Next Article in Journal
Morin Flavonoid Interaction with Albumin and Its Nanoparticle Conjugation: An Efficient Antioxidant Vehicle for Nutraceuticals
Previous Article in Journal
Low-Temperature Stress-Induced Hepatic Injury in Darkbarbel Catfish (Pelteobagrus vachelli): Mediated by Gut–Liver Axis Dysregulation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Does Gut Microbial Methylglyoxal Metabolism Impact Human Physiology?

by
Oluwatomisono I. Akinrimisi
1,
Kim Maasen
2,
Jean L. J. M. Scheijen
2,3,
Ina Nemet
4,5,
Max Nieuwdorp
1,6,
Casper G. Schalkwijk
2,3 and
Nordin M. J. Hanssen
1,6,*
1
Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Science Institute, Amsterdam University Medical Center, location AMC, 1105 AZ Amsterdam, The Netherlands
2
CARIM School for Cardiovascular Disease, Maastricht University, 6229 ER Maastricht, The Netherlands
3
Department of Internal Medicine, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands
4
Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
5
Center for Microbiome & Human Health, Cleveland Clinic, Cleveland, OH 44195, USA
6
Diabeter Center Amsterdam, 1066 EC Amsterdam, The Netherlands
*
Author to whom correspondence should be addressed.
Antioxidants 2025, 14(7), 763; https://doi.org/10.3390/antiox14070763
Submission received: 16 May 2025 / Revised: 15 June 2025 / Accepted: 18 June 2025 / Published: 21 June 2025
(This article belongs to the Special Issue Novel Antioxidant Mechanisms for Health and Diseases, 2nd Edition)

Abstract

Methylglyoxal (MGO) is a highly reactive dicarbonyl associated with oxidative stress, inflammation, and chronic diseases, particularly diabetic vascular complications and atherosclerosis through the formation of advanced glycation end products (AGEs). In the setting of human/host diseases, the formation of MGO has mainly been considered as the byproduct of glycolysis. Gut microbes play an important role in the development of cardiometabolic diseases. Here, we discuss a possibility that gut microbes can modulate the MGO pool within the host through (i) the alternation of the host metabolism, and (ii) direct MGO synthesis and/or detoxification by human commensal microorganisms. We also explore how dietary MGO impacts the composition of the gut microbiota and their potential role in modulating host health. This paradigm is highly innovative, with the current literature providing observations supporting this concept. Targeting the gut microbiome is emerging as an approach for treating cardiometabolic diseases through dietary, pre-, pro-, and postbiotic interventions, faecal microbiota transplantations, and the use of small molecule inhibitors of microbial enzymes. This can be a novel strategy to reduce MGO stress in the setting of cardiometabolic diseases and lowering the burden of diabetic complications and cardiovascular disease.

Graphical Abstract

1. Introduction

Methylglyoxal (MGO) is a highly reactive dicarbonyl that reacts with nucleophilic protein and DNA residues and forms advanced glycation end products (AGEs) that contribute to cell and tissue dysfunctions [1]. Within the host, MGO is formed by the spontaneous degradation of glycolytic intermediates. MGO is further metabolised by the evolutionarily conserved glyoxalase system [2,3], which converts MGO to D-lactate. The formation of MGO in human cells and its physiological implications have been well-documented in previous reviews [1,4]. Gut microbes have gained recognition for their important role in maintaining human health. Large cohort studies have repeatedly reported perturbations of the gut microbial community in a context of cardiometabolic diseases [5,6]. Microbial transplantation from mice with glucose intolerance into germ-free mice induce glucose intolerance [7]. In a human faecal microbiota transplant study, a faecal transfer from lean donors into individuals with metabolic syndrome resulted in an increase in gut microbial diversity and improved insulin sensitivity [8]. These data support the hypothesis that the dysbiosis of the gut microbiome contributes to metabolic dysfunction; however, how gut microbial alterations contribute to the metabolic diseases is still not fully understood. Here, we discuss the potential role of gut microbes in influencing the host’s pool of MGO through several mechanisms. Gut microbes may modulate the host MGO/AGE pool by altering the host metabolism. Additionally, microbes, including several human commensals, possess enzymatic capacities to either synthesise or degrade MGO [9,10,11,12,13,14], thereby directly affecting its production and/or detoxification. We also explore how dietary MGO impacts the overall composition of the gut microbiota and their potential role in modulating host health.

2. Host Production of Methylglyoxal: Linking Transient Increases in Glycolysis to Long-Term Cellular Dysfunction

In human cells, MGO is predominantly produced by the spontaneous degradation of the glycolytic intermediates glyceraldehyde-3-phosphate (G3P) and dihydroxyacetone phosphate (DHAP). The MGO content in food and beverages is relatively low, typically accounting for less than 1% of total human MGO exposure [1,15], and ~3% comes from the catabolism of threonine [1,16] and ~10% from the degradation of glycated proteins [1,17], but the majority of human MGO exposure, ~90%, is from endogenous formation during glycolysis, with most (~98%) being detoxified by the glyoxylase system in the cells under normal conditions [1,18]. Approximately 0.1–0.4% of the total glycolytic flux results in MGO formation [1,19,20,21,22] and is reported to be formed intracellularly at concentrations of 1–4 μM [16,23,24]. MGO is converted into D-lactate by the glyoxalase system composed of two enzymes, glyoxalase 1 (GLO1) and glyoxalase 2 (GLO2). GLO1, a rate-limiting enzyme, converts hemithioacetal spontaneously formed from MGO and reduced glutathione into S-D-lactoylglutathione. S-D-Lactoylglutathione is then hydrolysed by GLO2 to D-lactate with the accompanied regeneration of glutathione [25]. MGO that escapes the glyoxalase system spontaneously, non-enzymatically, reacts with proteins and DNA, changing their structure and function through the formation of AGEs. MGO reacts primarily with arginine guanidino residues and forms Nδ-(5-hydro-5-methyl-4-imidazolon-2-yl) ornithine (MG-H1), argpyrimidine, and tetrahydroimidazolone (THP), as well as with lysine amino group to form Nε-(carboxyethyl)lysine (CEL). N2-(1-Carboxyethyl)-2′-deoxyguanosine (CEdG) is a major MGO-derived DNA adduct [26]. The increased formation of MGO, thus, leads to the erosion of the intracellular milieu by modifying proteins and DNA, impairing normal cellular functions. Furthermore, MGO is thought to exert epigenetic effects by the direct modification of histones at critical lysine and arginine residues involved in both the nucleosome stability and reader domain binding altering gene expression [27]. Additionally, the induction of MGO production through hyperglycemia induced epigenetic changes in the promoter of the NF-κB p65 subunit in both cultured human aortic endothelial cells and in nondiabetic mice [28]. The accumulation of MGO can further impair cell energy homeostasis via mitochondrial dysfunction [29], the induction of oxidative stress, and the impaired availability of nitric oxide [30]. The clearance of these cellular modifications and impairments is further hampered by the modification of key proteasomal subunits by MGO [31]. Together, these processes may lead to a cycle of the accumulation of MGO and the build-up of modified proteins. The overexpression of Glo1 in bovine endothelial cells reduced intracellular hyperglycemia and the formation of AGEs [32], indicating that MGO is a major precursor in the formation of AGEs. Higher levels of MGO also drive GLO1 expression via its upstream regulator nuclear factor erythroid 2-related factor 2 (Nrf2) [33]. The mechanism linking glycolysis to glyoxalase activity has been partly unravelled, wherein MGO modifies KEAP1 to form a complex that enhances the transcription of NRF2 [34].

2.1. Infections—An Emerging Inducer of Methylglyoxal Stress

TNF alpha (Tumor Necrosis Factor alpha) is a pro-inflammatory cytokine, involved in the immune response and produced by macrophages. TNF alpha reduced the glyoxalase activity in cultured macrophages [35] and fibrosarcoma cell line L929 through the induction of GLO1 phosphorylation by protein kinase A (PKA), reducing its activity and the concomitant accumulation of MGO to cytotoxic levels [36]. Given its cytotoxic properties, the accumulation of MGO may aid in the inflammatory functions of TNF alpha, and we speculate that the production of MGO by the host serves as an antimicrobial agent during bacterial infections [37]. This is supported by a study demonstrating an increased MGO production during a mycobacterium infection of macrophages [38]. Elevated MGO levels have also been observed in individuals with septic shock [39,40], a condition characterised by a dysregulated immune response to infection. Furthermore, the glyoxalase system has been proposed as a defence mechanism in Group A Streptococcus against MGO produced by neutrophils [41]. Additionally, MGO detoxification has been shown to function as a virulence mechanism in Salmonella typhimurium [42] and Listeria monocytogenes [43], indicating that pathogens possess systems to counteract host-produced MGO [43].

2.2. How Essential Is Glyoxalase 1 for Maintaining Methylglyoxal Levels?

Drosophila Glo1 knockouts exhibited elevated MGO levels, increased insulin resistance, and greater weight gain, and developed hyperglycemia, although they remained viable [44]. In zebrafish, Glo1 deficiency did not alter normal development, likely due to the upregulation of compensatory pathways such as aldehyde dehydrogenase, and showed an enhanced sensitivity to insulin resistance and vascular injury in the retina [45]. Glo1 knockouts in Schwann cells showed no increase in MGO; there was an alternative detoxification of MGO by aldose reductase [46]. In addition, Glo1 knockout mice were susceptible to acetaminophen-induced hepatotoxicity [47]; Glo1 knockdown mice had a diabetic-nephropathy-like phenotype [48], while the manipulation of GLO1 activity did not alter the atherosclerotic burden in atherosclerosis-prone Apoe-/- mice [49]. These findings suggest that MGO plays a role in modulating the organismal physiology and reveal the complexity of its detoxification.

3. Methylglyoxal and the Microbiome

The interplay between MGO, the microbiome, and the host is complex. Although it has not been investigated definitively, various works in the literature elude to the concept that the microbiome may have a role to play in influencing the gut MGO pool and, in turn, systemic MGO levels, either through the diffusion of MGO across the intestinal barrier, or in a case where a disturbed intestinal barrier integrity allows microbial toxic compounds to enter the blood stream or, in fact, the microbes themselves [50,51]. Firstly, certain microbes possess specialised MGO metabolism genes with functions not found or demonstrated in humans such as methylglyoxal synthase (mgsA), glyoxylase 3 (glo3), and 1,2-propanediol oxidoreductase. Second, as described in Section 2.1, MGO may serve as a mediator between microbes and immune cells, particularly during infections. Third, SCFAs, which are microbial metabolites, have been shown to modulate MGO levels, and MGO itself may influence SCFA production [14,52,53]. Additionally, the dietary MGO present in various food and beverages interacts with gut microbes. Given the microbe’s role in modulating the host digestion and metabolism, the microbiome may indirectly regulate MGO concentrations in the host. Subsequent sections will explore the relevant literature regarding microbial communities and host MGO metabolism in more detail. Figure 1 illustrates the potential fates of MGO within the context of the host gut microbiome interactions.
Unlike in the host where MGO is produced spontaneously, i.e., non-enzymatically, microbes, including multiple commensals, produce MGO through the catalytic dephosphorization of dihydroxyacetone phosphate, by MgsA encoded by the mgsA gene [54]. Another microbial pathway in the formation of MGO is from aminoacetone generated from glycine and threonine via the oxidoreductase [55,56,57]. The microbial MGO metabolism has been demonstrated primarily through experiments using microbes grown in culture media [9,58,59,60,61,62,63,64]. Relatively few studies have explored the broader impact of MGO on the microbiome as a whole; these are summarised in Table 1 and Figure 2. There is limited overlap between studies, with studies also having differing methodologies and research objectives (Table 1). Microbial taxa show a positive association with MGO, with increased abundance, while others display a negative association (Table 1). We hypothesise that a positive association is due to intrinsic tolerance or the ability to metabolise MGO, whereas negative associations may arise from a lack of tolerance to MGO or from competitive exclusion by other microbes that are better adapted to utilise MGO as a resource within the microbial community. Across both clinical and experimental studies, MGO was shown to selectively influence the composition of the microbiome. Several taxa such as Ruminococcaceae [65,66], Bifidobacterium [52], and Lactobacilli [53] showed negative associations with MGO exposure, while potentially pro-inflammatory or opportunistic genera like Escherichia-Shigella, Klebsiella [52], and Bacteroides forsythus [67,68] tend to be enriched. Akkermansia displays context-dependent behaviour, appearing as both positively [65] and negatively [66] associated with MGO depending on the host condition and study model. While the in vivo data from aged mice revealed no significant changes in the overall alpha or beta diversity [65], the in vitro studies report both marked compositional shifts in microbial abundance [53] and demonstrated microbial differences using principal coordinate analysis [52]. These findings suggest that MGO does not necessarily alter global diversity metrics but can drive substantial taxonomic and functional shifts. We also believe that such changes are likely dependent on the dose of MGO used. Clinically, elevated MGO levels have been associated with conditions like rectal cancer [66] and periodontitis [67,68], where they correlate with specific microbial signatures, indicating that MGO may act not only as a microbial modulator but also as a potential biomarker, with the disease-related microbial shifts requiring further investigation.
Short-chain fatty acids (SCFAs), including the popular acetate, propionate, and butyrate, are important microbial metabolites. In Proteus and Morganella spp., the addition of butyric and propionic acids has been shown to reduce MgsA activity and decrease the MGO concentration in the culture medium [14]. SCFAs are produced by the gut microbe fermentation of dietary fibers; conditions present in the gut shaped by the already colonizing bacteria could influence the microbial production of methylglyoxal [14]. In the other direction, evidence also indicates that MGO may impact SCFA production. In in vitro fermentation using human faecal samples, the supplementation of MGO led to a reduction in SCFA levels for acetate, propionate, and butyrate [52]. A study simulating the digestion of dicarbonyl compounds in vitro and using faecal samples from three human donors demonstrated that MGO exposure resulted in diminished SCFA production [53]. Taken together, these findings suggest that MGO in high concentrations may exert detrimental effects on the gut microbiome, by reducing the number of beneficial bacteria capable of SCFA production.

3.1. Theories on Why Microorganisms Produce Methylglyoxal

Given the fact that MGO is cytotoxic, its active production by microbes seems counterintuitive. One hypothesis to explain this is that MGO production is a microbial adaptation to nutrient imbalance, particularly under conditions of excess carbon availability. In such environments, key glycolytic intermediates may accumulate to toxic levels, disrupting normal metabolic function [69,70]. MGO may act as a metabolic overflow pathway, helping to regulate the carbon flux and prevent the buildup of glycolytic intermediates [54,69]. Although MGO may be inherently toxic to the cell, microbes possess active protective mechanisms such as the glyoxalase system that converts MGO into less harmful compounds like D-lactate. This detoxification strategy allows microbes to temporarily tolerate MGO while benefiting from its role in maintaining the metabolic balance [69]. Overall, the hypothesis suggests that MGO production offers a short-term advantage during environmental shifts, when microbes encounter nutrient-rich but imbalanced conditions. Another theory is focused on the regulation of phosphate availability. MgsA converts DHAP into MGO, releasing phosphate in the process. This phosphate can then be utilised in the glyceraldehyde-3-phosphate dehydrogenase reaction during glycolysis [54]. Thus, MgsA may play a role in generating phosphate for glycolytic processes [54]. Moreover, the Catabolite Repression HPr-like protein (Crh) was found to regulate MgsA activity in Bacillus subtilis, with its non-phosphorylated form binding to and inhibiting MgsA [71]. Preferred carbon sources such as glucose or mannitol trigger the phosphorylation of Crh and reduce its binding to MgsA [71], suggesting that MGO is produced when the preferred carbon sources are present [71]. Lastly, the mechanisms of microbial cross-feeding in the gut are well-highlighted [72]; cross feeding involves the sharing of metabolites, including 1,2-propanediol [72] between microbes. In such systems, the metabolites of one microbe are released and utilised by another, fostering interactions that range from mutualism to competition. From a microbial ecological perspective, we speculate some microbes may release MGO into the environment to other microbes or as a way to inhibit the growth of other microbes. A metabolic flux analysis in Escherichia coli reveals a dynamic response to changing nutrient conditions, “famine” (increased glucose availability) and “feast” (decreased glucose availability). During feast conditions, the excretion of MGO and lactate occurred rapidly, followed by acetate at a later stage [73]. MGO levels in the medium reached 0.6–0.9 mg/L [73], well below the reported threshold of 20 mg/L that can inhibit microbial growth [73]. In contrast, under famine conditions, no byproducts were detected in the culture medium. This indicates that the MGO pathway is tightly regulated in Escherichia coli; even the overexpression of mgsA caused the accumulation of only ~0.08 mM MGO in the glucose medium [74], Escherichia coli is reported to lose viability at 0.6–0.7 mM MGO [69]. In Saccharomyces cerevisiae, MGO accounts for 0.3% of the glycolytic flux [62].

3.2. Microbial Detoxification of Methylglyoxal

Similar to the host MGO detoxification routes, bacteria also have a glyoxalase system responsible for the detoxification of MGO into D-lactate. Furthermore, another route identified in Escherichia coli is the glutathione-independent glyoxalase III (Glo3) where MGO is converted directly to D-lactate and is glutathione-independent [75]. Glo3 has primarily been characterised and its activity demonstrated in Escherichia coli [76,77]. The hchA gene, which encodes Hsp31 (Glo3), is important for MGO resistance and Hsp31 is a DJ-1 homolog that is widely evolutionarily distributed [76]; hchA-deficient strains of Escherichia coli in the stationary phase showed an increased sensitivity to MGO and accumulated higher levels of intracellular MGO [77]. The first physiologically relevant demonstration of glutathione-independent Glo3 activity in fungi was reported in Candida albicans, where the homozygous glx3 knockout mutants displayed sensitivity to exogenous MGO, and had increased intracellular MGO, and carbon-source-specific growth defects [78]. In contrast, GLO3 has not yet been identified in humans [79]. Other conversion routes include the enzymes alcohol dehydrogenase and methylglyoxal reductase [69,80], although the physiological relevance of these enzymes in Escherichia coli is questionable due to their low affinities for MGO [69]. Additionally, we have glycerol dehydrogenase, which converts MGO to D-lactaldehyde, and 1,2-propanediol oxidoreductase, which converts L-lactaldehyde to 1,2-propanediol [76]. A variety of microbes have been reported to produce 1,2-propanediol [81,82,83]; however, the conditions and substrate for production vary, with varied pathways for 1,2-propanediol production including the deoxyhexose pathway, lactate pathway, and MGO pathway described in this review [82]. The MGO pathway has been described previously where sugars such as glucose and fructose are substrates for glycolysis and MgsA converts the glycolytic intermediate DHAP into MGO. MGO is subsequently reduced to 1,2-propanediol through D-lactaldehyde in the presence of glycerol dehydrogenase, and then 1,2-propanediol oxidoreductase, or through acetol in the presence of alcohol dehydrogenase, and then glycerol dehydrogenase [82]. In Clostridium thermosaccharolyticum, phosphate concentrations of up to 113 mM had no significant effect on product (1,2-propanediol/acetol) formation through the MGO pathway [84], contrary to other reports where MgsA is inhibited by the presence of phosphate [54,85]. Additionally, Escherichia coli activates the potassium efflux systems KefB and KefC in response to the formation of a glutathione conjugate [69]. Homologs of KefC have been identified in other Gram-negative bacteria, including Salmonella typhimurium and Enterobacter aerogenes. This system appears to be absent in Gram-positive bacteria [69,86], with the exception of Staphylococcus aureus [86]. These systems may help protect the cell from MGO toxicity by exporting potassium ions out of the bacterial cell [87,88], accompanied by an influx of sodium ions and protons. This ion exchange leads to cytoplasmic acidification, resulting in a lower intracellular pH [89,90]. The proposed model suggests that this acidification reduces the harmful interactions between MGO and macromolecules, and may also stimulate DNA and protein repair mechanisms [69].

3.3. The Microbiome as a Potential Therapeutic Target to Lower Methylglyoxal Stress in the Host

To date, no known mice or human studies have investigated altering the gut microbiome using probiotic strains, commensal microbes, or faecal microbiota transplantation (FMT) with the specific aim of reducing systemic MGO levels in the host, although the influence of the microbiome on host health and metabolism is of increasing interest [91,92]. The microbiome has been reported to influence the host energy intake [93], metabolism of macronutrients [91] and micronutrients [94], dictate the appetite via the brain–gut axis [95,96], and modulate the immune system [97]. Microbes can also influence host metabolite levels which have diverse effects on the host [98,99]. SCFAs, produced by gut microbes from dietary fiber, are recognised as beneficial metabolites that help restore intestinal barrier integrity [100], support blood glucose regulation, enhance insulin sensitivity in individuals with type 2 diabetes [101,102], and carry out the regulation of energy homeostasis, and body weight control [103,104]. Thus, we hypothesise that the microbiome may influence MGO stress via host metabolic regulation, whereby a state of microbial dysbiosis negatively impacts the host metabolism. As previously mentioned, microbes possess several enzymes for MGO detoxification (Glo1, Glo2, and Glo3) and MGO synthesis (MgsA). Thus, the gut microbiome may act as a reservoir for MGO, allowing its entry into systemic circulation via the portal vein. Targeting the microbiome through supplementation with beneficial strains may help mitigate MGO-related stress. Lactobacillus spp. have gained popularity as probiotics due to their associated health benefits [105]. Interestingly, Lactobacilli sp. have been shown to convert MGO into 1,2-propanediol [9], while, in a mouse study, Lactobacilli were found to potentially prevent liver injury by stimulating the antioxidant pathway Nuclear factor Nrf2 [106], which is reported to help reduce MGO stress by promoting a GSH increase [107], and regulate the expression of GLO1 [33]. This suggests that the microbiome may function as a first line of defense against exogenous MGO, either by converting it into other metabolites or by directly neutralizing it. Conversely, in cases of gut dysbiosis, there may be an increase in MGO-producing proteins or enhanced MGO production by immune cells, which could result from an imbalance in microbial populations. For instance, the gut microbial production of MGO by Escherichia coli accelerated ageing in Caenorhabditis elegans [108], demonstrating that microbial MGO can directly impact the host physiology. Further research is needed in order to explore these hypotheses and advance our understanding for the greater good.

4. Dietary Methylglyoxal

Besides endogenous formation, we are also exposed to MGO through exogenous sources, mainly through diet. MGO is formed during food processing and preparation, as part of non-enzymatic browning (Maillard reaction), the caramelization reaction, and lipid peroxidation, or enzymatically by microorganisms in fermented foods [109]. The presence of MGO in foods and drinks has been evaluated in several studies, the most extensive of which is a comprehensive food composition database with an MGO content of 223 commonly consumed products quantified using the gold-standard UHPLC-MS/MS technique, covering a broad range of food groups, including vegetables, fruit, cheese, meat, fish, and ready-made products [110]. In this database, MGO concentrations ranged from 0.04 to 736 mg/kg, and were the highest in manuka honey, dried apricot, Dutch spiced cake, rusk, apple molasses, and digestive biscuit. MGO concentrations are generally high in fermented foods [15] and foods containing caramel or molasses (both rich in sugar and exposed to intense heat treatment). Several factors that impact dicarbonyl formation in foods have been described, i.e., the type of sugar used, the intensity of heat treatment and the accompanying reduction in water content, food processes such as ripening, fermentation, and storage, and the absence of reactants like amino acid side chains of proteins that may result in the subsequent formation of AGEs and, thus, reduce the dicarbonyl content [15]. In the Dutch population, MGO intake comes mainly from coffee consumption, followed by bread, and cookies/bakery products [110]. As stated previously, the MGO content in food and beverages is relatively low, typically accounting for less than 1% of total MGO exposure [1,15]. Several studies have attempted to perform the simulated digestion of MGO, yielding differing results [111,112,113]; simulated digestion experiments are different from actual conditions in the gastrointestinal tract [112]. Upon simulated in vitro gastrointestinal digestion, 5–20% of the initial MGO was recovered [114]; in addition, four healthy volunteers on a controlled diet were administered 500 μmol of MGO in manuka honey, and the renal excretion of MGO ranged from 0.1 to 0.4 μmol/day [114]. This study suggests that dietary MGO undergoes rapid degradation during intestinal digestion and, consequently, does not affect in vivo MGO levels [114]. On the contrary, in vitro gastrointestinal digestion (oral, gastric, and small intestinal phases) in another study reported ~75% intact MGO where they make it to the colon and can react with microbes [53], while a cross-sectional study in the Netherlands (The Maastricht Study) investigated the relationship between the habitual intake of MGO using dietary data. The study found that a higher habitual MGO intake was associated with lower levels of low-grade inflammation [115].

4.1. Methylglyoxal in Manuka Honey

Manuka honey is a honey derived from Leptospermum scoparium trees, which is known for its various therapeutic beneficial properties and very high MGO content [116,117], compared to other types of honey [118]. MGO has been reported to be responsible for the antimicrobial activity of manuka honey [118,119]. MGO derived from manuka honey has been shown to inhibit bacterial growth and prolong the lag phase of various bacterial species [120]. Manuka honey has also been shown to inhibit biofilm formation in bacterial species [121,122]. It has been reported that MGO from manuka honey may contribute to host mucosal-associated invariant T-cell-sensing microbes [123], which would be beneficial for immune homeostasis. MGO exhibits antibacterial activity against Gram-positive and Gram-negative bacteria [124,125,126]. Additionally, MGO has been shown to be effective against microbial biofilms [119] and multidrug-resistant strains [126], and enhance the susceptibility of microbes to antimicrobial agents [127,128]. MGO may prevent microbial growth through the damage of the structural integrity of bacterial flagella and fimbriae, leading to a loss of their function [124]. However, a mouse study reported that the dietary intake of manuka honey, which is rich in MGO, did not cause substantial alterations in the composition of the gut microbiota or the levels of microbial-derived SCFAs [129]. Although the authors of the study acknowledge some methodological limitations, for instance, the next-generation sequencing-based phylogenetic analysis may have identified more microbial taxa than the RT-qPCR method used in the study; conducting further research through stable isotope tracing and a flux analysis of the organic acid uptake, in addition, would have been beneficial. Lastly, in their study, manuka honey delivers ∼0.12 mM MGO [129]; they suspect higher concentrations may be required to show antimicrobial effects due to the loss of MGO during digestion processes [129].

4.2. Metabolism of Methylglyoxal in the Gut

MGO is a small compound, and it is conceivable that it is absorbed from foods in the intestine. Animal studies reported increased plasma concentrations of MGO after oral MGO administration via drinking water [130,131,132]. In humans, a small-scale study reported that a single oral dose of manuka honey containing a high amount of MGO (i.e., ~500 μmol) was given [114]. This single dose of MGO did not lead to changes in urinary MGO excretion, but no data on plasma MGO or AGEs was available. A recent human population cohort examined the habitual intake of MGO, and observed that a higher habitual intake of dietary MGO was associated with higher plasma concentrations of MGO and a higher skin autofluorescence, an estimate of skin AGEs that is thought to reflect AGEs in tissue [133]. Coffee and bread were the main dietary sources of MGO in this cohort, and the associations of dietary MGO with plasma MGO and with skin autofluorescence were mainly driven by coffee intake. These results suggest the dietary absorption of MGO, but we cannot exclude the possibility that MGO from the diet indirectly influences MGO in the body. A 1 mg/day higher intake of MGO (corresponding to, for example, ~400 mL coffee [110]) was associated with a 2–3% higher plasma MGO and skin autofluorescence. Notably, the concentrations of free MGO in (fasting) plasma might provide only a snapshot and be an underestimation of the actual contributions of dietary MGO. Besides the dietary absorption of MGO, some of the MGO from foods may react with proteins present in the gastrointestinal tract, for example, with proteins present in the food matrix [113,114,133], with digestive enzymes [134], or with intestinal proteins [111]. The modification of food proteins could lead to the de novo formation of AGEs, which can, thereafter, be absorbed or exert local effects in the gastrointestinal tract. The modification of digestive proteins may lead to altered digestion, whereas the modification of intestinal proteins may lead to gut inflammation, increased permeability, or an altered microbiome composition [109,135,136,137]. The reactivity of MGO during digestion has, so far, only been studied in vitro. Studies with human intestinal cells reported that MGO induced an increased secretion of the pro-inflammatory cytokines interleukin-6 (IL-6) and interleukin-8 (IL-8) [138,139]. A decrease in MGO concentration was observed in several studies during simulated gastrointestinal digestion [114,140,141]. However, an increase in MGO concentration has also been reported in soy sauce, possibly due to their release from reversibly-bound adducts [140]. This indicates that reactions during digestion may play an important role in the actual exposure to MGO throughout the gastrointestinal tract. It is important to note here that a reduction in MGO does not mean that they are harmless, per se, since the putative consequences of the reaction-products are unknown. Furthermore, it should be emphasised that these models might not represent in vivo digestion. Moreover, when investigating the gastrointestinal metabolism of MGO, it is essential that we consider the food matrix in which MGO is present. The release may be modulated by the complexity of the food matrix, or the presence of scavengers of MGO in the food matrix. Examples of potential scavengers are creatinine in meat [142], fiber-bound polyphenols from highland barley [52], and melanoidins in coffee, cacao, and bread, for which MGO-scavenging effects have been reported in vitro [143]. Thus, it is important that we consider the food matrix when predicting the effects of dietary MGO.

4.3. Consequences of Dietary Methylglyoxal

Studies in animals showed that high amounts of orally administered MGO increased vascular inflammation [131,144] and induced vascular changes such as impaired vasodilation [131,145], the thickening of the glomerular basement membrane [146], endothelial dysfunction [131], the induction of retinopathy-like changes, [147], and insulin resistance [148]. Moreover, the administration of exogenous MGO, either subcutaneously or intraperitoneally, induced β-cell dysfunction, insulin resistance and impaired glucose metabolism, and insulin sensitivity [149,150]. In contrast, a theory suggesting that MGO has beneficial effects within a hormetic model has been proposed, where MGO has harmful effects at high doses and beneficial effects at low doses [25]. A recent study in mice showed that the long-term exposure to orally administered MGO resulted in a moderate increase in plasma MGO; yet, this was accompanied by increased survival, less age-related solid tumors, and the absence of diabetes and renal insufficiency [130]. Moreover, in rats, a similar increase in plasma MGO after long-term administration with oral MGO was observed, but no differences were reported in oxidative stress and higher levels of antioxidants [132]. Similarly, the anti-oxidative effects of low doses of MGO were reported in vitro [34,151,152]. A possible explanation for this is that small increases in plasma MGO via exogenous intake may have beneficial effects, for example, through the upregulation of the antioxidant system and/or the glyoxalase system, a major pathway in the detoxification of MGO [151]. A large, cross-sectional cohort study in The Netherlands (The Maastricht Study) explored associations between habitual MGO intake (by matching the dietary data from the FFQ with our food composition database, which include the concentrations of MGO, GO, and 3-DG in more than 200 foods) and several health outcomes. Unexpectedly, a higher habitual intake of MGO was associated with lower low-grade inflammation scores [115]. Moreover, in the same cohort, a higher habitual MGO intake was associated with greater insulin sensitivity and less type 2 diabetes [153]. There were no consistent associations between MGO intakes and β-cell function. Possible explanations for the discrepancies between these studies in animals and humans are that experimental studies were often conducted with very high concentrations of MGO, out of the physiological range, whereas the exposure to MGO in humans is much lower. In addition, most studies in animals used a commercially available MGO, often contaminated with other reactive compounds. Additionally, it is also important to consider the food matrix when evaluating the role of dietary/exogenous MGO on the host. The associations between dietary MGO and health outcomes were mainly driven by coffee intake, thus far the largest identified contributor of MGO intake from the diet [153]. However, the effects of MGO in coffee cannot be disentangled from that of other compounds in coffee. Therefore, intervention studies with proper food matrix controls are needed in order to evaluate the role of dietary MGO on the host. Overall, these findings suggest that not only does MGO have harmful effects, but the consumption of dietary MGO might lead to small increases in plasma MGO, which may induce some beneficial effects.

5. Consequences of Methylglyoxal Stress

To date, no known mice or human studies have attempted to determine the proportional contributions of microbially derived MGO to the systemic circulation separate from MGO produced endogenously by the cells of the host. The predominant body of experimental literature has focused on either the exposure to (very) high levels of exogenous MGO or the manipulation of the glyoxalase system and quenchers of MGO in severe models of experimental diabetes, well-described in other reviews [1,4]. Therefore, we summarise the emerging evidence for the role of MGO in diabetic complications as well as other vascular and age-related conditions, and we provide hypotheses for how the manipulation of the microbiome may influence MGO stress.

5.1. Methylglyoxal and “Classic” Microvascular Diabetic Complications—Its Mechanisms of Action

MGO has been associated with neuroretinal dysfunction [147]; feeding rats 50 mM MGO in drinking water induced a retinopathy/like phenotype [147]. In line with this, the manipulation of intracellular MGO formation, by overexpressing Glo1, reduced diabetic retinopathy and nephropathy in rats [30,154]. A mouse study suggests that TRPC channels play a role in the accumulation of MGO, with the subsequent TRPC knockout mice protected against diabetic retinopathy, having lower MGO levels and increased GLO1 activity [155]. The gut–retina axis is a growing field of research, where the microbiota may play a role in disease severity. A study showed that feeding mice a diet of high glycemic load led to the acceleration of macular degeneration with the accumulation of AGEs; they found that bacterial species were associated with the pace of damage and protection [156].

5.2. Methylglyoxal as an Inducer of Cerebral Ageing and Cellular Senescence

Since endogenous MGO leads to oxidative stress and the formation of AGEs, the formation of MGO has been proposed to be one of the key pathways contributing to the ageing process. In the brain, the most energy-demanding organ with the poorest capacity to regenerate and divide, neurons largely convert lactate produced by astrocytes into pyruvate and acetyl-CoA to bypass the glycolytic steps that lead to the formation of MGO [21]. Such bypasses, however, do not seem to be afforded to peripheral nerves, as MGO is thought to be a key driver of diabetic neuropathy [157]. Senescence is emerging as a key cellular process involved in ageing. Cell senescence is a process where cell cycle arrests, and an associated senescence associated inflammatory signature (SASP) arises, leading to further dysfunction of the surrounding tissue. To what extent MGO induces cellular senescence in the strictest sense is still largely unknown. The gut microbial production of MGO by Escherichia coli accelerated ageing in Caenorhabditis elegans [108].
Exposure to MGO induces cell cycle arrest in human umbilical endothelial cells (HUVEC), without a clear increase in inflammatory signaling [158]. In contrast, in a mouse model of accelerated ageing by daily D-galactose injection, cortical MGO accumulation was linked to memory impairment, impaired glyoxalase activity, and increased inflammatory signaling [159]. Unfortunately, a single-cell approach to pinpointing the site of MGO accumulation in the brain is lacking, but older immunohistochemical studies using AGE staining suggest that the astrocytes and microglial cells are the primary cells of MGO accumulation [160], in line with their highest direct glycolytic rate. MGO has also been linked to direct signaling in the brain, either through binding directly to receptors and sodium channels, or through modifying neurotransmitters [161]. These observations together may aid the explanation that diabetes is both linked to depression as well as an increased risk for neurodegenerative disorders. We observed increased MGO-derived AGE levels in the brains of streptozotocin-injected rats, and we postulate that, by inducting endothelial dysfunction, MGO may contribute to the impairment of the blood–brain barrier, leading to further damage to the brain. In line with this hypothesis, Glo1 overexpression partly rescued the mild cognitive impairment in these rats [162]. Whether these mechanisms could be influenced by circulating MGO derived from excess production in the gut microbiome has not been studied, but it seems overall unlikely. Nonetheless, since the microbiome has established effects on insulin resistance, indirect effects of the gut microbiome on cerebral MGO levels are conceivable. Furthermore, the gut microbiome may produce MGO-quenching or GLO1-inducing compounds that alter MGO levels, but these kinds of potential mechanisms remain largely speculative.

5.3. Methylglyoxal and Cardiovascular Disease

Higher MGO levels are linked to an increased risk of cardiovascular events in type 1 diabetes [163] and type 2 diabetes, and in individuals with critical limb ischemia, regardless of diabetes status [164]. In carotid atherosclerotic plaques, we found that it was not the diabetes status but plaque macrophage content that was associated with higher plaque MGO levels [35]. Using immunohistochemical studies with antibodies against MG-H1, we determined the plaque shoulder bordering the necrotic core as the predominant site of MGO accumulation [35]. Oral and gut microbes are associated with plaques of patients with atherosclerosis [165]. Oral commensals have specifically been identified in the atherosclerotic plaques of patients with coronary artery disease [166]. Moreover, microbes can form biofilms in carotid arterial plaques and are believed to contribute to plaque rupture [167]. We previously found that overexpressing Glo1 failed to reduce the increased MGO levels in streptozotocin-injected mice, suggesting that there may be an additional, extracellular source of MGO, such as diet and/or a perturbed microbiome [168]. We are currently not aware of any antibiotic or faecal microbiota transplantation studies to shed further light on these issues. The complexity of this system is further underlined by a publication showing that either the overexpression or knockdown of Glo1-altered albuminuria in Apoe-deficient mice had no effect on atherosclerosis [49]. Therefore, the formation and detoxification of MGO is highly context-dependent. In addition to its effects on arteries, MGO seems to have direct detrimental effects on the cardiac muscle. Harm by MGO has been mostly described in the context of increased intracellular formation in the setting of diabetes, using Glo1-overexpressing rodents. One study found that Glo1 overexpression improved cardiac dysfunction in streptozotocin-injected mice, and that this was mainly due to an improvement in endothelial dysfunction. Largely in line with our atherosclerosis experiment, this study only found a modest reduction in dicarbonyl stress in the hearts of the Glo1-overexpressing mice [169].

5.4. Methylglyoxal and Cancer

The formation of MGO in cancer has mainly been studied in the setting of increased glycolysis by the so-called Warburg effect [170]. In turn, MGO may accelerate tumor progression by inducing a direct modification of DNA or by the induction of epigenetic signatures. Recently, it was, in fact, shown in breast cancer that MGO can bypass the seminal two-hit tumor suppressor hypothesis by inactivating the BRCA2 gene product because the MGO-modified protein undergoes proteolysis [171]. The role of exogenous MGO exposure was studied in breast and glioma cells, showing that exogenous MGO (at low doses) induced NRF2 and GLO2, and promoted tumor growth [33]. A small study of 26 individuals with rectal cancer identified MGO as the key metabolite produced by the faecal microbiome, predicting the response to chemotherapy [66]. Compared to wild-type cells, colorectal cancer cells with KRAS mutations survive better under MGO stress through AKT activation. Additionally, MGO stress makes wild-type cells resistant to cetuximab [172]. The link between endogenous and microbial MGO production and cancer is not straightforward. Nonetheless, these cancer studies raise the question as to whether similar interplays also occur in other organs.

6. Conclusions

MGO is a ubiquitous compound produced endogenously by the host metabolism and by microbes in the gut, and is present in various dietary sources. A review of the current literature suggests that the effects of MGO are highly context- and dose-dependent, with elevated endogenous exposure more commonly associated with detrimental outcomes. While the mechanisms by which MGO influences cellular physiology have been well-characterised, and the critical role of the microbiome in human health is increasingly recognised, relatively few studies have bridged the gap between MGO biology and host–microbiome interactions. To advance this field, future research should prioritise the use of stable-isotope-labeled MGO to accurately trace its gastrointestinal fate, microbial transformation, and systemic absorption. In addition, in vivo experiments are needed in order to elucidate how microbes interact with MGO within complex microbial communities, rather than in isolated cultures with study designs involving antibiotics, faecal microbiota transplantation, and germ-free mice. Lastly, large-scale population studies examining the associations between dietary MGO intake, circulating MGO levels, and health outcomes will also be critical. While the role of microbially derived MGO in human physiology remains unclear, the tools and methodologies now exist to explore this important frontier in host–microbe interactions.

Author Contributions

Conceptualization: M.N. and C.G.S.; writing: O.I.A., K.M., and N.M.J.H.; review and editing: J.L.J.M.S. and I.N.; visualization: O.I.A.; final editing: O.I.A. and N.M.J.H.; funding acquisition: N.M.J.H. All authors have read and agreed to the published version of the manuscript.

Funding

O.I.A. and N.M.J.H. are supported by a ZONMW-VIDI grant 2023 (09150172210019). NMJH is supported by a Senior Clinical Dekker grant by the Dutch Heart Foundation (grant number 2021T055). M.N. is supported by a personal NWO VICI grant 2020 (09150182010020) and an ERC Advanced Grant (101141346).

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

N.M.J.H. has received a honorarium from Novo Nordisk, Bayer, and Boehringer Ingelheim. M.N. is the founder and a scientific advisory board member of Caelus Pharmaceuticals and Advanced Microbiome Interventions, the Netherlands. However, none of these bear any relevance to the content of the current paper.

Abbreviations

AGEsAdvanced glycation end products
DHAPDihydroxyacetone phosphate
FFQsFood Frequency Questionnaires
G3PGlyceraldehyde-3-phosphate
GLO1Glyoxalase 1
GLO2Glyoxalase 2
GSHGlutathione
MGOMethylglyoxal
SCFAShort-chain fatty acids

References

  1. Schalkwijk, C.G.; Stehouwer, C.D.A. Methylglyoxal, a Highly Reactive Dicarbonyl Compound, in Diabetes, Its Vascular Complications, and Other Age-Related Diseases. Physiol. Rev. 2020, 100, 407–461. [Google Scholar] [CrossRef] [PubMed]
  2. Inoue, Y.; Kimura, A. Methylglyoxal and regulation of its metabolism in microorganisms. Adv. Microb. Physiol. 1995, 37, 177–227. [Google Scholar] [PubMed]
  3. Ranganathan, S.; Walsh, E.S.; Godwin, A.K.; Tew, K.D. Cloning and characterization of human colon glyoxalase-I. J. Biol. Chem. 1993, 268, 5661–5667. [Google Scholar] [CrossRef]
  4. Berends, E.; van Oostenbrugge, R.J.; Foulquier, S.; Schalkwijk, C.G. Methylglyoxal, a highly reactive dicarbonyl compound, as a threat for blood brain barrier integrity. Fluids Barriers CNS 2023, 20, 75. [Google Scholar] [CrossRef] [PubMed]
  5. Baars, D.P.; Fondevila, M.F.; Meijnikman, A.S.; Nieuwdorp, M. The central role of the gut microbiota in the pathophysiology and management of type 2 diabetes. Cell Host Microbe 2024, 32, 1280–1300. [Google Scholar] [CrossRef]
  6. Fromentin, S.; Forslund, S.K.; Chechi, K.; Aron-Wisnewsky, J.; Chakaroun, R.; Nielsen, T.; Tremaroli, V.; Ji, B.; Prifti, E.; Myridakis, A.; et al. Microbiome and metabolome features of the cardiometabolic disease spectrum. Nat. Med. 2022, 28, 303–314. [Google Scholar] [CrossRef]
  7. Suez, J.; Korem, T.; Zeevi, D.; Zilberman-Schapira, G.; Thaiss, C.A.; Maza, O.; Israeli, D.; Zmora, N.; Gilad, S.; Weinberger, A.; et al. Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature 2014, 514, 181–186. [Google Scholar] [CrossRef]
  8. Vrieze, A.; Van Nood, E.; Holleman, F.; Salojärvi, J.; Kootte, R.S.; Bartelsman, J.F.W.M.; Dallinga-Thie, G.M.; Ackermans, M.T.; Serlie, M.J.; Oozeer, R.; et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 2012, 143, 913–916.e7. [Google Scholar] [CrossRef]
  9. Gandhi, N.N.; Cobra, P.F.; Steele, J.L.; Markley, J.L.; Rankin, S.A. Lactobacillus demonstrate thiol-independent metabolism of methylglyoxal: Implications toward browning prevention in Parmesan cheese. J. Dairy Sci. 2018, 101, 968–978. [Google Scholar] [CrossRef]
  10. Huang, K.; Rudolph, F.B.; Bennett, G.N. Characterization of methylglyoxal synthase from Clostridium acetobutylicum ATCC 824 and its use in the formation of 1,2-propanediol. Appl. Environ. Microbiol. 1999, 65, 3244–3247. [Google Scholar] [CrossRef]
  11. Saxena, R.K.; Anand, P.; Saran, S.; Isar, J.; Agarwal, L. Microbial production and applications of 1,2-propanediol. Indian J. Microbiol. 2010, 50, 2–11. [Google Scholar] [CrossRef]
  12. Sun, S.; Shu, L.; Lu, X.; Wang, Q.; Tišma, M.; Zhu, C.; Shi, J.; Baganz, F.; Lye, G.J.; Hao, J. 1,2-Propanediol production from glycerol via an endogenous pathway of Klebsiella pneumoniae. Appl. Microbiol. Biotechnol. 2021, 105, 9003–9016. [Google Scholar] [CrossRef]
  13. Bennett, G.N.; San, K.Y. Microbial formation, biotechnological production and applications of 1,2-propanediol. Appl. Microbiol. Biotechnol. 2001, 55, 1–9. [Google Scholar] [CrossRef] [PubMed]
  14. Baskaran, S.; Prasanna Rajan, D.; Balasubramanian, K.A. Formation of methylglyoxal by bacteria isolated from human faeces. J. Med. Microbiol. 1989, 28, 211–215. [Google Scholar] [CrossRef] [PubMed]
  15. Degen, J.; Hellwig, M.; Henle, T. 1,2-dicarbonyl compounds in commonly consumed foods. J. Agric. Food Chem. 2012, 60, 7071–7079. [Google Scholar] [CrossRef] [PubMed]
  16. Vašková, J.; Kováčová, G.; Pudelský, J.; Palenčár, D.; Mičková, H. Methylglyoxal Formation—Metabolic Routes and Consequences. Antioxidants 2025, 14, 212. [Google Scholar] [CrossRef]
  17. Thornalley, P.J.; Langborg, A.; Minhas, H.S. Formation of glyoxal, methylglyoxal and 3-deoxyglucosone in the glycation of proteins by glucose. Biochem. J. 1999, 344 Pt 1, 109–116. [Google Scholar] [CrossRef]
  18. Rabbani, N.; Thornalley, P.J. Dicarbonyl proteome and genome damage in metabolic and vascular disease. Biochem. Soc. Trans. 2014, 42, 425–432. [Google Scholar] [CrossRef]
  19. Thornalley, P.J. Modification of the glyoxalase system in human red blood cells by glucose in vitro. Biochem. J. 1988, 254, 751–755. [Google Scholar] [CrossRef]
  20. Borysiuk, K.; Ostaszewska-Bugajska, M.; Vaultier, M.N.; Hasenfratz-Sauder, M.P.; Szal, B. Enhanced Formation of Methylglyoxal-Derived Advanced Glycation End Products in Arabidopsis Under Ammonium Nutrition. Front. Plant Sci. 2018, 9, 667. [Google Scholar] [CrossRef]
  21. Allaman, I.; Bélanger, M.; Magistretti, P.J. Methylglyoxal, the dark side of glycolysis. Front Neurosci. 2015, 9, 23. [Google Scholar] [CrossRef] [PubMed]
  22. Kalapos, M.P. Methylglyoxal and glucose metabolism: A historical perspective and future avenues for research. Drug Metabol. Drug Interact 2008, 23, 69–91. [Google Scholar] [CrossRef] [PubMed]
  23. Lai, S.W.T.; Lopez Gonzalez, E.D.J.; Zoukari, T.; Ki, P.; Shuck, S.C. Methylglyoxal and Its Adducts: Induction, Repair, and Association with Disease. Chem. Res. Toxicol. 2022, 35, 1720–1746. [Google Scholar] [CrossRef]
  24. Rabbani, N.; Thornalley, P.J. Measurement of methylglyoxal by stable isotopic dilution analysis LC-MS/MS with corroborative prediction in physiological samples. Nat. Protoc. 2014, 9, 1969–1979. [Google Scholar] [CrossRef]
  25. Morgenstern, J.; Campos Campos, M.; Nawroth, P.; Fleming, T. The Glyoxalase System—New Insights into an Ancient Metabolism. Antioxidants 2020, 9, 939. [Google Scholar] [CrossRef] [PubMed]
  26. Synold, T.; Xi, B.; Wuenschell, G.E.; Tamae, D.; Figarola, J.L.; Rahbar, S.; Termini, J. Advanced Glycation End Products of DNA: Quantification of N2-(1-carboxyethyl)-2’-deoxyguanosine (CEdG) in Biological Samples by LC-ESI-MS/MS. Chem. Res. Toxicol. 2008, 21, 2148–2155. [Google Scholar] [CrossRef]
  27. Galligan, J.J.; Wepy, J.A.; Streeter, M.D.; Kingsley, P.J.; Mitchener, M.M.; Wauchope, O.R.; Beavers, W.N.; Rose, K.L.; Wang, T.; Spiegel, D.A.; et al. Methylglyoxal-derived posttranslational arginine modifications are abundant histone marks. Proc. Natl. Acad. Sci. USA 2018, 115, 9228–9233. [Google Scholar] [CrossRef]
  28. El-Osta, A.; Brasacchio, D.; Yao, D.; Pocai, A.; Jones, P.L.; Roeder, R.G.; Cooper, M.E.; Brownlee, M. Transient high glucose causes persistent epigenetic changes and altered gene expression during subsequent normoglycemia. J. Exp. Med. 2008, 205, 2409–2417. [Google Scholar] [CrossRef]
  29. Chan, C.; Huang, D.; Huang, Y.; Hsu, S.; Kang, L.; Shen, C.; Lin, W. Methylglyoxal induces cell death through endoplasmic reticulum stress-associated ROS production and mitochondrial dysfunction. J. Cell Mol. Med. 2016, 20, 1749–1760. [Google Scholar] [CrossRef]
  30. Brouwers, O.; Niessen, P.M.G.; Miyata, T.; Østergaard, J.A.; Flyvbjerg, A.; Peutz-Kootstra, C.J.; Sieber, J.; Mundel, P.H.; Brownlee, M.; Janssen, B.J.A.; et al. Glyoxalase-1 overexpression reduces endothelial dysfunction and attenuates early renal impairment in a rat model of diabetes. Diabetologia 2014, 57, 224–235. [Google Scholar] [CrossRef]
  31. Queisser, M.A.; Yao, D.; Geisler, S.; Hammes, H.P.; Lochnit, G.; Schleicher, E.D.; Brownlee, M.; Preissner, K.T. Hyperglycemia impairs proteasome function by methylglyoxal. Diabetes 2010, 59, 670–678. [Google Scholar] [CrossRef] [PubMed]
  32. Shinohara, M.; Thornalley, P.J.; Giardino, I.; Beisswenger, P.; Thorpe, S.R.; Onorato, J.; Brownlee, M. Overexpression of glyoxalase-I in bovine endothelial cells inhibits intracellular advanced glycation endproduct formation and prevents hyperglycemia-induced increases in macromolecular endocytosis. J. Clin. Investig. 1998, 101, 1142–1147. [Google Scholar] [CrossRef]
  33. Nokin, M.J.; Durieux, F.; Bellier, J.; Peulen, O.; Uchida, K.; Spiegel, D.A.; Cochrane, J.R.; Hutton, C.A.; Castronovo, V.; Bellahcène, A. Hormetic potential of methylglyoxal, a side-product of glycolysis, in switching tumours from growth to death. Sci. Rep. 2017, 7, 11722. [Google Scholar] [CrossRef]
  34. Bollong, M.J.; Lee, G.; Coukos, J.S.; Yun, H.; Zambaldo, C.; Chang, J.W.; Chin, E.N.; Ahmad, I.; Chatterjee, A.K.; Lairson, L.L.; et al. A metabolite-derived protein modification integrates glycolysis with KEAP1-NRF2 signaling. Nature 2018, 562, 600–604. [Google Scholar] [CrossRef] [PubMed]
  35. Hanssen, N.M.J.; Wouters, K.; Huijberts, M.S.; Gijbels, M.J.; Sluimer, J.C.; Scheijen, J.L.J.M.; Heeneman, S.; Biessen, E.A.L.; Daemen, M.J.A.P.; Brownlee, M.; et al. Higher levels of advanced glycation endproducts in human carotid atherosclerotic plaques are associated with a rupture-prone phenotype. Eur. Heart J. 2014, 35, 1137–1146. [Google Scholar] [CrossRef]
  36. Van Herreweghe, F.; Mao, J.; Chaplen, F.W.R.; Grooten, J.; Gevaert, K.; Vandekerckhove, J.; Vancompernolle, K. Tumor necrosis factor-induced modulation of glyoxalase I activities through phosphorylation by PKA results in cell death and is accompanied by the formation of a specific methylglyoxal-derived AGE. Proc. Natl. Acad. Sci. USA 2002, 99, 949–954. [Google Scholar] [CrossRef]
  37. Zhang, X.; Schalkwijk, C.G.; Wouters, K. Immunometabolism and the modulation of immune responses and host defense: A role for methylglyoxal? Biochim. Biophys. Acta BBA Mol. Basis Dis. 2022, 1868, 166425. [Google Scholar] [CrossRef] [PubMed]
  38. Rachman, H.; Kim, N.; Ulrichs, T.; Baumann, S.; Pradl, L.; Nasser Eddine, A.; Bild, M.; Rother, M.; Kuban, R.J.; Lee, J.S.; et al. Critical role of methylglyoxal and AGE in mycobacteria-induced macrophage apoptosis and activation. PLoS ONE 2006, 1, e29. [Google Scholar] [CrossRef]
  39. Brenner, T.; Fleming, T.; Uhle, F.; Silaff, S.; Schmitt, F.; Salgado, E.; Ulrich, A.; Zimmermann, S.; Bruckner, T.; Martin, E.; et al. Methylglyoxal as a new biomarker in patients with septic shock: An observational clinical study. Crit. Care 2014, 18, 683. [Google Scholar] [CrossRef]
  40. Schmoch, T.; Uhle, F.; Siegler, B.H.; Fleming, T.; Morgenstern, J.; Nawroth, P.P.; Weigand, M.A.; Brenner, T. The Glyoxalase System and Methylglyoxal-Derived Carbonyl Stress in Sepsis: Glycotoxic Aspects of Sepsis Pathophysiology. Int. J. Mol. Sci. 2017, 18, 657. [Google Scholar] [CrossRef]
  41. Zhang, M.M.; Ong, C.L.Y.; Walker, M.J.; McEwan, A.G. Defence against methylglyoxal in Group A Streptococcus: A role for Glyoxylase I in bacterial virulence and survival in neutrophils? Pathog. Dis. 2016, 74, ftv122. [Google Scholar] [CrossRef]
  42. Chakraborty, S.; Chaudhuri, D.; Balakrishnan, A.; Chakravortty, D. Salmonella methylglyoxal detoxification by STM3117-encoded lactoylglutathione lyase affects virulence in coordination with Salmonella pathogenicity island 2 and phagosomal acidification. Microbiol. Read. Engl. 2014, 160 Pt 9, 1999–2017. [Google Scholar] [CrossRef] [PubMed]
  43. Anaya-Sanchez, A.; Feng, Y.; Berude, J.C.; Portnoy, D.A. Detoxification of methylglyoxal by the glyoxalase system is required for glutathione availability and virulence activation in Listeria monocytogenes. PLoS Pathog. 2021, 17, e1009819. [Google Scholar] [CrossRef] [PubMed]
  44. Moraru, A.; Wiederstein, J.; Pfaff, D.; Fleming, T.; Miller, A.K.; Nawroth, P.; Teleman, A.A. Elevated Levels of the Reactive Metabolite Methylglyoxal Recapitulate Progression of Type 2 Diabetes. Cell Metab. 2018, 27, 926–934.e8. [Google Scholar] [CrossRef] [PubMed]
  45. Lodd, E.; Wiggenhauser, L.M.; Morgenstern, J.; Fleming, T.H.; Poschet, G.; Büttner, M.; Tabler, C.T.; Wohlfart, D.P.; Nawroth, P.P.; Kroll, J. The combination of loss of glyoxalase1 and obesity results in hyperglycemia. JCI Insight 2019, 4, e126154. [Google Scholar] [CrossRef]
  46. Morgenstern, J.; Fleming, T.; Schumacher, D.; Eckstein, V.; Freichel, M.; Herzig, S.; Nawroth, P. Loss of Glyoxalase 1 Induces Compensatory Mechanism to Achieve Dicarbonyl Detoxification in Mammalian Schwann Cells. J. Biol. Chem. 2017, 292, 3224–3238. [Google Scholar] [CrossRef]
  47. Dobariya, P.; Xie, W.; Rao, S.P.; Xie, J.; Seelig, D.M.; Vince, R.; Lee, M.K.; More, S.S. Deletion of Glyoxalase 1 Exacerbates Acetaminophen-Induced Hepatotoxicity in Mice. Antioxidants 2024, 13, 648. [Google Scholar] [CrossRef]
  48. Giacco, F.; Du, X.; D’Agati, V.D.; Milne, R.; Sui, G.; Geoffrion, M.; Brownlee, M. Knockdown of Glyoxalase 1 Mimics Diabetic Nephropathy in Nondiabetic Mice. Diabetes 2014, 63, 291–299. [Google Scholar] [CrossRef]
  49. Geoffrion, M.; Du, X.; Irshad, Z.; Vanderhyden, B.C.; Courville, K.; Sui, G.; D’Agati, V.D.; Ott-Braschi, S.; Rabbani, N.; Thornalley, P.J.; et al. Differential effects of glyoxalase 1 overexpression on diabetic atherosclerosis and renal dysfunction in streptozotocin-treated, apolipoprotein E-deficient mice. Physiol. Rep. 2014, 2, e12043. [Google Scholar] [CrossRef]
  50. Spadoni, I.; Zagato, E.; Bertocchi, A.; Paolinelli, R.; Hot, E.; Di Sabatino, A.; Caprioli, F.; Bottiglieri, L.; Oldani, A.; Viale, G.; et al. A gut-vascular barrier controls the systemic dissemination of bacteria. Science 2015, 350, 830–834. [Google Scholar] [CrossRef]
  51. Min, S.; Than, N.; Shin, Y.C.; Hu, G.; Shin, W.; Ambrosini, Y.M.; Kim, H.J. Live probiotic bacteria administered in a pathomimetic Leaky Gut Chip ameliorate impaired epithelial barrier and mucosal inflammation. Sci. Rep. 2022, 12, 22641. [Google Scholar] [CrossRef] [PubMed]
  52. Li, J.; Zhang, H.; Liu, W.; Yang, X.; Zhu, L.; Wu, G.; Zhang, H. Methylglyoxal scavenging capacity of fiber-bound polyphenols from highland barley during colonic fermentation and its modulation on methylglyoxal-interfered gut microbiota. Food Chem. 2024, 434, 137409. [Google Scholar] [CrossRef] [PubMed]
  53. Brighina, S.; Poveda Turrado, C.; Restuccia, C.; Walton, G.; Fallico, B.; Oruna-Concha, M.J.; Arena, E. Detrimental effect on the gut microbiota of 1,2-dicarbonyl compounds after in vitro gastro-intestinal and fermentative digestion. Food Chem. 2021, 341 Pt 1, 128237. [Google Scholar] [CrossRef]
  54. Cooper, R.A. Metabolism of methylglyoxal in microorganisms. Annu Rev. Microbiol. 1984, 38, 49–68. [Google Scholar] [CrossRef]
  55. Willetts, A.J.; Turner, J.M. Threonine metabolism in a strain of Bacillus subtilis: Enzymes acting on methylglyoxal. Biochim. Biophys. Acta BBA -Gen. Subj. 1970, 222, 668–670. [Google Scholar] [CrossRef]
  56. Elliott, W.H. Methylglyoxal Formation from Aminoacetone by Ox Plasma. Nature 1960, 185, 467–468. [Google Scholar] [CrossRef]
  57. Green, M.L.; Lewis, J.B. The oxidation of aminoacetone by a species of Arthrobacter. Biochem. J. 1968, 106, 267–270. [Google Scholar] [CrossRef] [PubMed]
  58. McLeod, A.; Snipen, L.; Naterstad, K.; Axelsson, L. Global transcriptome response in Lactobacillus sakei during growth on ribose. BMC Microbiol. 2011, 11, 145. [Google Scholar] [CrossRef]
  59. Galimberti, S.; Rocchetti, G.; Di Rico, F.; Rossetti, C.; Fontana, A.; Lucini, L.; Callegari, M.L. Untargeted metabolomics provide new insights into the implication of Lactobacillus helveticus strains isolated from natural whey starter in methylglyoxal-mediated browning. Food Res. Int. 2023, 174, 113644. [Google Scholar] [CrossRef]
  60. Zhao, Q.; Su, Y.; Wang, Z.; Chen, C.; Wu, T.; Huang, Y. Identification of glutathione (GSH)-independent glyoxalase III from Schizosaccharomyces pombe. BMC Evol. Biol. 2014, 14, 86. [Google Scholar] [CrossRef]
  61. Scheckhuber, C.Q. Penicillium chrysogenum as a model system for studying cellular effects of methylglyoxal. BMC Microbiol. 2015, 15, 138. [Google Scholar] [CrossRef] [PubMed]
  62. Martins, A.M.; Cordeiro, C.A.; Ponces Freire, A.M. In situ analysis of methylglyoxal metabolism in Saccharomyces cerevisiae. FEBS Lett. 2001, 499, 41–44. [Google Scholar] [CrossRef]
  63. Ozyamak, E.; Black, S.S.; Walker, C.A.; Maclean, M.J.; Bartlett, W.; Miller, S.; Booth, I.R. The critical role of S-lactoylglutathione formation during methylglyoxal detoxification in Escherichia coli. Mol. Microbiol. 2010, 78, 1577–1590. [Google Scholar] [CrossRef] [PubMed]
  64. Darling, T.N.; Blum, J.J. D-lactate production by Leishmania braziliensis through the glyoxalase pathway. Mol. Biochem. Parasitol. 1988, 28, 121–127. [Google Scholar] [CrossRef]
  65. Tirelli, E.; Pucci, M.; Squillario, M.; Bignotti, G.; Messali, S.; Zini, S.; Bugatti, M.; Cadei, M.; Memo, M.; Caruso, A.; et al. Effects of methylglyoxal on intestine and microbiome composition in aged mice. Food Chem. Toxicol. 2025, 197, 115276. [Google Scholar] [CrossRef]
  66. Zhou, H.; Wang, L.; Lin, Z.; Jiang, C.; Chen, X.; Wang, K.; Liu, L.; Shao, L.; Pan, J.; Li, J.; et al. Methylglyoxal from gut microbes boosts radiosensitivity and radioimmunotherapy in rectal cancer by triggering endoplasmic reticulum stress and cGAS-STING activation. J. Immunother. Cancer 2023, 11, e007840. [Google Scholar] [CrossRef]
  67. Maiden, M.F.J.; Pham, C.; Kashket, S. Glucose toxicity effect and accumulation of methylglyoxal by the periodontal anaerobe Bacteroides forsythus. Anaerobe 2004, 10, 27–32. [Google Scholar] [CrossRef]
  68. Kashket, S.; Maiden, M.F.J.; Haffajee, A.D.; Kashket, E.R. Accumulation of methylglyoxal in the gingival crevicular fluid of chronic periodontitis patients. J. Clin. Periodontol. 2003, 30, 364–367. [Google Scholar] [CrossRef] [PubMed]
  69. Ferguson, G.P.; Tötemeyer, S.; MacLean, M.J.; Booth, I.R. Methylglyoxal production in bacteria: Suicide or survival? Arch. Microbiol. 1998, 170, 209–218. [Google Scholar] [CrossRef]
  70. Chakraborty, S.; Karmakar, K.; Chakravortty, D. Cells producing their own nemesis: Understanding methylglyoxal metabolism. IUBMB Life 2014, 66, 667–678. [Google Scholar] [CrossRef]
  71. Landmann, J.J.; Busse, R.A.; Latz, J.H.; Singh, K.D.; Stülke, J.; Görke, B. Crh, the paralogue of the phosphocarrier protein HPr, controls the methylglyoxal bypass of glycolysis in Bacillus subtilis. Mol. Microbiol. 2011, 82, 770–787. [Google Scholar] [CrossRef]
  72. Culp, E.J.; Goodman, A.L. Cross-feeding in the gut microbiome: Ecology and mechanisms. Cell Host Microbe 2023, 31, 485–499. [Google Scholar] [CrossRef]
  73. Weber, J.; Kayser, A.; Rinas, U. Metabolic flux analysis of Escherichia coli in glucose-limited continuous culture. II. Dynamic response to famine and feast, activation of the methylglyoxal pathway and oscillatory behaviour. Microbiology 2005, 151, 707–716. [Google Scholar] [CrossRef]
  74. Tötemeyer, S.; Booth, N.A.; Nichols, W.W.; Dunbar, B.; Booth, I.R. From famine to feast: The role of methylglyoxal production in Escherichia coli. Mol. Microbiol. 1998, 27, 553–562. [Google Scholar] [CrossRef] [PubMed]
  75. Misra, K.; Banerjee, A.B.; Ray, S.; Ray, M. Glyoxalase III from Escherichia coli: A single novel enzyme for the conversion of methylglyoxal into D-lactate without reduced glutathione. Biochem. J. 1995, 305 Pt 3, 999–1003. [Google Scholar] [CrossRef]
  76. Lee, C.; Park, C. Bacterial Responses to Glyoxal and Methylglyoxal: Reactive Electrophilic Species. Int. J. Mol. Sci. 2017, 18, 169. [Google Scholar] [CrossRef] [PubMed]
  77. Subedi, K.P.; Choi, D.; Kim, I.; Min, B.; Park, C. Hsp31 of Escherichia coli K-12 is glyoxalase III. Mol. Microbiol. 2011, 81, 926–936. [Google Scholar] [CrossRef]
  78. Hasim, S.; Hussin, N.A.; Alomar, F.; Bidasee, K.R.; Nickerson, K.W.; Wilson, M.A. A Glutathione-independent Glyoxalase of the DJ-1 Superfamily Plays an Important Role in Managing Metabolically Generated Methylglyoxal in Candida albicans. J. Biol. Chem. 2014, 289, 1662–1674. [Google Scholar] [CrossRef] [PubMed]
  79. Farrera, D.O.; Galligan, J.J. The human glyoxalase gene family in health and disease. Chem. Res. Toxicol. 2022, 35, 1766–1776. [Google Scholar] [CrossRef]
  80. Misra, K.; Banerjee, A.B.; Ray, S.; Ray, M. Reduction of methylglyoxal in Escherichia coli K12 by an aldehyde reductase and alcohol dehydrogenase. Mol. Cell Biochem. 1996, 156, 117–124. [Google Scholar] [CrossRef]
  81. Sánchez-Riera, F.; Cameron, D.C.; Cooney, C.L. Influence of environmental factors in the production of R(−)-1,2-propanediol by clostridium thermosaccharolyticum. Biotechnol. Lett. 1987, 9, 449–454. [Google Scholar] [CrossRef]
  82. Tao, Y.M.; Bu, C.Y.; Zou, L.H.; Hu, Y.L.; Zheng, Z.J.; Ouyang, J. A comprehensive review on microbial production of 1,2-propanediol: Micro-organisms, metabolic pathways, and metabolic engineering. Biotechnol. Biofuels 2021, 14, 216. [Google Scholar] [CrossRef] [PubMed]
  83. Suzuki, T.; and Onishi, H. Aerobic Dissimilation of l-Rhamnose and the Production of l-Rhamnonic Acid and 1,2-Propanediol by Yeasts. Agric. Biol. Chem. 1968, 32, 888–893. [Google Scholar]
  84. Cameron, D.C.; Cooney, C.L. A Novel Fermentation: The Production of R(–)–1,2–Propanediol and Acetol by Clostridium thermosaccharolyticum. Bio/Technology 1986, 4, 651–654. [Google Scholar] [CrossRef]
  85. Tran-Din, K.; Gottschalk, G. Formation of d(-)-1,2-propanediol and d(-)-lactate from glucose by Clostridium sphenoides under phosphate limitation. Arch. Microbiol. 1985, 142, 87–92. [Google Scholar] [CrossRef]
  86. Douglas, R.M.; Roberts, J.A.; Munro, A.W.; Ritchie, G.Y.; Lamb, A.J.; Booth, I.R. The distribution of homologues of the Escherichia coli KefC K+-efflux system in other bactèrial species. Microbiology 1991, 137, 1999–2005. [Google Scholar] [CrossRef]
  87. Elmore, M.J.; Lamb, A.J.; Ritchie, G.Y.; Douglas, R.M.; Munro, A.; Gajewska, A.; Booth, I.R. Activation of potassium efflux from Escherichia coli by glutathione metabolites. Mol. Microbiol. 1990, 4, 405–412. [Google Scholar] [CrossRef]
  88. Ferguson, G.P.; Munro, A.W.; Douglas, R.M.; McLaggan, D.; Booth, I.R. Activation of potassium channels during metabolite detoxification in Escherichia coli. Mol. Microbiol. 1993, 9, 1297–1303. [Google Scholar] [CrossRef]
  89. Ferguson, G.P.; Nikolaev, Y.; McLaggan, D.; Maclean, M.; Booth, I.R. Survival during exposure to the electrophilic reagent N-ethylmaleimide in Escherichia coli: Role of KefB and KefC potassium channels. J. Bacteriol. 1997, 179, 1007–1012. [Google Scholar] [CrossRef]
  90. Ferguson, G.P.; McLaggan, D.; Booth, I.R. Potassium channel activation by glutathione-S-conjugates in Escherichia coli: Protection against methylglyoxal is mediated by cytoplasmic acidification. Mol. Microbiol. 1995, 17, 1025–1033. [Google Scholar] [CrossRef]
  91. Cox, T.O.; Lundgren, P.; Nath, K.; Thaiss, C.A. Metabolic control by the microbiome. Genome Med. 2022, 14, 80. [Google Scholar] [CrossRef] [PubMed]
  92. Mei, Z.; Wang, F.; Bhosle, A.; Dong, D.; Mehta, R.; Ghazi, A.; Zhang, Y.; Liu, Y.; Rinott, E.; Ma, S.; et al. Strain-specific gut microbial signatures in type 2 diabetes identified in a cross-cohort analysis of 8117 metagenomes. Nat. Med. 2024, 30, 2265–2276. [Google Scholar] [CrossRef]
  93. Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444, 1027–1031. [Google Scholar] [CrossRef] [PubMed]
  94. Hadadi, N.; Berweiler, V.; Wang, H.; Trajkovski, M. Intestinal microbiota as a route for micronutrient bioavailability. Curr. Opin. Endocr. Metab. Res. 2021, 20, 100285. [Google Scholar] [CrossRef]
  95. Fetissov, S.O. Role of the gut microbiota in host appetite control: Bacterial growth to animal feeding behaviour. Nat. Rev. Endocrinol. 2017, 13, 11–25. [Google Scholar] [CrossRef] [PubMed]
  96. Chen, F.; Hou, K.; Chen, Z.S. Gut microbes regulate the feeding center: A new discovery of Gut Brain Axis. Signal Transduct. Target. Ther. 2022, 7, 1–2. [Google Scholar] [CrossRef]
  97. Blok, L.; Hanssen, N.; Nieuwdorp, M.; Rampanelli, E. From Microbes to Metabolites: Advances in Gut Microbiome Research in Type 1 Diabetes. Metabolites 2025, 15, 138. [Google Scholar] [CrossRef]
  98. Nemet, I.; Li, X.S.; Haghikia, A.; Li, L.; Wilcox, J.; Romano, K.A.; Buffa, J.A.; Witkowski, M.; Demuth, I.; König, M.; et al. Atlas of gut microbe-derived products from aromatic amino acids and risk of cardiovascular morbidity and mortality. Eur. Heart J. 2023, 44, 3085–3096. [Google Scholar] [CrossRef]
  99. Dekkers, K.F.; Sayols-Baixeras, S.; Baldanzi, G.; Nowak, C.; Hammar, U.; Nguyen, D.; Varotsis, G.; Brunkwall, L.; Nielsen, N.; Eklund, A.C.; et al. An online atlas of human plasma metabolite signatures of gut microbiome composition. Nat. Commun. 2022, 13, 5370. [Google Scholar] [CrossRef]
  100. Pérez-Reytor, D.; Puebla, C.; Karahanian, E.; García, K. Use of Short-Chain Fatty Acids for the Recovery of the Intestinal Epithelial Barrier Affected by Bacterial Toxins. Front Physiol. 2021, 12, 650313. [Google Scholar] [CrossRef]
  101. Puddu, A.; Sanguineti, R.; Montecucco, F.; Viviani, G.L. Evidence for the gut microbiota short-chain fatty acids as key pathophysiological molecules improving diabetes. Mediat. Inflamm. 2014, 2014, 162021. [Google Scholar] [CrossRef] [PubMed]
  102. Mandaliya, D.K.; Seshadri, S. Short Chain Fatty Acids, pancreatic dysfunction and type 2 diabetes. Pancreatology 2019, 19, 280–284. [Google Scholar]
  103. Hernández, M.A.G.; Canfora, E.E.; Jocken, J.W.E.; Blaak, E.E. The Short-Chain Fatty Acid Acetate in Body Weight Control and Insulin Sensitivity. Nutrients 2019, 11, 1943. [Google Scholar] [CrossRef] [PubMed]
  104. Canfora, E.E.; Jocken, J.W.; Blaak, E.E. Short-chain fatty acids in control of body weight and insulin sensitivity. Nat. Rev. Endocrinol. 2015, 11, 577–591. [Google Scholar] [CrossRef]
  105. Shah, A.B.; Baiseitova, A.; Zahoor, M.; Ahmad, I.; Ikram, M.; Bakhsh, A.; Shah, M.A.; Ali, I.; Idress, M.; Ullah, R.; et al. Probiotic significance of Lactobacillus strains: A comprehensive review on health impacts, research gaps, and future prospects. Gut Microbes 2024, 16, 2431643. [Google Scholar] [CrossRef] [PubMed]
  106. Saeedi, B.J.; Liu, K.H.; Owens, J.A.; Hunter-Chang, S.; Camacho, M.C.; Eboka, R.U.; Chandrasekharan, B.; Baker, N.F.; Darby, T.M.; Robinson, B.S.; et al. Gut-Resident Lactobacilli Activate Hepatic Nrf2 and Protect Against Oxidative Liver Injury. Cell Metab. 2020, 31, 956–968.e5. [Google Scholar] [CrossRef]
  107. Nishimoto, S.; Koike, S.; Inoue, N.; Suzuki, T.; Ogasawara, Y. Activation of Nrf2 attenuates carbonyl stress induced by methylglyoxal in human neuroblastoma cells: Increase in GSH levels is a critical event for the detoxification mechanism. Biochem. Biophys. Res. Commun. 2017, 483, 874–879. [Google Scholar] [CrossRef]
  108. Shin, M.G.; Lee, J.W.; Han, J.S.; Lee, B.; Jeong, J.H.; Park, S.H.; Kim, J.H.; Jang, S.; Park, M.; Kim, S.Y.; et al. Bacteria-derived metabolite, methylglyoxal, modulates the longevity of C. elegans through TORC2/SGK-1/DAF-16 signaling. Proc. Natl. Acad. Sci. USA 2020, 117, 17142–17150. [Google Scholar] [CrossRef]
  109. Hellwig, M.; Gensberger-Reigl, S.; Henle, T.; Pischetsrieder, M. Food-derived 1,2-dicarbonyl compounds and their role in diseases. Semin. Cancer Biol. 2018, 49, 1–8. [Google Scholar] [CrossRef]
  110. Maasen, K.; Scheijen, J.L.J.M.; Opperhuizen, A.; Stehouwer, C.D.A.; Van Greevenbroek, M.M.; Schalkwijk, C.G. Quantification of dicarbonyl compounds in commonly consumed foods and drinks; presentation of a food composition database for dicarbonyls. Food Chem. 2021, 339, 128063. [Google Scholar] [CrossRef]
  111. Amoroso, A.; Maga, G.; Daglia, M. Cytotoxicity of α-dicarbonyl compounds submitted to in vitro simulated digestion process. Food Chem. 2013, 140, 654–659. [Google Scholar] [CrossRef]
  112. Treibmann, S.; Venema, K.; Henle, T. Glycation reactions of methylglyoxal during digestion in a dynamic, in vitro model of the upper gastrointestinal tract (TIM-1). Food Sci. Nutr. 2024, 12, 4702–4712. [Google Scholar] [CrossRef]
  113. Hamzalıoğlu, A.; Gökmen, V. Investigations on the reactions of α-dicarbonyl compounds with amino acids and proteins during in vitro digestion of biscuits. Food Funct. 2016, 7, 2544–2550. [Google Scholar] [CrossRef]
  114. Degen, J.; Vogel, M.; Richter, D.; Hellwig, M.; Henle, T. Metabolic transit of dietary methylglyoxal. J. Agric. Food Chem. 2013, 61, 10253–10260. [Google Scholar] [CrossRef]
  115. Maasen, K.; Eussen, S.J.P.M.; Dagnelie, P.C.; Houben, A.J.H.M.; Webers, C.A.B.; Schram, M.T.; Berendschot, T.T.J.M.; Stehouwer, C.D.A.; Opperhuizen, A.; van Greevenbroek, M.M.J.; et al. Habitual intake of dietary methylglyoxal is associated with less low-grade inflammation: The Maastricht Study. Am. J. Clin. Nutr. 2022, 116, 1715–1728. [Google Scholar] [CrossRef] [PubMed]
  116. Kaźmierczak-Barańska, J.; Karwowski, B.T. The Antioxidant Potential of Commercial Manuka Honey from New Zealand—Biochemical and Cellular Studies. Curr. Issues Mol. Biol. 2024, 46, 6366–6376. [Google Scholar] [CrossRef] [PubMed]
  117. Niaz, K.; Maqbool, F.; Bahadar, H.; Abdollahi, M. Health Benefits of Manuka Honey as an Essential Constituent for Tissue Regeneration. Curr. Drug Metab. 2017, 18, 881–892. [Google Scholar] [CrossRef] [PubMed]
  118. Mavric, E.; Wittmann, S.; Barth, G.; Henle, T. Identification and quantification of methylglyoxal as the dominant antibacterial constituent of Manuka (Leptospermum scoparium) honeys from New Zealand. Mol. Nutr. Food Res. 2008, 52, 483–489. [Google Scholar] [CrossRef]
  119. Kilty, S.J.; Duval, M.; Chan, F.T.; Ferris, W.; Slinger, R. Methylglyoxal: (active agent of manuka honey) in vitro activity against bacterial biofilms. Int. Forum Allergy Rhinol. 2011, 1, 348–350. [Google Scholar] [CrossRef]
  120. Lu, J.; Carter, D.A.; Turnbull, L.; Rosendale, D.; Hedderley, D.; Stephens, J.; Gannabathula, S.; Steinhorn, G.; Schlothauer, R.C.; Whitchurch, C.B.; et al. The Effect of New Zealand Kanuka, Manuka and Clover Honeys on Bacterial Growth Dynamics and Cellular Morphology Varies According to the Species. PLoS ONE 2013, 8, e55898. [Google Scholar] [CrossRef]
  121. Maddocks, S.E.; Lopez, M.S.; Rowlands, R.S.; Cooper, R.A. Manuka honey inhibits the development of Streptococcus pyogenes biofilms and causes reduced expression of two fibronectin binding proteins. Microbiology 2012, 158, 781–790. [Google Scholar] [CrossRef]
  122. Lu, J.; Turnbull, L.; Burke, C.M.; Liu, M.; Carter, D.A.; Schlothauer, R.C.; Whitchurch, C.B.; Harry, E.J. Manuka-type honeys can eradicate biofilms produced by Staphylococcus aureus strains with different biofilm-forming abilities. PeerJ 2014, 2, e326. [Google Scholar] [CrossRef] [PubMed]
  123. Tang, J.S.; Compton, B.J.; Marshall, A.; Anderson, R.; Li, Y.; van der Woude, H.; Hermans, I.F.; Painter, G.F.; Gasser, O. Mānuka honey-derived methylglyoxal enhances microbial sensing by mucosal-associated invariant T cells. Food Funct. 2020, 11, 5782–5787. [Google Scholar] [CrossRef]
  124. Rabie, E.; Serem, J.C.; Oberholzer, H.M.; Gaspar, A.R.M.; Bester, M.J. How methylglyoxal kills bacteria: An ultrastructural study. Ultrastruct. Pathol. 2016, 40, 107–111. [Google Scholar] [CrossRef] [PubMed]
  125. Brighina, S.; Restuccia, C.; Arena, E.; Palmeri, R.; Fallico, B. Antibacterial activity of 1,2-dicarbonyl compounds and the influence of the in vitro assay system. Food Chem. 2020, 311, 125905. [Google Scholar] [CrossRef]
  126. Hayashi, K.; Fukushima, A.; Hayashi-Nishino, M.; Nishino, K. Effect of methylglyoxal on multidrug-resistant Pseudomonas aeruginosa. Front Microbiol. 2014, 5, 180. [Google Scholar] [CrossRef] [PubMed]
  127. Hayes, G.; Wright, N.; Gardner, S.L.; Telzrow, C.L.; Wommack, A.J.; Vigueira, P.A. Manuka honey and methylglyoxal increase the sensitivity of Staphylococcus aureus to linezolid. Lett. Appl. Microbiol. 2018, 66, 491–495. [Google Scholar] [CrossRef]
  128. Alluhaim, W.; Alkhulaifi, M.M.; Alzahrani, R.R.; Alrfaei, B.M.; Yassin, A.E.B.; Alghoribi, M.F.; Alsaadi, A.M.; Al-Asmari, A.I.; Al-Fahad, A.J.; Ali, R.; et al. Effectiveness of a Novel Liposomal Methylglyoxal–Tobramycin Formulation in Reducing Biofilm Formation and Bacterial Adhesion. Antibiotics 2025, 14, 3. [Google Scholar] [CrossRef]
  129. Rosendale, D.; Butts, C.A.; de Guzman, C.E.; Maddox, I.S.; Martell, S.; McIntyre, L.; Skinner, M.A.; Dinnan, H.; Ansell, J. Consumption of antimicrobial manuka honey does not significantly perturb the microbiota in the hind gut of mice. PeerJ 2016, 4, e2787. [Google Scholar] [CrossRef]
  130. Zunkel, K.; Simm, A.; Bartling, B. Long-term intake of the reactive metabolite methylglyoxal is not toxic in mice. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2020, 141, 111333. [Google Scholar] [CrossRef]
  131. Sena, C.M.; Matafome, P.; Crisóstomo, J.; Rodrigues, L.; Fernandes, R.; Pereira, P.; Seiça, R.M. Methylglyoxal promotes oxidative stress and endothelial dysfunction. Pharmacol. Res. 2012, 65, 497–506. [Google Scholar] [CrossRef] [PubMed]
  132. Watanabe, K.; Okada, K.; Fukabori, R.; Hayashi, Y.; Asahi, K.; Terawaki, H.; Kobayashi, K.; Watanabe, T.; Nakayama, M. Methylglyoxal (MG) and Cerebro-Renal Interaction: Does Long-Term Orally Administered MG Cause Cognitive Impairment in Normal Sprague-Dawley Rats? Toxins 2014, 6, 254–269. [Google Scholar] [CrossRef]
  133. Maasen, K.; Eussen, S.J.P.M.; Scheijen, J.L.J.M.; van der Kallen, C.J.H.; Dagnelie, P.C.; Opperhuizen, A.; Stehouwer, C.D.A.; van Greevenbroek, M.M.J.; Schalkwijk, C.G. Higher habitual intake of dietary dicarbonyls is associated with higher corresponding plasma dicarbonyl concentrations and skin autofluorescence: The Maastricht Study. Am. J. Clin. Nutr. 2022, 115, 34–44. [Google Scholar] [CrossRef] [PubMed]
  134. Daglia, M.; Ferrari, D.; Collina, S.; Curti, V. Influence of in vitro simulated gastroduodenal digestion on methylglyoxal concentration of Manuka (Lectospermum scoparium) honey. J. Agric. Food Chem. 2013, 61, 2140–2145. [Google Scholar] [CrossRef]
  135. Hellwig, M.; Geissler, S.; Matthes, R.; Peto, A.; Silow, C.; Brandsch, M.; Henle, T. Transport of free and peptide-bound glycated amino acids: Synthesis, transepithelial flux at Caco-2 cell monolayers, and interaction with apical membrane transport proteins. Chembiochem Eur. J. Chem. Biol. 2011, 12, 1270–1279. [Google Scholar] [CrossRef]
  136. Kellow, N.J.; Coughlan, M.T. Effect of diet-derived advanced glycation end products on inflammation. Nutr. Rev. 2015, 73, 737–759. [Google Scholar] [CrossRef]
  137. Snelson, M.; Coughlan, M.T. Dietary Advanced Glycation End Products: Digestion, Metabolism and Modulation of Gut Microbial Ecology. Nutrients 2019, 11, 215. [Google Scholar] [CrossRef]
  138. Kuntz, S.; Kunz, C.; Rudloff, S. Carbonyl compounds methylglyoxal and glyoxal affect interleukin-8 secretion in intestinal cells by superoxide anion generation and activation of MAPK p38. Mol. Nutr. Food Res. 2010, 54, 1458–1467. [Google Scholar] [CrossRef] [PubMed]
  139. Kuntz, S.; Rudloff, S.; Ehl, J.; Bretzel, R.G.; Kunz, C. Food derived carbonyl compounds affect basal and stimulated secretion of interleukin-6 and -8 in Caco-2 cells. Eur. J. Nutr. 2009, 48, 499–503. [Google Scholar] [CrossRef]
  140. Papetti, A.; Mascherpa, D.; Gazzani, G. Free α-dicarbonyl compounds in coffee, barley coffee and soy sauce and effects of in vitro digestion. Food Chem. 2014, 164, 259–265. [Google Scholar] [CrossRef]
  141. Papetti, A.; Mascherpa, D.; Marrubini, G.; Gazzani, G. Effect of in vitro digestion on free α-dicarbonyl compounds in balsamic vinegars. J. Food Sci. 2013, 78, C514–C519. [Google Scholar] [CrossRef]
  142. Treibmann, S.; Spengler, F.; Degen, J.; Löbner, J.; Henle, T. Studies on the Formation of 3-Deoxyglucosone- and Methylglyoxal-Derived Hydroimidazolones of Creatine during Heat Treatment of Meat. J. Agric. Food Chem. 2019, 67, 5874–5881. [Google Scholar] [CrossRef] [PubMed]
  143. Zhang, H.; Zhang, H.; Troise, A.D.; Fogliano, V. Melanoidins from Coffee, Cocoa, and Bread Are Able to Scavenge α-Dicarbonyl Compounds under Simulated Physiological Conditions. J. Agric. Food Chem. 2019, 67, 10921–10929. [Google Scholar] [CrossRef]
  144. Tikellis, C.; Pickering, R.J.; Tsorotes, D.; Huet, O.; Cooper, M.E.; Jandeleit-Dahm, K.; Thomas, M.C. Dicarbonyl stress in the absence of hyperglycemia increases endothelial inflammation and atherogenesis similar to that observed in diabetes. Diabetes 2014, 63, 3915–3925. [Google Scholar] [CrossRef] [PubMed]
  145. Berlanga, J.; Cibrian, D.; Guillén, I.; Freyre, F.; Alba, J.S.; Lopez-Saura, P.; Merino, N.; Aldama, A.; Quintela, A.M.; Triana, M.E.; et al. Methylglyoxal administration induces diabetes-like microvascular changes and perturbs the healing process of cutaneous wounds. Clin. Sci. Lond. Engl. 2005, 109, 83–95. [Google Scholar] [CrossRef] [PubMed]
  146. Golej, J.; Hoeger, H.; Radner, W.; Unfried, G.; Lubec, G. Oral administration of methylglyoxal leads to kidney collagen accumulation in the mouse. Life Sci. 1998, 63, 801–807. [Google Scholar] [CrossRef]
  147. Schlotterer, A.; Kolibabka, M.; Lin, J.; Acunman, K.; Dietrich, N.; Sticht, C.; Fleming, T.; Nawroth, P.; Hammes, H.P. Methylglyoxal induces retinopathy-type lesions in the absence of hyperglycemia: Studies in a rat model. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2019, 33, 4141–4153. [Google Scholar] [CrossRef]
  148. Guo, Q.; Mori, T.; Jiang, Y.; Hu, C.; Osaki, Y.; Yoneki, Y.; Sun, Y.; Hosoya, T.; Kawamata, A.; Ogawa, S.; et al. Methylglyoxal contributes to the development of insulin resistance and salt sensitivity in Sprague-Dawley rats. J. Hypertens. 2009, 27, 1664–1671. [Google Scholar] [CrossRef]
  149. Nigro, C.; Raciti, G.A.; Leone, A.; Fleming, T.H.; Longo, M.; Prevenzano, I.; Fiory, F.; Mirra, P.; D’Esposito, V.; Ulianich, L.; et al. Methylglyoxal impairs endothelial insulin sensitivity both in vitro and in vivo. Diabetologia 2014, 57, 1485–1494. [Google Scholar] [CrossRef]
  150. Dhar, A.; Dhar, I.; Jiang, B.; Desai, K.M.; Wu, L. Chronic methylglyoxal infusion by minipump causes pancreatic beta-cell dysfunction and induces type 2 diabetes in Sprague-Dawley rats. Diabetes 2011, 60, 899–908. [Google Scholar] [CrossRef]
  151. Zemva, J.; Fink, C.A.; Fleming, T.H.; Schmidt, L.; Loft, A.; Herzig, S.; Knieß, R.A.; Mayer, M.; Bukau, B.; Nawroth, P.P.; et al. Hormesis enables cells to handle accumulating toxic metabolites during increased energy flux. Redox Biol. 2017, 13, 674–686. [Google Scholar] [CrossRef] [PubMed]
  152. Dafre, A.L.; Goldberg, J.; Wang, T.; Spiegel, D.A.; Maher, P. Methylglyoxal, the foe and friend of glyoxalase and Trx/TrxR systems in HT22 nerve cells. Free Radic. Biol. Med. 2015, 89, 8–19. [Google Scholar] [CrossRef]
  153. Maasen, K.; Eussen, S.J.; Dagnelie, P.C.; Stehouwer, C.D.; Opperhuizen, A.; van Greevenbroek, M.M.; Schalkwijk, C.G. Habitual Intake of Dietary Dicarbonyls is Associated with Greater Insulin Sensitivity and Lower Prevalence of Type 2 Diabetes: The Maastricht Study. Am. J. Clin. Nutr. 2023, 118, 151–161. [Google Scholar] [CrossRef]
  154. Berner, A.K.; Brouwers, O.; Pringle, R.; Klaassen, I.; Colhoun, L.; McVicar, C.; Brockbank, S.; Curry, J.W.; Miyata, T.; Brownlee, M.; et al. Protection against methylglyoxal-derived AGEs by regulation of glyoxalase 1 prevents retinal neuroglial and vasodegenerative pathology. Diabetologia 2012, 55, 845–854. [Google Scholar] [CrossRef] [PubMed]
  155. Sachdeva, R.; Schlotterer, A.; Schumacher, D.; Matka, C.; Mathar, I.; Dietrich, N.; Medert, R.; Kriebs, U.; Lin, J.; Nawroth, P.; et al. TRPC proteins contribute to development of diabetic retinopathy and regulate glyoxalase 1 activity and methylglyoxal accumulation. Mol. Metab. 2018, 9, 156–167. [Google Scholar] [CrossRef] [PubMed]
  156. Rowan, S.; Jiang, S.; Korem, T.; Szymanski, J.; Chang, M.L.; Szelog, J.; Cassalman, C.; Dasuri, K.; McGuire, C.; Nagai, R.; et al. Involvement of a gut-retina axis in protection against dietary glycemia-induced age-related macular degeneration. Proc. Natl. Acad. Sci. USA 2017, 114, E4472–E4481. [Google Scholar] [CrossRef]
  157. Bierhaus, A.; Fleming, T.; Stoyanov, S.; Leffler, A.; Babes, A.; Neacsu, C.; Sauer, S.K.; Eberhardt, M.; Schnölzer, M.; Lasitschka, F.; et al. Methylglyoxal modification of Nav1.8 facilitates nociceptive neuron firing and causes hyperalgesia in diabetic neuropathy. Nat. Med. 2012, 18, 926–933. [Google Scholar] [CrossRef]
  158. Braun, J.D.; Pastene, D.O.; Breedijk, A.; Rodriguez, A.; Hofmann, B.B.; Sticht, C.; von Ochsenstein, E.; Allgayer, H.; van den Born, J.; Bakker, S.; et al. Methylglyoxal down-regulates the expression of cell cycle associated genes and activates the p53 pathway in human umbilical vein endothelial cells. Sci. Rep. 2019, 9, 1152. [Google Scholar] [CrossRef]
  159. Li, H.; Zheng, L.; Chen, C.; Liu, X.; Zhang, W. Brain Senescence Caused by Elevated Levels of Reactive Metabolite Methylglyoxal on D-Galactose-Induced Aging Mice. Front. Neurosci. 2019, 13, 1004. [Google Scholar] [CrossRef]
  160. Takeda, A.; Yasuda, T.; Miyata, T.; Goto, Y.; Wakai, M.; Watanabe, M.; Yasuda, Y.; Horie, K.; Inagaki, T.; Doyu, M.; et al. Advanced glycation end products co-localized with astrocytes and microglial cells in Alzheimer’s disease brain. Acta Neuropathol. 1998, 95, 555–558. [Google Scholar] [CrossRef]
  161. de Almeida, G.R.L.; Szczepanik, J.C.; Selhorst, I.; Cunha, M.P.; Dafre, A.L. The expanding impact of methylglyoxal on behavior-related disorders. Prog. Neuropsychopharmacol. Biol. Psychiatry 2023, 120, 110635. [Google Scholar] [CrossRef]
  162. Berends, E.; Pencheva, M.G.; van de Waarenburg, M.P.H.; Scheijen, J.L.J.M.; Hermes, D.J.H.P.; Wouters, K.; van Oostenbrugge, R.J.; Foulquier, S.; Schalkwijk, C.G. Glyoxalase 1 overexpression improves neurovascular coupling and limits development of mild cognitive impairment in a mouse model of type 1 diabetes. J. Physiol. 2024, 602, 6209–6223. [Google Scholar] [CrossRef] [PubMed]
  163. Hanssen, N.M.J.; Scheijen, J.L.J.M.; Jorsal, A.; Parving, H.H.; Tarnow, L.; Rossing, P.; Stehouwer, C.D.A.; Schalkwijk, C.G. Higher Plasma Methylglyoxal Levels Are Associated with Incident Cardiovascular Disease in Individuals with Type 1 Diabetes: A 12-Year Follow-up Study. Diabetes 2017, 66, 2278–2283. [Google Scholar] [CrossRef] [PubMed]
  164. Hanssen, N.M.; Teraa, M.; Scheijen, J.L.; Van de Waarenburg, M.; Gremmels, H.; Stehouwer, C.D.; Verhaar, M.C.; Schalkwijk, C.G. Plasma Methylglyoxal Levels Are Associated with Amputations and Mortality in Severe Limb Ischemia Patients with and Without Diabetes. Diabetes Care 2021, 44, 157–163. [Google Scholar] [CrossRef]
  165. Koren, O.; Spor, A.; Felin, J.; Fåk, F.; Stombaugh, J.; Tremaroli, V.; Behre, C.J.; Knight, R.; Fagerberg, B.; Ley, R.E.; et al. Human oral, gut, and plaque microbiota in patients with atherosclerosis. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. S1), 4592–4598. [Google Scholar] [CrossRef]
  166. Chhibber-Goel, J.; Singhal, V.; Bhowmik, D.; Vivek, R.; Parakh, N.; Bhargava, B.; Sharma, A. Linkages between oral commensal bacteria and atherosclerotic plaques in coronary artery disease patients. Npj Biofilms Microbiomes 2016, 2, 7. [Google Scholar] [CrossRef]
  167. Lanter, B.B.; Sauer, K.; Davies, D.G. Bacteria Present in Carotid Arterial Plaques Are Found as Biofilm Deposits Which May Contribute to Enhanced Risk of Plaque Rupture. MBio 2014, 5, 10–1128. [Google Scholar] [CrossRef] [PubMed]
  168. Hanssen, N.M.; Brouwers, O.; Gijbels, M.J.; Wouters, K.; Wijnands, E.; Cleutjens, J.P.; De Mey, J.G.; Miyata, T.; Biessen, E.A.; Stehouwer, C.D.; et al. Glyoxalase 1 overexpression does not affect atherosclerotic lesion size and severity in ApoE−/− mice with or without diabetes. Cardiovasc. Res. 2014, 104, 160–170. [Google Scholar] [CrossRef]
  169. Vulesevic, B.; McNeill, B.; Giacco, F.; Maeda, K.; Blackburn, N.J.R.; Brownlee, M.; Milne, R.W.; Suuronen, E.J. Methylglyoxal-Induced Endothelial Cell Loss and Inflammation Contribute to the Development of Diabetic Cardiomyopathy. Diabetes 2016, 65, 1699–1713. [Google Scholar] [CrossRef]
  170. Dube, G.; Tiamiou, A.; Bizet, M.; Boumahd, Y.; Gasmi, I.; Crake, R.; Bellier, J.; Nokin, M.J.; Calonne, E.; Deplus, R.; et al. Methylglyoxal: A novel upstream regulator of DNA methylation. J. Exp. Clin. Cancer Res. 2023, 42, 78. [Google Scholar] [CrossRef]
  171. Kong, L.R.; Gupta, K.; Wu, A.J.; Perera, D.; Ivanyi-Nagy, R.; Ahmed, S.M.; Tan, T.Z.; Tan, S.L.W.; Fuddin, A.; Sundaramoorthy, E.; et al. A glycolytic metabolite bypasses “two-hit” tumor suppression by BRCA2. Cell 2024, 187, 2269–2287.e16. [Google Scholar] [CrossRef] [PubMed]
  172. Bellier, J.; Nokin, M.J.; Caprasse, M.; Tiamiou, A.; Blomme, A.; Scheijen, J.L.; Koopmansch, B.; MacKay, G.M.; Chiavarina, B.; Costanza, B.; et al. Methylglyoxal Scavengers Resensitize KRAS-Mutated Colorectal Tumors to Cetuximab. Cell Rep. 2020, 30, 1400–1416.e6. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Illustration depicting the fate of MGO in human gut microbiome interactions. Red arrows indicate detrimental consequences, blue arrows indicate beneficial effects, solid black arrows represent pathways, and dashed arrows illustrate suggestive factors that may influence MGO buildup or loss. MgsA (methylglyoxal synthase), Glo3 (glyoxylase 3), FucO (1,2 propanediol oxidoreductase), G3P (glyceraldehyde 3 phosphate), DHAP (dihydroxyacetone phosphate).
Figure 1. Illustration depicting the fate of MGO in human gut microbiome interactions. Red arrows indicate detrimental consequences, blue arrows indicate beneficial effects, solid black arrows represent pathways, and dashed arrows illustrate suggestive factors that may influence MGO buildup or loss. MgsA (methylglyoxal synthase), Glo3 (glyoxylase 3), FucO (1,2 propanediol oxidoreductase), G3P (glyceraldehyde 3 phosphate), DHAP (dihydroxyacetone phosphate).
Antioxidants 14 00763 g001
Figure 2. Illustration summarizing (Table 1), showing microbes that increase or decrease in abundance in response to MGO. Red arrows are positive associations with MGO while green arrows are negative associations.
Figure 2. Illustration summarizing (Table 1), showing microbes that increase or decrease in abundance in response to MGO. Red arrows are positive associations with MGO while green arrows are negative associations.
Antioxidants 14 00763 g002
Table 1. Studies investigating the influence of MGO on the microbiome. A visual summary is provided (Figure 2).
Table 1. Studies investigating the influence of MGO on the microbiome. A visual summary is provided (Figure 2).
Study SummaryPositively Associated with MGONegatively Associated with MGOReference
Oral MGO administration in aged mice caused shifts in specific taxa and metabolic pathways, with no change in alpha or beta diversity; mice were given MGO (100 mg/kg/day) for 4 weeks.Lachnospiraceae, AkkermansiaHarryflintia, Intestinimonas, Ruminococcaceae Family, Anaerotruncus[65]
Faecal and serum samples from 26 patients with locally advanced rectal cancer showed that MGO predicts treatment response and correlates with specific microbes.Lactobacillus murinusStreptococcus anginosus, Akkermansia, Ruminococcaceae, Eubacterium, Prevotellaceae[66]
In vitro fermentation of human faecal samples with MGO was analysed using 16S rRNA gene sequencing. Principal coordinate analysis (PCoA) of the microbiome shows separation between the blank group and the MGO group.Escherichia-Shigella and KlebsiellaMegasphaera, Bifidobacterium, Megamonas[52]
Simulated digestion of dicarbonyls and in vitro fermentation with 3 human faecal samples showed that MGO and other 1,2-dicarbonyls reduced gut microbial abundance and activity. Low MGO level used—6.0 mg/kg; high MGO level used—219.6 mg/kg. Lactobacilli[53]
In 14 patients with chronic periodontitis, MGO accumulated in gingival crevicular fluid at high levels compared to the reference group.Bacteroides forsythus [67,68]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Akinrimisi, O.I.; Maasen, K.; Scheijen, J.L.J.M.; Nemet, I.; Nieuwdorp, M.; Schalkwijk, C.G.; Hanssen, N.M.J. Does Gut Microbial Methylglyoxal Metabolism Impact Human Physiology? Antioxidants 2025, 14, 763. https://doi.org/10.3390/antiox14070763

AMA Style

Akinrimisi OI, Maasen K, Scheijen JLJM, Nemet I, Nieuwdorp M, Schalkwijk CG, Hanssen NMJ. Does Gut Microbial Methylglyoxal Metabolism Impact Human Physiology? Antioxidants. 2025; 14(7):763. https://doi.org/10.3390/antiox14070763

Chicago/Turabian Style

Akinrimisi, Oluwatomisono I., Kim Maasen, Jean L. J. M. Scheijen, Ina Nemet, Max Nieuwdorp, Casper G. Schalkwijk, and Nordin M. J. Hanssen. 2025. "Does Gut Microbial Methylglyoxal Metabolism Impact Human Physiology?" Antioxidants 14, no. 7: 763. https://doi.org/10.3390/antiox14070763

APA Style

Akinrimisi, O. I., Maasen, K., Scheijen, J. L. J. M., Nemet, I., Nieuwdorp, M., Schalkwijk, C. G., & Hanssen, N. M. J. (2025). Does Gut Microbial Methylglyoxal Metabolism Impact Human Physiology? Antioxidants, 14(7), 763. https://doi.org/10.3390/antiox14070763

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop