dumas M-E, Bruneau A, Cherbuy C, Langella P, Callebert J, Launay J-M, Chabry J, Barik J, Le Merrer J, Glaichenhaus N, Davidovic L, The microbial metabolite p-Cresol induces autistic-like behaviors in mice by remodeling the gut microbiota, Microbiome. 9 (2021) 157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [287].Kawanabe-Matsuda H, Takeda K, Nakamura M, Makino S, Karasaki T, Kakimi K, Nishimukai M, Ohno T, Omi J, Kano K, Uwamizu A, Yagita H, Boneca IG, Eberl G, Aoki J, Smyth MJ, Okumura K, Dietary Lactobacillus-Derived Exopolysaccharide Enhances Immune-Checkpoint Blockade Therapy, Cancer Discov. 12 (2022) 1336–1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [288].Gaignard C, Laroche C, Pierre G, Dubessay P, Delattre C, Gardarin C, Gourvil P, Probert I, Dubuffet A, Michaud P, Screening of marine microalgae: Investigation of new exopolysaccharide producers, Algal Research. 44 (2019) 101711. [Google Scholar]

RESOURCES