17 Jul 2022

Redox and the gut microbiome

Host redox biology shapes the gut microbiome and vice versa; relationships which may be important in oxidative stress-associated disease and ageing. Here’s an overview…

Gut oxygen

Reduction-oxidation (redox) processes play fundamental roles in biology, while shifting redox environments have shaped the evolution of life on this planet. The primordial earth was virtually anoxic when life appeared ~3.8 billion years ago, with the advent of photosystem II (i.e. early photosynthesis) and geochemical changes eventually increasing atmospheric oxygen (O2) (i.e. Great Oxidation Event). This exposed life to a double-edged sword: a toxic oxidant and an energetically favourable respiratory acceptor. Consequently, while some committed anaerobes became confined to anoxic zones, others went aerobic, creating the present dichotomy 1,2. Moreover, aerobic metabolism facilitated the evolution of complex multicellular metazoa 3,4, and novel biogeographical redox environments therein. In particular, the human gut is populated by trillions of microbes, predominantly anaerobes, which have co-diversified with us acquiring traits such as O2 intolerance 5.

Gut O2 is dynamic and follows gradients 6; levels decline radially from epithelium to lumen and distally from small to large intestine, culminating in the largely anoxic and reducing environment of the colonic lumen—reminiscent of primordial earth. The aerotolerance of gut microbes broadly parallels their distribution, with enrichment of aerobic and facultative anaerobes in the mucosa and obligate anaerobes in the lumen 6,7. Further, it also relates to their initial colonisation by succession; infancy being characterised by initial abundance of facultative anaerobes, with obligate anaerobes increasing over 4 months 8 and 1 year 9. And notably, in children and adults, acute secretory diarrhea (i.e. V. cholerae or E. coli) depletes the gut microbiome, while repopulation also proceeded with facultative then obligate anaerobes over 30 days 10. As such, it has often been presumed that initial consumption of gut O2 by aerobes underlies the successive growth of obligate anaerobes. However, luminal anoxia was largely unaffected in germ-free mice, while in vitro experiments implicated chemical oxidation and microbial biomass as major O2 sinks 11. Also, initial colonisation by aerobic or anaerobic bacteria supported that of dominant anaerobe Bacteroides thetaiotaomicron; suggesting early colonisation by aerotolerant bacteria may simply reflect better survival ex vivo and vectorization in vivo 12. Several human studies are also informative. In 88 African-American newborns, E. coli (family Enterobacteriaceae) was the most common early coloniser and evidenced to perform anaerobic fermentation of amino acids (e.g. serine/threonine to succinate/acetate), while at 1 month bacterial diversity and carbohydrate fermentation genes increased 13. Going further, in 153 East Asian newborns followed for 4 months, Enterobacteriaceae and other facultative/aerobic bacteria were increasingly displaced by Bifidobacteria and strict anaerobes, correlating with faecal acetic acid; all of which was delayed with C-section and normalised by a Bifido-based synbiotic 8. And in 98 Swedish newborns, the microbiome was initially enriched in TCA cycle functionality (vs. pyruvate at 4 months), suggesting a more oxidative environment; while at 12 months cessation of breastfeeding marked the transition toward an adult/maternal microbiome, which included a decline in Bifidobacteria and emergence of Clostridia spp. 9. Thus Bifidobacteria and breastfeeding may shape early anaerobiosis.

Gut O2 principally arises from bloodstream delivery to the submucosa for cellular respiration, where diffusion from the epithelium 14 establishes a dynamic radial gradient 6,7; and perhaps in relation to surface area (decreases from small–large intestine) 11. Modulating intestinal oxygenation (e.g. via hyperbaric oxygen, intermittent hypoxia or mechanical ventilation) correspondingly alters the gut microbiome 14. Conversely, microbial metabolism also modulates epithelial metabolism. In particular, unabsorbed carbohydrates (i.e. fibres and resistant starches) are normally the major carbon source for microbes and (anaerobically) fermented to short-chain fatty acids (SCFAs), mainly acetate (C2:0), propionate (C3:0) and butyrate (C4:0); the latter being the preferred carbon source for colonocytes and their (aerobic) respiration. Here colonocytes exist in a state of ‘physiological hypoxia’, where butyrate supports O2 consumption and stabilises hypoxia-inducible factor (HIF), a transcription factor coordinating barrier function and antimicrobial defence 14,15 (incl. β-defensin-1 16). Further, butyrate also induces PPARg-dependant β-oxidation and inhibition of iNOS, thereby limiting O2 and nitrate (NO3), respectively, which can serve as respiratory acceptors for growth of facultative Enterobacteriaceae 17–19. Consequently, antibiotic suppression of the gut microbiome can increase host-derived O2/nitrate 15,17,18,20. Notably, to a lesser extent than butyrate, acetate also supported epithelial O2 consumption (but not HIF) 15. This may be particularly relevant during infancy, where facultative Bifidobacteria (supported by breastfeeding) may be an early driver of anaerobiosis, and perhaps via metabolism of carbohydrates to acetate in the ‘bifid shunt’ 8. Whereas cessation of breastfeeding and increased Clostridia spp. possessing the acetyl-CoA pathway (e.g. E. rectale and F. prausnitzii) have correlated faecal butyrate over 12 21 and 24 months 22.

Gut redox

O2 can directly oxidise a subset of biomolecules (e.g. flavins, quinones and metal centres), where its reduction can generate reactive oxygen species (ROS) with capacity to oxidise many others (e.g. lipids, proteins and DNA). This can occur incidentally or intentionally, while elaborate antioxidant systems serve to eliminate ROS and buffer redox changes. In particular, cellular redox poise is determined by the thiol/disulfide redox couples, whose redox potentials (Eh in mV) are held low (favouring reduction), with dynamic changes regulating cell physiology via redox signalling 23. For instance, the intestinal epitheliums rapid turnover and cell transitions (i.e. proliferation–differentiation–apoptosis) are regulated by progressive oxidation of glutathione (GSH) and thioredoxin (Trx) 24,25. The epithelium further secretes mucus and antimicrobial peptides, which co-associate to form a frontline physiochemical barrier to microbes 26. Mucus is composed of mucin oligomers assembled via disulfide bridges (S–S bonds) which stabilise its structure and confer redox-dependent fluidity, which may be modulated by luminal thiols 24,27. Moreover, reduction of α-defensin-6 (small intestine) and β-defensin-1 (esp. colon) unmasks their antimicrobial activity against commensal bacteria and opportunistic fungi, which may be mediated by epithelial Trx and the reducing luminal milieu 28–30. Reciprocally, commensal microbes stimulate epithelial receptors (e.g. TLRs and FPRs) and ROS-dependent pathways regulating cell turnover, gut barrier and immune functions 24,31,32. In particular, oxidative/xenobiotic signals are sensed by Nrf2, a master transcription factor mediating redox and epithelial homeostasis 33. Accordingly, eubiotic Lactobacilli (e.g. LGG) can induce cytoprotection via NOX1/ROS-dependant activation of Nrf2 34. And of microbial metabolites, butyrate induces Nrf2 35,36 (which may also support PPARg/β-oxidation 37–39), while urolithin A induced AhR/Nrf2-dependent enhancement of gut barrier integrity and anti-inflammatory activity 40.

In a study on 70 diverse individuals, gut microbiome aerotolerance correlated the faecal redox potential (r = 0.41) 41; a relationship which may involve various factors. Foremost, microbes depend on antioxidant activity for aerotolerance, while anaerobic metabolism may be particularly susceptible to poisoning by O2/ROS due to its dependence on radical chemistry and low-potential metal centres 1,2. Episodic disruptions can reversibly inhibit growth, whereas excessive aeration and internal Fenton chemistry can be lethal 1; i.e. electron transfer from ferrous iron (Fe(II)) to hydrogen peroxide (H2O2) forms hydroxyl radicals (HO) with capacity to irreparably damage DNA. Among antioxidants, bacteria express various catalases, a family of heme/iron or manganese-containing enzymes which decompose H2O2 to H2O and O2 42; and often used to identify aerobic/facultative bacteria (i.e. catalase test). In humans and mice, microbial catalase expression was greater in rectal mucosa vs. stool, paralleling radial O2 gradients 7. Moreover, one of the major butyrate-producing bacteria in humans, Faecalibacterium prausnitzii (i.e. ~5–10% of adult microbiome), may utilise an extracellular flavin–thiol electron shuttle (i.e. Fig. 3; resembling GSH reductase) to survive close to the epithelium; and at the expense of electron disposal via SCFA production 43. Notably, antioxidants (e.g. GSH/cysteine, ascorbic acid and uric acid) can maintain the viability of such highly sensitive anaerobic bacteria in ambient air 44,45 and even enhance Clostridia butyrate production 46.

The gut microbiome may also directly affect luminal reducing activity. Accordingly, in mice on broad antibiotics for 5 days, an increased faecal redox potential occurred within hours, which was not associated with luminal O2 and even occurred ex vivo 20. Subsequent microbiome recovery followed ecological succession (and required dispersal via cohousing), where an early increase in Enterobacteriaceae associated with a lowering of the faecal redox potential; suggesting they may act as pioneer taxa directly affecting luminal redox and driving their own displacement by secondary colonisers 20. Mechanistically, antibiotics led to a sustained increase in nitrate, a potent respiratory acceptor after O2 20 (e.g. standard redox potentials), which can particularly be exploited by Enterobacteriaceae with their high prevalence of nitrate reductase 47,48. In ruminants 49 and pigs 50, luminal redox also correlates negatively with pH and acetate, and positively with propionate. Faecal organic acids themselves may have some very mild antioxidant activity (e.g. lactic and acetic acids 51), although are tied to microbial hydrogen (H2) economy 49. H2 is a diffusible gas which selectively scavenges hydroxyl radicals 52, while also representing a primordial electron donor with potential for broad interspecies transfer and metabolism (i.e. 70% of human colonic microbes contain hydrogenases) 53; generally being released during carbohydrate fermentation and oxidised in sulfate reduction, methanogenesis and reductive acetogenesis 54. Of SCFAs, production of acetate and butyrate may favour net H2 release (e.g. via NADH–ferredoxin–hydrogenase; Fig. 1 55), whereas propionate its consumption 49,54. Notably, in 32 healthy newborns followed for 6 months, luminal H2 increased and stabilised after 1 month, while faecal SCFAs kept increasing, but with a lower proportion acetate (i.e. 99–82%) and higher propionate (i.e. 0–15%) 56. Finally, some bacteria can metabolise various sulfur-containing molecules (e.g. sulfate, cysteine and taurine) to hydrogen sulfide (H2S) gas 57, a highly reducing thiol which can modulate mucus integrity 27 and be oxidised by colonocyte mitochondria (via SQR) to support bioenergetics 58.

Microbial redox is a direct target of host immunity. In particular, the oxidative/destructive power of ROS/RNS is weaponised by innate immune cells, including phagocytes and epithelia, as a conserved form of defence. In response to microbes, mammalian epithelial barriers can produce NOX-dependent superoxide (O2), DUOX-dependant hydrogen peroxide (H2O2) 59–61 and iNOS-dependent nitric oxide (NO) 62–64, all of which may mediate antimicrobial effects. For instance, in mice, stomach infection with Helicobacter felis (a model for H. pylori) induced epithelial DUOX2 at the apical surface and release of H2O2 into the mucus layer, causing bacterial oxidative stress and limiting infection/inflammation 65; and similarly, intestinal DUOX2 regulated microbial redox and access to lymphatic tissues, thereby limiting immune responses 66. In humans, DUOX2 was upregulated in IBD/IBS 66, while rare loss-of-function variants associated with elevated blood IL-17C and mucosal expansion of Gram-negative Enterobacteriaceae 67. Notably, IL-17 cytokines support type-3 immunity against extracellular pathogens 68; including recruitment of neutrophils expressing myeloperoxidase (MPO) for production of hypochlorous acid (HOCl) 61, a chlorine-based oxidant and potent antimicrobial (also used commercially as a disinfectant). In animal models, microbes also induce co-expression of NOX and iNOS 62, and formation of peroxynitrite (ONOO; i.e. O2 + NO) 64, another antimicrobial 62–64. Moreover, epithelial NOX deficiency increased H2O2-producing Lactobacilli which mediated compensatory colonisation resistance 69 and ameliorated colitis 70. Notably, all these oxidants have potential to engage host redox signalling, where antioxidant responses may even support immunity. For instance, sulforaphane-rich broccoli sprouts induced a Nrf2-depentant reduction in gastric H. pylori colonisation and inflammation in mice and humans 71. Also, in a urinary tract infection (UTI) model, urothelial cell infection with E.coli (typically thought to originate from faeces 72) induced ROS and Nrf2, which subsequently suppressed oxidative stress and inflammation, and promoted bacterial expulsion via regulation of the RAB27B GTPase 73.

Gut dysbiosis

A dysfunctional gut–microbiome redox dialog may underlie disease. For instance, in human gastritis, tissue oxidative stress correlated microbiome redox traits (i.e. O2 respiration and antioxidant genes) used to define a ‘microbial-redox index’, as a putative indicator of mucosal health 74. Moreover, many conditions may feature general shifts from obligate to facultative anaerobes (e.g. C-section 8, malnutrition 41,75, HIV 76,77, IBD 78,79, CRC 80, anxiety/depression 81, ME/CFS 82,83, post-stroke 84–86, Parkinson’s 87,88 and MCI–Alzheimer’s 89–91), marking an increase in aerotolerant bacteria and implicating O2 as a common driver of colonic dysbiosis 78,92. Accordingly, mitochondrial dysfunction is a common feature of disease phenotypes, while mutations conferring increased mtROS decreased microbiome species diversity and major families of SCFA-producing obligate anaerobes (i.e. Lachnospiraceae and Ruminococcaceae/Oscillospira) 93. Further, in IBD, a pro-inflammatory response to β-fructan fibres (i.e. FOS/inulin) was accompanied by low faecal SCFA-producing bacteria and riboflavin (i.e. vitamin B2) 94, an antioxidant cofactor for F. prausnitzii 43; while impaired mucosal butyrate oxidation was associated with markedly decreased mitochondrial thiolase activity, which can result from increased mtROS/H2O2 95. On the other hand, oxidising conditions can favour the growth of facultative anaerobes and opportunistic pathogens (e.g. Salmonella 17,96, E. coli 78, C. rodentium 97 and V. cholerae 98) by conferring bioenergetic advantage; a situation promoted by epithelial inflammation and metabolic dysfunction 19. For instance, gut inflammation generates various novel carbon sources and respiratory acceptors 99; in particular, the iNOS–NO pathway can yield nitrate (NO3). As above, oxidising conditions may initially favour formation of peroxynitrite, which can rapidly isomerise to nitrate 48,86, a reaction also catalysed by haemoglobin (i.e. ferrous heme) 100,101, itself a faecal correlate of intestinal inflammation and cancer 102. Nitrate may induce blooms of Enterobacteriaceae—a common feature of inflammatory dysbiosis 47,78,103,104. Similarly, antibiotics can disrupt redox dynamics in the gut and promote outgrowth of Enterobacteriaceae 20; here depletion of SCFA-producing bacteria 15 was reported to increase epithelial O2/nitrate, allowing aerobic expansion of Salmonella and E. coli 17,18.

A primary function of the gut is to digest food and assimilate water and nutrients, which themselves have inherent redox activity. Even common liquids (e.g. spring, tap or soda water) may have markedly different redox potentials of relevance 41; in a colitis model, H2-enriched/reduced water restored microbial SCFA production, colonocyte metabolism (i.e. PPARg and iNOS), gut barrier and anaerobiosis, while suppressing E. coli expansion 105. Reducing activity may also be conferred by dietary antioxidants (e.g. ascorbate 106,107, polyphenols 75,108 and fibres 109,110). In particular, since many phyto-antioxidants are poorly absorbed, the gut was suggested to be a major site of action 111. An early 2-week trial with an antioxidant-rich diet (with matched macros and fibre) increased stool bulk and antioxidant content, but not Lactobacillus or Bifidobacteria (selective measurement only) 108; while in porcine models, antioxidant supplementation (as NAC 112 or antiox-blend 113) did increase intestinal levels of Lactobacillus and Bifidobacteria, at the expense of E. coli. Moreover, more recent trials with high-dose or delayed-release (colon-targeted) ascorbate have modulated the gut microbiome 106 and increased SCFA/butyrate production 107, respectively. Another important nutrient is iron, a redox-active transition metal which is incorporated into metalloproteins employed throughout metabolism 114; hence deficiency may disrupt many pathways mentioned herein (e.g. ↓ RBC/O2, NOX/MPO and catalase; ↑ mtROS) 114–116. However, free/labile iron can catalyse the Fenton reaction forming hydroxyl radicals, while high-dose iron supplementation has increased faecal ROS 117 and Enterobacteriaceae 118,119, in association with inflammation 120. Accordingly, in vitro, iron can favour the growth and virulence of enteric pathogens 121; and similarly, chronic exposure of titanium dioxide (a white pigment—E171) nanoparticles to E. coli induced adaptive morphogenesis and commensal–pathogen transition via hydroxyl radicals 122.

Children with severe malnutrition have positive/oxidised faecal redox potentials and corresponding dysbiosis 41,75. On the other hand, high fat/lard diets can also induce intestinal oxidative stress which highly correlates bacterial dysbiosis 123 and its amelioration by tea polyphenols 124. Moreover, pre-IBD was associated with a history of antibiotics and high fat intake (combined RR = 8.6; higher saturated fat p = 0.03), which in mice cooperatively impaired colonocyte mitochondrial O2 consumption resulting in overgrowth of Enterobacteriaceae, ameliorated by the PPARg agonist 5-ASA (i.e. Mesalamine) 125. Diets high in saturated fat specifically can also induce taurine-conjugated bile acids and growth of H2S-producing bacteria 126 with disruption of the gut barrier 127–129. Taurine provides organic sulfonate (SO3) for sulfite respiration by Bilophila 57,130; and notably, sodium sulfite (a preservative—E221) also induced Bilophila and E.coli in vitro 131. Excess H2S can reduce disulfide bonds in the mucus network (e.g. 2x > NAC) 27, increasing microbe/toxin exposure to the epithelium and inflammatory sequelae 132, and also impair colonocyte respiration (i.e. SCFA metabolism) 57,58,130,133. High intake of sulfur-rich proteins (e.g. meat) may also increase colon delivery of sulfur amino acids (i.e. Met/Cys) for sulfide generation 58,134. Moreover, diets high in red meat specifically can increase colonic nitrite in humans, perhaps via heme iron-dependent delivery of nitrosating agents 135; while in mice, heme (red pigment) can also suppress Gram-positive Firmicutes and induce nitrate-reducing bacteria 136 and faecal sulfide 27.

As above, host–microbe interactions regulate epithelial redox and physiology 24. Crucially, a healthy gut microbiome produces metabolites with growth-repressive effects on tumorigenesis 137, which is attenuated in people with colorectal cancer (CRC) 138. In particular, butyrate prevents genotoxicity via induction of phase 2 detox pathways (i.e. GSTs 139 and Nrf2 35,36), and has anti-proliferative and pro-apoptotic effects on colon cancer cells via induction of ROS and depletion of GSH 140–142; similarly to phytochemicals (e.g. glucosinolates and polyphenols) 36,137,139,140. Note, ROS-inducers may have differential effects on cancer cells due to their higher basal ROS and antioxidant dependence 143. Moreover, proliferating/cancer cells exhibit aerobic glycolysis (aka. Warburg effect) due to increased glucose processing saturating and outpacing mitochondrial metabolism 144. Consequently, in colon cancer cells, inefficient butyrate oxidation promoted nuclear accumulation and function as a HDAC inhibitor 145, which further decreased its oxidation 146; although others report that butyrate can suppress glucose metabolism 147 and promote its own oxidation with differential glutamine metabolism, while inhibiting cell proliferation 148. Recently, CRC was also associated with downregulation of Lactobacillus reuteri and its metabolite reuterin, which can induce protein oxidation in colon cancer cells and suppresses tumour growth 138. Conversely, chronic colonic inflammation promotes dysplasia and cancer 149. For instance, epithelial TLR4 activity was upregulated in IBD and CRC, and in mice induced tumorigenesis dependent on both DUOX2 (produces H2O2) and dysbiosis, which further increased ROS, potentially establishing a feedforward loop 149. Notably, in vitro this pathway was induced by adherent-invasive E. coli (contains LPS), but not F. prausnitzii 149. Moreover, inflammation shifts colonocyte metabolism toward glycolysis 19, while epithelial oxygenation can promote aerobic expansion of colibactin-producing E. coli, a pro-oncogenic driver species 150,151. Also, nitrate/nitrite-reducing bacteria may promote formation of carcinogenic N-nitroso compounds (NOCs; e.g. nitrosamines) 135; while sulfide-producing bacteria may open the mucus barrier to dietary heme, facilitating epithelial damage-induced hyperproliferation 27.

Systemic pathways

Beyond local effects, the gut microbiome may influence systemic redox and health via pathways affecting nutrient metabolism and redox signalling. Indeed germ-free mice reveal a broad effect of the gut microbiome on blood metabolites 152,153 and induction of phase 2 metabolism 154,155. For instance, blood indole 3-propionic acid (IPA) results entirely from bacterial deamination of tryptophan 154 and is associated with microbiome diversity (and F. prausnitzii) 156 and cardiometabolic health 157–159. IPA is a potent antioxidant in vitro (i.e. > melatonin) 160, which may underlie some beneficial effects in cell/animal models 157,158. On the other hand, in women newly diagnosed with breast cancer, gut microbiome indole biosynthesis was suppressed and correlated lymphocyte infiltration to tumours, while serum levels of IPA induced AhR/PXR-dependent oxidative/nitrosative stress (via Nrf2; iNOS and mtROS) and cytostasis in breast cancer cells 161. Beyond IPA, the gut microbiome was also linked to induction of the hepatic Nrf2-antioxidant pathway and protection against xenobiotics via Lactobacilli (e.g. LGG) and production of 5-methoxyindoleacetic acid (5-MIAA) 155. And beyond indoles, gut microbes can directly regulate systemic metabolism of the major thiol-antioxidant GSH by consuming serine/glycine precursors and limiting host availability, which may be of particular importance in age-related metabolic disease (e.g. NAFLD) 153,162. Reciprocally, biliary secretions also deliver significant GSH to the intestine 25, while liver diseases can involve GSH deficiency 163.

Age-related oxidative stress and organ dysfunction has been linked to the meta-organismal trimethylamine oxide (TMAO) pathway 164–168. Here dietary precursors (esp. carnitine and choline) fuel microbial production of TMA, which is converted by liver FMO3 to TMAO, which circulates blood before being excreted by kidneys or reduced/detoxified by the ubiquitous enzyme mARC1 169. In animals and humans, blood TMAO increases with age 164–168, and in relation to oxidative stress and vascular dysfunction, where ex vivo experiments implicate excess superoxide and impaired NO bioavailability 165,167,168. In animals this occurs alongside gut dysbiosis, including overgrowth of Proteobacteria and Desulfovibrio (a sulfate-reducing and TMA-producing genus), and increased FMO3 expression 168, while suppression of microbial TMA lyase (with DMB) normalises vascular dysfunction 165,167. Similarly, western diet-induced TMAO, vascular dysfunction, exercise intolerance and frailty were ameliorated by DMB 170. Mechanistically, a high fat diet was reported to induce colonocyte mitochondrial dysfunction, thereby increasing luminal O2/nitrate, E. coli respiration-dependent choline catabolism and blood TMAO 171. In the vasculature, TMAO may induce inflammation/oxidation via gradual accumulation of advanced glycation end products (AGEs) 172 and suppression of SIRT1/3 164,173 and Nrf2 174. Several conditions are also associated with small intestinal bacterial overgrowth (SIBO) or dysbiosis, which can be associated with vitamin B12 deficiency—thought to result from increased nutrient competition with the host and/or production of inactive cobalamin analogues 175–178. B12 is a cofactor for the remethylation of homocysteine, a source of cysteine for thiol-based redox molecules. On the other hand, gut microbes are also a potential source of several vitamins for the host 179, including folate 180–182 (another cofactor for homocysteine remethylation) and the NAD-precursor niacinamide (NAM) 183; both linked to redox biology. Moreover, an RCT with B-vitamins lowered homocysteine and TMAO proportionally 184, which might involve modulation of gut microbiome or host metabolism.

As above, SCFAs are the major end products of carbohydrate/fibre fermentation which support colonocyte metabolism; and this extends to systemic metabolism 185,186. For instance, in an RCT on 12 overweight men, acute colonic infusion of physiological SCFA mixtures increased fasting fat oxidation and resting energy expenditure 187. Accordingly, SCFAs are absorbed into peripheral circulation and can regulate extraintestinal cells via G-protein coupled receptors (GPRs) and histone deacetylases (HDACs), while butyrate in particular can induce Nrf2-dependent effects in disease models 188,189. For instance, susceptibility to sepsis-associated encephalopathy was associated with lower faecal/plasma/hippocampal butyrate, while sodium butyrate (NaB) administration normalised plasma/hippocampal butyrate, improved survival and neuroinflammation, and suppressed microglial activity in vitro via a GPR109A/Nrf2/HO-1 pathway 190. In a uveitis model, NaB suppressed the ocular inflammatory response and shifted IL-6R-dependent Th17 differentiation to Treg via a Nrf2/HO-1 pathway 191. And regarding Alzheimer’s, in neuronal cells under high cholesterol, NaB suppressed BACE1-dependent amyloidogenesis via an SMCT1/Nrf2/SOD1 pathway 192. More generally, the ability of SCFAs to support the (epithelial) gut barrier extends to the (endothelial) blood–brain barrier (BBB) 193, where propionate at least was shown to suppress mtROS and induce Nrf2 194. Of further interest, in a 2-week trial on 20 adults, switching from a western to low fat/high fibre diet not only increased colonic butyrate, but also folate 182; a required nutrient for various butyrate-producing Clostridiales (e.g. Fig. 2) 195 and the survival of colonic Tregs 196. Thus folate may support the production and protective effects of SCFAs in inflammatory/autoimmune disease (incl. uveitis) 197. Conversely, in a 2-week trial on 10 obese adults, an isocaloric low carb diet (i.e. C4%/F72%/P24%) rapidly lowered liver fat and VLDL-TGs, alongside carbohydrate-fermenting bacteria and SCFAs, while increasing folate-producing Streptococcus/Lactococcus and faecal/serum folate in association with β-hydroxybutyrate and 1C metabolism 181. Notably, SCFAs and ketones are similar molecules (esp. butyrate and β-hydroxybutyrate, respectively), but with differential immuno-metabolic effects 198–201, as may ketogenic diets long-term 202–204.

In addition to dietary fibre, plant polyphenols also have limited absorption and accumulate in the colon; here microbial metabolism generates smaller secondary metabolites with high bioavailability and bioactivity (e.g. antioxidant and anti-inflammatory), which can persist in circulation for days 205,206 and target distant organs such as the brain 207. For instance, the major green tea polyphenol, EGCG (C22H18O11), is poorly absorbed but degraded by gut microbes to pyrogallol (C6H6O3), a major metabolite excreted in human urine and more potent activator of Nrf2 in vitro 208. And dietary ellagitannins/ellagic acid (i.e. pomegranate, berries, nuts, tea, etc.) are metabolised to urolithins, which in rodents induce Nrf2-dependent health effects in the intestine 40 and elsewhere (e.g. liver 209, heart 210, brain 211, retina 212, etc.). Notably, polyphenols may also support SCFA production. For instance, in an 8-week RCT on older adults (n=51, ≥60yrs; MaPLE trial), a polyphenol-rich diet lowered intestinal permeability (i.e. zonulin) and blood pressure, while increasing butyrate-producing bacteria (e.g. Ruminococcaceae and Faecalibacterium) 213 in association with serum polyphenol metabolites and methylxanthines 214. In animal models, polyphenols can also restore SCFA production during colitis 215,216, post-antibiotic dysbiosis 217 and TMJ inflammation 218. Furthermore, diets high in carbs/fibre 156,219,220 and polyphenols 221,222 may increase blood IPA. For instance, in a MaPLE trial post-hoc analysis, the polyphenol-rich diet increased blood IPA, but only in those with normal kidney function, and in association with changes to CRP and gut microbiome (esp. + Clostridiales and – Enterobacteriales) 221.

Commensal microbes may also modulate metabolism of systemic gaseous mediators. In particular, endogenous nitric oxide (NO) is eventually oxidised to nitrate (NO3), some of which is taken up by the salivary glands and secreted into the oral cavity where it can be reduced by (nitrate-respiring) bacteria to nitrite (NO2), which can be absorbed and support systemic production of vasoactive NO 223. This pathway may be induced by exercise; for instance, in an RCT on 23 healthy adults, acute administration of antibacterial mouthwash (0.2% chlorohexidine) attenuated post-exercise (at 1–2hrs) hypotension, tissue oxygenation and salivary/blood nitrite 224. Moreover, this pathway may be supplemented by diet; for instance, in a 10-day RCT on 18 healthy young and older people, nitrate-rich beetroot juice altered the oral microbiome (e.g. Proteobacteria/Neisseria and Rothia), increased plasma nitrite and lowered blood pressure (old group only), which were correlated (e.g. NO2/SBP r = –0.73) 225. Notably, a prior meta-analysis of such RCTs suggests that even nitrate-depleted beetroot juice (as a placebo vs. others) may slightly lower blood pressure, implicating other components 226. Importantly, the fate of gastric nitrite also depends on the local redox environment. In particular, ingestion of antioxidant polyphenols may support gastric and systemic NO production, via direct reduction of nitrite and generation of relatively stable NO-donors (e.g. S-nitrosothiols), respectively, while supressing formation of carcinogenic NOCs 135, which may underlie their positive health effects 227 (contrasting those of heme iron 135). As above, intestinal microbes are also a source of H2, which can diffuse into circulation and be exhaled in breath, with interim potential to support systemic redox homeostasis and health 228,229. In particular, H2 can rapidly penetrate biomembranes and diffuse into organelles, where it may function as a selective antioxidant, removing cytotoxic radicals without disrupting normal ROS/RNS signalling (unlike vitamin C) 52,230. Furthermore, the murine gut microbiota was also shown to support systemic transsulfuration activity and endogenous H2S 231, which generally has antioxidant and anti-inflammatory activity 232.

The gut microbiome also shapes systemic immunity and consequently immunometabolism. In particular, ageing and disease are associated with impaired gut barrier function and increased translocation of LPS into the body 233–235 and brain 236; while blood LPS correlated NOX2 (r = 0.441) and oxidative stress in elderly people with neurodegenerative diseases 237. Accordingly, LPS/endotoxin is recognised by TLR4 and stimulates innate immunity and ROS/RNS, particularly via the phagocyte oxidative/respiratory burst 61,238. In mice cognitive ageing was accompanied by depletion of butyrate-producing bacteria (i.e. ↓ Roseburia and F. prausnitzii), gut/brain barrier dysfunction, elevated blood/brain LPS and systemic/cerebral inflammation 239; while age-related microglial activation was ameliorated by NaB or soluble fibre 240. Moreover, to gut–brain reciprocity, increased faecal Enterobacteriaceae has been identified as an independent risk factor for poor outcome in stroke; while in mice brain ischaemia induced rapid intestinal ischemia and oxidant production (i.e. NOX1, DUOX2 and iNOS), with ROS/RNS-dependent nitrate, Enterobacteriaceae expansion and barrier dysfunction (i.e. LPS, LBP and D-lac), which reciprocally induced systemic inflammation and worsened brain infarction 86. Note, the initial brain-to-gut pathway was suggested to involve sympathetic/β-adrenergic activation 86. A follow-up study further showed that post-stroke cognitive impairment was associated with low butyrate/Lachnospiraceae at baseline and increased Enterobacteriaceae at 3 months, when they negatively and positively correlated blood LPS, respectively; and was transmissible to mice via the gut microbiome (i.e. FMT), where it was associated with BBB disruption, hippocampal microglial activation and degeneration, and thalamic amyloid-β deposition, all of which could be replicated by injection of LPS (from E. coli) and rescued by NaB 241. Finally, in people with HIV, an increased abundance of aerotolerant bacteria was associated with worse emotional states (incl. perceived stress) and a history of depression 77; while in mice psychosocial stress induced a gut inflammatory­/oxidative response (i.e. DUOX2 and iNOS) associated with dysbiosis (incl. Lachnospiraceae) and increased microbial catalase expression 242.

Perspective

Redox processes play a defining role in metazoan and microbial biology and consequently shape the symbiosis between us and our microbiome; a relationship which may be important in oxidative stress-associated disease and ageing. From an evolutionary perspective, the huge diversity of the gut metagenome may serve to greatly augment host (or ‘holobiont’) digestive and biosynthetic capacity, supporting survival and reproductive fitness; while reciprocally, gut microbes have co-diversified with humans, acquiring traits such as reduced genomes and O2 intolerance, implying host dependency 5. In particular, maintenance of an anoxic and reducing colonic microenvironment may support a mutual redox metabolism, wherein commensal microbes (esp. obligate anaerobes) partially oxidise indigestible carbohydrates/fibres via anaerobic fermentation to SCFA end products which can be salvaged by host cells and fully oxidised via aerobic respiration, meanwhile engaging signalling pathways coordinating epithelial and immuno-metabolic homeostasis. On the contrary, increasingly aerobic and oxidising conditions may generally favour shifts from obligate to facultative anaerobes, resulting in heterogeneous and dysbiotic relationships of competition and parasitism, typified by barrier breakdown and immuno-metabolic dysregulation, and potentially representing a common pathway underlying gastrointestinal 19 and systemic disease 99. In essence, 2 key molecules may be particularly representative of this dichotomy: butyrate is a product of redox symbiosis which transduces redox homeostasis, while LPS is a product of oxidative dysbiosis which transduces oxidative stress. Many environmental factors discussed above may modulate this balance and which could be employed to support a mutual redox symbiosis and systemic health.

In memory of HC: ‘Keep smiling’.

References

1.           Lu, Z. & Imlay, J. A. When anaerobes encounter oxygen: mechanisms of oxygen toxicity, tolerance and defence. Nat. Rev. Microbiol. 19, 774–785 (2021).

2.           Khademian, M. & Imlay, J. A. How Microbes Evolved to Tolerate Oxygen. Trends Microbiol. 29, 428–440 (2021).

3.           Thannickal, V. J. Oxygen in the evolution of complex life and the price we pay. Am. J. Respir. Cell Mol. Biol. 40, 507–10 (2009).

4.           Jones, D. P. & Sies, H. The Redox Code. Antioxid. Redox Signal. 23, 734–46 (2015).

5.           Suzuki, T. A. et al. Codiversification of gut microbiota with humans. Science 377, 1328–1332 (2022).

6.           Espey, M. G. Role of oxygen gradients in shaping redox relationships between the human intestine and its microbiota. Free Radic. Biol. Med. 55, 130–40 (2013).

7.           Albenberg, L. et al. Correlation between intraluminal oxygen gradient and radial partitioning of intestinal microbiota. Gastroenterology 147, 1055–63.e8 (2014).

8.           Lay, C. et al. A synbiotic intervention modulates meta-omics signatures of gut redox potential and acidity in elective caesarean born infants. BMC Microbiol. 21, 191 (2021).

9.           Bäckhed, F. et al. Dynamics and Stabilization of the Human Gut Microbiome during the First Year of Life. Cell Host Microbe 17, 690–703 (2015).

10.        David, L. A. et al. Gut microbial succession follows acute secretory diarrhea in humans. MBio 6, e00381-15 (2015).

11.        Friedman, E. S. et al. Microbes vs. chemistry in the origin of the anaerobic gut lumen. Proc. Natl. Acad. Sci. U. S. A. 115, 4170–4175 (2018).

12.        Halpern, D. et al. Do Primocolonizing Bacteria Enable Bacteroides thetaiotaomicron Intestinal Colonization Independently of the Capacity To Consume Oxygen? mSphere 6, (2021).

13.        Bittinger, K. et al. Bacterial colonization reprograms the neonatal gut metabolome. Nat. Microbiol. 5, 838–847 (2020).

14.        Pral, L. P., Fachi, J. L., Corrêa, R. O., Colonna, M. & Vinolo, M. A. R. Hypoxia and HIF-1 as key regulators of gut microbiota and host interactions. Trends Immunol. 42, 604–621 (2021).

15.        Kelly, C. J. et al. Crosstalk between Microbiota-Derived Short-Chain Fatty Acids and Intestinal Epithelial HIF Augments Tissue Barrier Function. Cell Host Microbe 17, 662–71 (2015).

16.        Kelly, C. J. et al. Fundamental role for HIF-1α in constitutive expression of human β defensin-1. Mucosal Immunol. 6, 1110–8 (2013).

17.        Rivera-Chávez, F. et al. Depletion of Butyrate-Producing Clostridia from the Gut Microbiota Drives an Aerobic Luminal Expansion of Salmonella. Cell Host Microbe 19, 443–454 (2016).

18.        Byndloss, M. X. et al. Microbiota-activated PPAR-γ signaling inhibits dysbiotic Enterobacteriaceae expansion. Science 357, 570–575 (2017).

19.        Litvak, Y., Byndloss, M. X. & Bäumler, A. J. Colonocyte metabolism shapes the gut microbiota. Science 362, (2018).

20.        Reese, A. T. et al. Antibiotic-induced changes in the microbiota disrupt redox dynamics in the gut. Elife 7, (2018).

21.        Nilsen, M. et al. Butyrate Levels in the Transition from an Infant- to an Adult-Like Gut Microbiota Correlate with Bacterial Networks Associated with Eubacterium Rectale and Ruminococcus Gnavus. Genes (Basel). 11, (2020).

22.        Tsukuda, N. et al. Key bacterial taxa and metabolic pathways affecting gut short-chain fatty acid profiles in early life. ISME J. 15, 2574–2590 (2021).

23.        Go, Y.-M. & Jones, D. P. The redox proteome. J. Biol. Chem. 288, 26512–20 (2013).

24.        Circu, M. L. & Aw, T. Y. Redox biology of the intestine. Free Radic. Res. 45, 1245–66 (2011).

25.        Circu, M. L. & Aw, T. Y. Intestinal redox biology and oxidative stress. Semin. Cell Dev. Biol. 23, 729–37 (2012).

26.        Dupont, A., Heinbockel, L., Brandenburg, K. & Hornef, M. W. Antimicrobial peptides and the enteric mucus layer act in concert to protect the intestinal mucosa. Gut Microbes 5, 761–5 (2014).

27.        Ijssennagger, N. et al. Gut microbiota facilitates dietary heme-induced epithelial hyperproliferation by opening the mucus barrier in colon. Proc. Natl. Acad. Sci. U. S. A. 112, 10038–43 (2015).

28.        Schroeder, B. O. et al. Paneth cell α-defensin 6 (HD-6) is an antimicrobial peptide. Mucosal Immunol. 8, 661–71 (2015).

29.        Schroeder, B. O. et al. Reduction of disulphide bonds unmasks potent antimicrobial activity of human β-defensin 1. Nature 469, 419–23 (2011).

30.        Jaeger, S. U. et al. Cell-mediated reduction of human β-defensin 1: a major role for mucosal thioredoxin. Mucosal Immunol. 6, 1179–90 (2013).

31.        Neish, A. S. & Jones, R. M. Redox signaling mediates symbiosis between the gut microbiota and the intestine. Gut Microbes 5, 250–3 (2014).

32.        van der Post, S., Birchenough, G. M. H. & Held, J. M. NOX1-dependent redox signaling potentiates colonic stem cell proliferation to adapt to the intestinal microbiota by linking EGFR and TLR activation. Cell Rep. 35, 108949 (2021).

33.        Wen, Z. et al. A Protective Role of the NRF2-Keap1 Pathway in Maintaining Intestinal Barrier Function. Oxid. Med. Cell. Longev. 2019, 1759149 (2019).

34.        Jones, R. M. et al. Lactobacilli Modulate Epithelial Cytoprotection through the Nrf2 Pathway. Cell Rep. 12, 1217–1225 (2015).

35.        Yaku, K. et al. The enhancement of phase 2 enzyme activities by sodium butyrate in normal intestinal epithelial cells is associated with Nrf2 and p53. Mol. Cell. Biochem. 370, 7–14 (2012).

36.        Yaku, K., Matsui-Yuasa, I., Konishi, Y. & Kojima-Yuasa, A. AMPK synergizes with the combined treatment of 1’-acetoxychavicol acetate and sodium butyrate to upregulate phase II detoxifying enzyme activities. Mol. Nutr. Food Res. 57, 1198–208 (2013).

37.        Polvani, S., Tarocchi, M. & Galli, A. PPARγ and Oxidative Stress: Con(β) Catenating NRF2 and FOXO. PPAR Res. 2012, 641087 (2012).

38.        Dinkova-Kostova, A. T. & Abramov, A. Y. The emerging role of Nrf2 in mitochondrial function. Free Radic. Biol. Med. 88, 179–88 (2015).

39.        Lin, T.-Y., Cantley, L. C. & DeNicola, G. M. NRF2 Rewires Cellular Metabolism to Support the Antioxidant Response. in A Master Regulator of Oxidative Stress - The Transcription Factor Nrf2 21–48 (InTech, 2016). doi:10.5772/65141

40.        Singh, R. et al. Enhancement of the gut barrier integrity by a microbial metabolite through the Nrf2 pathway. Nat. Commun. 10, 89 (2019).

41.        Million, M. & Raoult, D. Linking gut redox to human microbiome. Hum. Microbiome J. 10, 27–32 (2018).

42.        Yuan, F. et al. The Richness and Diversity of Catalases in Bacteria. Front. Microbiol. 12, 645477 (2021).

43.        Khan, M. T. et al. The gut anaerobe Faecalibacterium prausnitzii uses an extracellular electron shuttle to grow at oxic-anoxic interphases. ISME J. 6, 1578–85 (2012).

44.        Khan, M. T., van Dijl, J. M. & Harmsen, H. J. M. Antioxidants keep the potentially probiotic but highly oxygen-sensitive human gut bacterium Faecalibacterium prausnitzii alive at ambient air. PLoS One 9, e96097 (2014).

45.        La Scola, B., Khelaifia, S., Lagier, J.-C. & Raoult, D. Aerobic culture of anaerobic bacteria using antioxidants: a preliminary report. Eur. J. Clin. Microbiol. Infect. Dis. 33, 1781–3 (2014).

46.        Million, M. et al. The Antioxidants Glutathione, Ascorbic Acid and Uric Acid Maintain Butyrate Production by Human Gut Clostridia in The Presence of Oxygen In Vitro. Sci. Rep. 10, 7705 (2020).

47.        Winter, S. E. & Bäumler, A. J. Dysbiosis in the inflamed intestine: chance favors the prepared microbe. Gut Microbes 5, 71–3 (2014).

48.        Lopez, C. A. et al. Phage-mediated acquisition of a type III secreted effector protein boosts growth of salmonella by nitrate respiration. MBio 3, (2012).

49.        Huang, Y., Marden, J. P., Julien, C. & Bayourthe, C. Redox potential: An intrinsic parameter of the rumen environment. J. Anim. Physiol. Anim. Nutr. (Berl). 102, 393–402 (2018).

50.        Lizardo, R. et al. Redox potential of cecum content of growing pigs and its relation with pH and VFA concentration. J. Anim. Sci. 90 Suppl 4, 409–11 (2012).

51.        Zhang, B. et al. Effects of Organic Acids, Amino Acids and Phenolic Compounds on Antioxidant Characteristic of Zhenjiang Aromatic Vinegar. Molecules 24, (2019).

52.        Ohsawa, I. et al. Hydrogen acts as a therapeutic antioxidant by selectively reducing cytotoxic oxygen radicals. Nat. Med. 13, 688–94 (2007).

53.        Wolf, P. G., Biswas, A., Morales, S. E., Greening, C. & Gaskins, H. R. H2 metabolism is widespread and diverse among human colonic microbes. Gut Microbes 7, 235–45 (2016).

54.        Smith, N. W., Shorten, P. R., Altermann, E. H., Roy, N. C. & McNabb, W. C. Hydrogen cross-feeders of the human gastrointestinal tract. Gut Microbes 10, 270–288 (2019).

55.        Yu, R., Wang, R., Bi, T., Sun, W. & Zhou, Z. Blocking the butyrate-formation pathway impairs hydrogen production in Clostridium perfringens. Acta Biochim. Biophys. Sin. (Shanghai). 45, 408–15 (2013).

56.        Sobko, T., Norman, M., Norin, E., Gustafsson, L. E. & Lundberg, J. O. Birth-related increase in intracolonic hydrogen gas and nitric oxide as indicator of host-microbial interactions. Allergy 60, 396–400 (2005).

57.        Carbonero, F., Benefiel, A. C., Alizadeh-Ghamsari, A. H. & Gaskins, H. R. Microbial pathways in colonic sulfur metabolism and links with health and disease. Front. Physiol. 3, 448 (2012).

58.        Blachier, F., Beaumont, M. & Kim, E. Cysteine-derived hydrogen sulfide and gut health: a matter of endogenous or bacterial origin. Curr. Opin. Clin. Nutr. Metab. Care 22, 68–75 (2019).

59.        Lindquist, R. L., Bayat-Sarmadi, J., Leben, R., Niesner, R. & Hauser, A. E. NAD(P)H Oxidase Activity in the Small Intestine Is Predominantly Found in Enterocytes, Not Professional Phagocytes. Int. J. Mol. Sci. 19, (2018).

60.        Lipinski, S. et al. DUOX2-derived reactive oxygen species are effectors of NOD2-mediated antibacterial responses. J. Cell Sci. 122, 3522–30 (2009).

61.        Rada, B. & Leto, T. L. Oxidative innate immune defenses by Nox/Duox family NADPH oxidases. Contrib. Microbiol. 15, 164–87 (2008).

62.        Matziouridou, C. et al. iNOS- and NOX1-dependent ROS production maintains bacterial homeostasis in the ileum of mice. Mucosal Immunol. 11, 774–784 (2018).

63.        Gookin, J. L. et al. NF-kappaB-mediated expression of iNOS promotes epithelial defense against infection by Cryptosporidium parvum in neonatal piglets. Am. J. Physiol. Gastrointest. Liver Physiol. 290, G164-74 (2006).

64.        Gookin, J. L., Allen, J., Chiang, S., Duckett, L. & Armstrong, M. U. Local peroxynitrite formation contributes to early control of Cryptosporidium parvum infection. Infect. Immun. 73, 3929–36 (2005).

65.        Grasberger, H., El-Zaatari, M., Dang, D. T. & Merchant, J. L. Dual oxidases control release of hydrogen peroxide by the gastric epithelium to prevent Helicobacter felis infection and inflammation in mice. Gastroenterology 145, 1045–54 (2013).

66.        Grasberger, H. et al. Increased Expression of DUOX2 Is an Epithelial Response to Mucosal Dysbiosis Required for Immune Homeostasis in Mouse Intestine. Gastroenterology 149, 1849–59 (2015).

67.        Grasberger, H. et al. DUOX2 variants associate with preclinical disturbances in microbiota-immune homeostasis and increased inflammatory bowel disease risk. J. Clin. Invest. 131, (2021).

68.        Annunziato, F., Romagnani, C. & Romagnani, S. The 3 major types of innate and adaptive cell-mediated effector immunity. J. Allergy Clin. Immunol. 135, 626–35 (2015).

69.        Knaus, U. G., Hertzberger, R., Pircalabioru, G. G., Yousefi, S. P. M. & Branco Dos Santos, F. Pathogen control at the intestinal mucosa - H2O2 to the rescue. Gut Microbes 8, 67–74 (2017).

70.        Singh, A. K., Hertzberger, R. Y. & Knaus, U. G. Hydrogen peroxide production by lactobacilli promotes epithelial restitution during colitis. Redox Biol. 16, 11–20 (2018).

71.        Yanaka, A. et al. Dietary sulforaphane-rich broccoli sprouts reduce colonization and attenuate gastritis in Helicobacter pylori-infected mice and humans. Cancer Prev. Res. (Phila). 2, 353–60 (2009).

72.        Jones-Freeman, B. et al. The microbiome and host mucosal interactions in urinary tract diseases. Mucosal Immunol. 14, 779–792 (2021).

73.        Joshi, C. S., Mora, A., Felder, P. A. & Mysorekar, I. U. NRF2 promotes urothelial cell response to bacterial infection by regulating reactive oxygen species and RAB27B expression. Cell Rep. 37, 109856 (2021).

74.        Panigrahi, M. K., Kaliaperumal, V., Akella, A., Venugopal, G. & Ramadass, B. Mapping microbiome-redox spectrum and evaluating Microbial-Redox Index in chronic gastritis. Sci. Rep. 12, 8450 (2022).

75.        Million, M. et al. Increased Gut Redox and Depletion of Anaerobic and Methanogenic Prokaryotes in Severe Acute Malnutrition. Sci. Rep. 6, 26051 (2016).

76.        Dubourg, G. et al. Gut microbiota associated with HIV infection is significantly enriched in bacteria tolerant to oxygen. BMJ open Gastroenterol. 3, e000080 (2016).

77.        Pérez-Santiago, J. et al. Gut microbiota dysbiosis is associated with worse emotional states in HIV infection. J. Neurovirol. 27, 228–238 (2021).

78.        Rivera-Chávez, F., Lopez, C. A. & Bäumler, A. J. Oxygen as a driver of gut dysbiosis. Free Radic. Biol. Med. 105, 93–101 (2017).

79.        Strus, M. et al. A role of hydrogen peroxide producing commensal bacteria present in colon of adolescents with inflammatory bowel disease in perpetuation of the inflammatory process. J. Physiol. Pharmacol. 60 Suppl 6, 49–54 (2009).

80.        Liu, W. et al. Study of the Relationship between Microbiome and Colorectal Cancer Susceptibility Using 16SrRNA Sequencing. Biomed Res. Int. 2020, 7828392 (2020).

81.        Simpson, C. A. et al. The gut microbiota in anxiety and depression - A systematic review. Clin. Psychol. Rev. 83, 101943 (2021).

82.        Varesi, A., Deumer, U.-S., Ananth, S. & Ricevuti, G. The Emerging Role of Gut Microbiota in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS): Current Evidence and Potential Therapeutic Applications. J. Clin. Med. 10, (2021).

83.        Giloteaux, L. et al. Reduced diversity and altered composition of the gut microbiome in individuals with myalgic encephalomyelitis/chronic fatigue syndrome. Microbiome 4, 30 (2016).

84.        Ling, Y. et al. Gut Microbiome Signatures Are Biomarkers for Cognitive Impairment in Patients With Ischemic Stroke. Front. Aging Neurosci. 12, 511562 (2020).

85.        Ling, Y. et al. Structural Change of Gut Microbiota in Patients with Post-Stroke Comorbid Cognitive Impairment and Depression and Its Correlation with Clinical Features. J. Alzheimers. Dis. 77, 1595–1608 (2020).

86.        Xu, K. et al. Rapid gut dysbiosis induced by stroke exacerbates brain infarction in turn. Gut (2021). doi:10.1136/gutjnl-2020-323263

87.        Keshavarzian, A. et al. Colonic bacterial composition in Parkinson’s disease. Mov. Disord. 30, 1351–60 (2015).

88.        Hirayama, M. & Ohno, K. Parkinson’s Disease and Gut Microbiota. Ann. Nutr. Metab. 77 Suppl 2, 28–35 (2021).

89.        Hou, M., Xu, G., Ran, M., Luo, W. & Wang, H. APOE-ε4 Carrier Status and Gut Microbiota Dysbiosis in Patients With Alzheimer Disease. Front. Neurosci. 15, 619051 (2021).

90.        Liu, P. et al. Altered microbiomes distinguish Alzheimer’s disease from amnestic mild cognitive impairment and health in a Chinese cohort. Brain. Behav. Immun. 80, 633–643 (2019).

91.        Li, B. et al. Mild cognitive impairment has similar alterations as Alzheimer’s disease in gut microbiota. Alzheimers. Dement. 15, 1357–1366 (2019).

92.        Rigottier-Gois, L. Dysbiosis in inflammatory bowel diseases: the oxygen hypothesis. ISME J. 7, 1256–61 (2013).

93.        Yardeni, T. et al. Host mitochondria influence gut microbiome diversity: A role for ROS. Sci. Signal. 12, (2019).

94.        Armstrong, H. K. et al. Unfermented β-fructan fibers fuel inflammation in select inflammatory bowel disease patients. Gastroenterology (2022). doi:10.1053/j.gastro.2022.09.034

95.        Santhanam, S., Venkatraman, A. & Ramakrishna, B. S. Impairment of mitochondrial acetoacetyl CoA thiolase activity in the colonic mucosa of patients with ulcerative colitis. Gut 56, 1543–9 (2007).

96.        Faber, F. et al. Host-mediated sugar oxidation promotes post-antibiotic pathogen expansion. Nature 534, 697–9 (2016).

97.        Miller, B. M. et al. Anaerobic Respiration of NOX1-Derived Hydrogen Peroxide Licenses Bacterial Growth at the Colonic Surface. Cell Host Microbe 28, 789-797.e5 (2020).

98.        Van Alst, A. J. & DiRita, V. J. Aerobic Metabolism in Vibrio cholerae Is Required for Population Expansion during Infection. MBio 11, (2020).

99.        Shelton, C. D. & Byndloss, M. X. Gut Epithelial Metabolism as a Key Driver of Intestinal Dysbiosis Associated with Noncommunicable Diseases. Infect. Immun. 88, 1–14 (2020).

100.      Romero, N. et al. Reaction of human hemoglobin with peroxynitrite. Isomerization to nitrate and secondary formation of protein radicals. J. Biol. Chem. 278, 44049–57 (2003).

101.      Herold, S. & Shivashankar, K. Metmyoglobin and methemoglobin catalyze the isomerization of peroxynitrite to nitrate. Biochemistry 42, 14036–46 (2003).

102.      Mooiweer, E., Fidder, H. H., Siersema, P. D., Laheij, R. J. F. & Oldenburg, B. Fecal hemoglobin and calprotectin are equally effective in identifying patients with inflammatory bowel disease with active endoscopic inflammation. Inflamm. Bowel Dis. 20, 307–14 (2014).

103.      Winter, S. E. & Bäumler, A. J. Why related bacterial species bloom simultaneously in the gut: Principles underlying the ‘like will to like’ concept. Cell. Microbiol. 16, 179–184 (2014).

104.      Winter, S. E., Lopez, C. a & Bäumler, A. J. The dynamics of gut-associated microbial communities during inflammation. EMBO Rep. 14, 319–27 (2013).

105.      Ge, L. et al. Microbial hydrogen economy alleviates colitis by reprogramming colonocyte metabolism and reinforcing intestinal barrier. Gut Microbes 14, 2013764 (2022).

106.      Otten, A. T. et al. Vitamin C Supplementation in Healthy Individuals Leads to Shifts of Bacterial Populations in the Gut-A Pilot Study. Antioxidants (Basel, Switzerland) 10, (2021).

107.      Pham, V. T. et al. Effects of colon-targeted vitamins on the composition and metabolic activity of the human gut microbiome- a pilot study. Gut Microbes 13, 1–20 (2021).

108.      Bianchi, M. A. et al. Ability of a high-total antioxidant capacity diet to increase stool weight and bowel antioxidant status in human subjects. Br. J. Nutr. 104, 1500–7 (2010).

109.      Franco-Robles, E. & López, M. G. Implication of Fructans in Health: Immunomodulatory and Antioxidant Mechanisms. ScientificWorldJournal. 2015, 289267 (2015).

110.      Pasqualetti, V. et al. Antioxidant activity of inulin and its role in the prevention of human colonic muscle cell impairment induced by lipopolysaccharide mucosal exposure. PLoS One 9, e98031 (2014).

111.      Halliwell, B., Zhao, K. & Whiteman, M. The gastrointestinal tract: a major site of antioxidant action? Free Radic. Res. 33, 819–30 (2000).

112.      Xu, C. C. et al. Regulation of N-acetyl cysteine on gut redox status and major microbiota in weaned piglets. J. Anim. Sci. 92, 1504–11 (2014).

113.      Xu, J. et al. Regulation of an antioxidant blend on intestinal redox status and major microbiota in early weaned piglets. Nutrition 30, 584–9 (2014).

114.      Galaris, D., Barbouti, A. & Pantopoulos, K. Iron homeostasis and oxidative stress: An intimate relationship. Biochim. Biophys. acta. Mol. cell Res. 1866, 118535 (2019).

115.      Frost, J. N. et al. Plasma iron controls neutrophil production and function. Sci. Adv. 8, eabq5384 (2022).

116.      Li, L. & Frei, B. Prolonged exposure to LPS increases iron, heme, and p22phox levels and NADPH oxidase activity in human aortic endothelial cells: inhibition by desferrioxamine. Arterioscler. Thromb. Vasc. Biol. 29, 732–8 (2009).

117.      Orozco, M. N., Arriaga, C., Solomons, N. W. & Schümann, K. Equivalent effects on fecal reactive oxygen species generation with oral supplementation of three iron compounds: ferrous sulfate, sodium iron EDTA and iron polymaltose. Ann. Nutr. Metab. 60, 108–14 (2012).

118.      Tompkins, G. R., O’Dell, N. L., Bryson, I. T. & Pennington, C. B. The effects of dietary ferric iron and iron deprivation on the bacterial composition of the mouse intestine. Curr. Microbiol. 43, 38–42 (2001).

119.      Sun, J., Hu, X.-L., Le, G.-W. & Shi, Y.-H. Inhibition of Fe-induced colon oxidative stress by lactobacilli in mice. World J. Microbiol. Biotechnol. 29, 209–16 (2013).

120.      Paganini, D. & Zimmermann, M. B. The effects of iron fortification and supplementation on the gut microbiome and diarrhea in infants and children: a review. Am. J. Clin. Nutr. 106, 1688S-1693S (2017).

121.      Kortman, G. A. M., Boleij, A., Swinkels, D. W. & Tjalsma, H. Iron availability increases the pathogenic potential of Salmonella typhimurium and other enteric pathogens at the intestinal epithelial interface. PLoS One 7, e29968 (2012).

122.      Zhang, Q., Xia, T. & Zhang, C. Chronic Exposure to Titanium Dioxide Nanoparticles Induces Commensal-to-Pathogen Transition in Escherichia coli. Environ. Sci. Technol. 54, 13186–13196 (2020).

123.      Qiao, Y., Sun, J., Ding, Y., Le, G. & Shi, Y. Alterations of the gut microbiota in high-fat diet mice is strongly linked to oxidative stress. Appl. Microbiol. Biotechnol. 97, 1689–97 (2013).

124.      Ma, H. et al. Correlation Analysis of Intestinal Redox State with the Gut Microbiota Reveals the Positive Intervention of Tea Polyphenols on Hyperlipidemia in High Fat Diet Fed Mice. J. Agric. Food Chem. 67, 7325–7335 (2019).

125.      Lee, J.-Y. et al. High-Fat Diet and Antibiotics Cooperatively Impair Mitochondrial Bioenergetics to Trigger Dysbiosis that Exacerbates Pre-inflammatory Bowel Disease. Cell Host Microbe 28, 273-284.e6 (2020).

126.      Devkota, S. et al. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10-/- mice. Nature 487, 104–8 (2012).

127.      Lam, Y. Y. et al. Effects of dietary fat profile on gut permeability and microbiota and their relationships with metabolic changes in mice. Obesity (Silver Spring). 23, 1429–39 (2015).

128.      Natividad, J. M. et al. Bilophila wadsworthia aggravates high fat diet induced metabolic dysfunctions in mice. Nat. Commun. 9, 2802 (2018).

129.      Zhuang, P. et al. Eicosapentaenoic and Docosahexaenoic Acids Differentially Alter Gut Microbiome and Reverse High-Fat Diet–Induced Insulin Resistance. Mol. Nutr. Food Res. 64, 1900946 (2020).

130.      Shen, W., Gaskins, H. R. & McIntosh, M. K. Influence of dietary fat on intestinal microbes, inflammation, barrier function and metabolic outcomes. J. Nutr. Biochem. 25, 270–280 (2014).

131.      Gerasimidis, K. et al. The impact of food additives, artificial sweeteners and domestic hygiene products on the human gut microbiome and its fibre fermentation capacity. Eur. J. Nutr. 59, 3213–3230 (2020).

132.      Ijssennagger, N., van der Meer, R. & van Mil, S. W. C. Sulfide as a Mucus Barrier-Breaker in Inflammatory Bowel Disease? Trends Mol. Med. 22, 190–199 (2016).

133.      Teigen, L. M. et al. Dietary Factors in Sulfur Metabolism and Pathogenesis of Ulcerative Colitis. Nutrients 11, 1–20 (2019).

134.      Yao, C. K., Muir, J. G. & Gibson, P. R. Review article: insights into colonic protein fermentation, its modulation and potential health implications. Aliment. Pharmacol. Ther. 43, 181–96 (2016).

135.      Kobayashi, J. Effect of diet and gut environment on the gastrointestinal formation of N-nitroso compounds: A review. Nitric oxide  Biol. Chem. 73, 66–73 (2018).

136.      IJssennagger, N. et al. Dietary heme alters microbiota and mucosa of mouse colon without functional changes in host-microbe cross-talk. PLoS One 7, e49868 (2012).

137.      Beyer-Sehlmeyer, G. et al. Butyrate is only one of several growth inhibitors produced during gut flora-mediated fermentation of dietary fibre sources. Br. J. Nutr. 90, 1057–70 (2003).

138.      Bell, H. N. et al. Reuterin in the healthy gut microbiome suppresses colorectal cancer growth through altering redox balance. Cancer Cell 40, 185-200.e6 (2022).

139.      Pool-Zobel, B., Veeriah, S. & Böhmer, F.-D. Modulation of xenobiotic metabolising enzymes by anticarcinogens -- focus on glutathione S-transferases and their role as targets of dietary chemoprevention in colorectal carcinogenesis. Mutat. Res. 591, 74–92 (2005).

140.      Odom, R. Y., Dansby, M. Y., Rollins-Hairston, A. M., Jackson, K. M. & Kirlin, W. G. Phytochemical induction of cell cycle arrest by glutathione oxidation and reversal by N-acetylcysteine in human colon carcinoma cells. Nutr. Cancer 61, 332–9 (2009).

141.      Giardina, C. & Inan, M. S. Nonsteroidal anti-inflammatory drugs, short-chain fatty acids, and reactive oxygen metabolism in human colorectal cancer cells. Biochim. Biophys. Acta 1401, 277–88 (1998).

142.      Matthews, G. M., Howarth, G. S. & Butler, R. N. Short-chain fatty acids induce apoptosis in colon cancer cells associated with changes to intracellular redox state and glucose metabolism. Chemotherapy 58, 102–9 (2012).

143.      Basak, D., Uddin, M. N. & Hancock, J. The Role of Oxidative Stress and Its Counteractive Utility in Colorectal Cancer (CRC). Cancers (Basel). 12, 1–31 (2020).

144.      Wang, Y. et al. Saturation of the mitochondrial NADH shuttles drives aerobic glycolysis in proliferating cells. Mol. Cell (2022). doi:10.1016/j.molcel.2022.07.007

145.      Donohoe, D. R. et al. The Warburg effect dictates the mechanism of butyrate-mediated histone acetylation and cell proliferation. Mol. Cell 48, 612–26 (2012).

146.      Han, A., Bennett, N., Ahmed, B., Whelan, J. & Donohoe, D. R. Butyrate decreases its own oxidation in colorectal cancer cells through inhibition of histone deacetylases. Oncotarget 9, 27280–27292 (2018).

147.      Geng, H.-W., Yin, F.-Y., Zhang, Z.-F., Gong, X. & Yang, Y. Butyrate Suppresses Glucose Metabolism of Colorectal Cancer Cells via GPR109a-AKT Signaling Pathway and Enhances Chemotherapy. Front. Mol. Biosci. 8, 634874 (2021).

148.      Blouin, J.-M. et al. Butyrate elicits a metabolic switch in human colon cancer cells by targeting the pyruvate dehydrogenase complex. Int. J. cancer 128, 2591–601 (2011).

149.      Burgueño, J. F. et al. Epithelial TLR4 Signaling Activates DUOX2 to Induce Microbiota-Driven Tumorigenesis. Gastroenterology 160, 797-808.e6 (2021).

150.      Cevallos, S. A. et al. Increased Epithelial Oxygenation Links Colitis to an Expansion of Tumorigenic Bacteria. MBio 10, (2019).

151.      Zhu, W. et al. Editing of the gut microbiota reduces carcinogenesis in mouse models of colitis-associated colorectal cancer. J. Exp. Med. 216, 2378–2393 (2019).

152.      Zheng, X. et al. The footprints of gut microbial-mammalian co-metabolism. J. Proteome Res. 10, 5512–22 (2011).

153.      Mardinoglu, A. et al. The gut microbiota modulates host amino acid and glutathione metabolism in mice. Mol. Syst. Biol. 11, 834 (2015).

154.      Wikoff, W. R. et al. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc. Natl. Acad. Sci. U. S. A. 106, 3698–703 (2009).

155.      Saeedi, B. J. et al. Gut-Resident Lactobacilli Activate Hepatic Nrf2 and Protect Against Oxidative Liver Injury. Cell Metab. 31, 956-968.e5 (2020).

156.      Menni, C. et al. Circulating levels of the anti-oxidant indoleproprionic acid are associated with higher gut microbiome diversity. Gut Microbes 10, 688–695 (2019).

157.      Konopelski, P. & Mogilnicka, I. Biological Effects of Indole-3-Propionic Acid, a Gut Microbiota-Derived Metabolite, and Its Precursor Tryptophan in Mammals’ Health and Disease. Int. J. Mol. Sci. 23, (2022).

158.      Zhang, B. et al. The Mechanism Underlying the Influence of Indole-3-Propionic Acid: A Relevance to Metabolic Disorders. Front. Endocrinol. (Lausanne). 13, 841703 (2022).

159.      Xue, H. et al. Gut Microbially Produced Indole-3-Propionic Acid Inhibits Atherosclerosis by Promoting Reverse Cholesterol Transport and Its Deficiency Is Causally Related to Atherosclerotic Cardiovascular Disease. Circ. Res. 131, 404–420 (2022).

160.      Chyan, Y. J. et al. Potent neuroprotective properties against the Alzheimer beta-amyloid by an endogenous melatonin-related indole structure, indole-3-propionic acid. J. Biol. Chem. 274, 21937–42 (1999).

161.      Sári, Z. et al. Indolepropionic Acid, a Metabolite of the Microbiome, Has Cytostatic Properties in Breast Cancer by Activating AHR and PXR Receptors and Inducing Oxidative Stress. Cancers (Basel). 12, (2020).

162.      Mardinoglu, A. et al. Personal model-assisted identification of NAD+ and glutathione metabolism as intervention target in NAFLD. Mol. Syst. Biol. 13, 916 (2017).

163.      Vairetti, M. et al. Changes in Glutathione Content in Liver Diseases: An Update. Antioxidants (Basel, Switzerland) 10, (2021).

164.      Ke, Y. et al. Gut flora-dependent metabolite Trimethylamine-N-oxide accelerates endothelial cell senescence and vascular aging through oxidative stress. Free Radic. Biol. Med. 116, 88–100 (2018).

165.      Brunt, V. E. et al. Trimethylamine-N-Oxide Promotes Age-Related Vascular Oxidative Stress and Endothelial Dysfunction in Mice and Healthy Humans. Hypertension 76, 101–112 (2020).

166.      Brunt, V. E. et al. The gut microbiome-derived metabolite trimethylamine N-oxide modulates neuroinflammation and cognitive function with aging. GeroScience 43, 377–394 (2021).

167.      Li, T., Chen, Y., Gua, C. & Li, X. Elevated Circulating Trimethylamine N-Oxide Levels Contribute to Endothelial Dysfunction in Aged Rats through Vascular Inflammation and Oxidative Stress. Front. Physiol. 8, 350 (2017).

168.      Brunt, V. E. et al. Suppression of the gut microbiome ameliorates age-related arterial dysfunction and oxidative stress in mice. J. Physiol. 597, 2361–2378 (2019).

169.      Schneider, J., Girreser, U., Havemeyer, A., Bittner, F. & Clement, B. Detoxification of Trimethylamine N-Oxide by the Mitochondrial Amidoxime Reducing Component mARC. Chem. Res. Toxicol. 31, 447–453 (2018).

170.      Brunt, V. E. et al. Suppression of trimethylamine N-oxide with DMB mitigates vascular dysfunction, exercise intolerance, and frailty associated with a Western-style diet in mice. J. Appl. Physiol. 133, 798–813 (2022).

171.      Yoo, W. et al. High-fat diet-induced colonocyte dysfunction escalates microbiota-derived trimethylamine N-oxide. Science 373, 813–818 (2021).

172.      Brunt, V. E. et al. Gut Microbiome-Derived Metabolite Trimethylamine N-Oxide Induces Aortic Stiffening and Increases Systolic Blood Pressure With Aging in Mice and Humans. Hypertens. (Dallas, Tex.  1979) 78, 499–511 (2021).

173.      Chen, M.-L. et al. Trimethylamine-N-Oxide Induces Vascular Inflammation by Activating the NLRP3 Inflammasome Through the SIRT3-SOD2-mtROS Signaling Pathway. J. Am. Heart Assoc. 6, (2017).

174.      González-Correa, C. et al. Trimethylamine N-Oxide Promotes Autoimmunity and a Loss of Vascular Function in Toll-like Receptor 7-Driven Lupus Mice. Antioxidants (Basel, Switzerland) 11, (2021).

175.      Elphick, D. A. et al. Small bowel bacterial overgrowth in symptomatic older people: Can it be diagnosed earlier? Gerontology 51, 396–401 (2005).

176.      Madigan, K. E., Bundy, R. & Weinberg, R. B. Distinctive Clinical Correlates of Small Intestinal Bacterial Overgrowth with Methanogens. Clin. Gastroenterol. Hepatol. (2021). doi:10.1016/j.cgh.2021.09.035

177.      Degnan, P. H., Taga, M. E. & Goodman, A. L. Vitamin B12 as a modulator of gut microbial ecology. Cell Metab. 20, 769–78 (2014).

178.      Greco, A. et al. Glucose breath test and Crohn’s disease: Diagnosis of small intestinal bacterial overgrowth and evaluation of therapeutic response. Scand. J. Gastroenterol. 50, 1376–81 (2015).

179.      Magnúsdóttir, S., Ravcheev, D., de Crécy-Lagard, V. & Thiele, I. Systematic genome assessment of B-vitamin biosynthesis suggests co-operation among gut microbes. Front. Genet. 6, (2015).

180.      Lakoff, A. et al. Folate is absorbed across the human colon: evidence by using enteric-coated caplets containing 13C-labeled [6S]-5-formyltetrahydrofolate. Am. J. Clin. Nutr. 100, 1278–86 (2014).

181.      Mardinoglu, A. et al. An Integrated Understanding of the Rapid Metabolic Benefits of a Carbohydrate-Restricted Diet on Hepatic Steatosis in Humans. Cell Metab. 27, 559-571.e5 (2018).

182.      Chan, Y.-M., Aufreiter, S., O’Keefe, S. J. & O’Connor, D. L. Switching to a fibre-rich and low-fat diet increases colonic folate contents among African Americans. Appl. Physiol. Nutr. Metab. 44, 127–132 (2019).

183.      Blacher, E. et al. Potential roles of gut microbiome and metabolites in modulating ALS in mice. Nature 572, 474–480 (2019).

184.      Obeid, R. et al. Plasma trimethylamine-N-oxide following supplementation with vitamin D or D plus B vitamins. Mol. Nutr. Food Res. 61, (2017).

185.      Kasubuchi, M., Hasegawa, S., Hiramatsu, T., Ichimura, A. & Kimura, I. Dietary Gut Microbial Metabolites, Short-chain Fatty Acids, and Host Metabolic Regulation. Nutrients 7, 2839–2849 (2015).

186.      Sukkar, A. H., Lett, A. M., Frost, G. & Chambers, E. S. Regulation of energy expenditure and substrate oxidation by short-chain fatty acids. J. Endocrinol. 242, R1–R8 (2019).

187.      Canfora, E. E. et al. Colonic infusions of short-chain fatty acid mixtures promote energy metabolism in overweight/obese men: a randomized crossover trial. Sci. Rep. 7, 2360 (2017).

188.      Endo, H., Niioka, M., Kobayashi, N., Tanaka, M. & Watanabe, T. Butyrate-producing probiotics reduce nonalcoholic fatty liver disease progression in rats: new insight into the probiotics for the gut-liver axis. PLoS One 8, e63388 (2013).

189.      González-Bosch, C., Boorman, E., Zunszain, P. A. & Mann, G. E. Short-chain fatty acids as modulators of redox signaling in health and disease. Redox Biol. 47, 102165 (2021).

190.      Zhang, H. et al. Gut Microbiota Mediates the Susceptibility of Mice to Sepsis-Associated Encephalopathy by Butyric Acid. J. Inflamm. Res. 15, 2103–2119 (2022).

191.      Chen, X. et al. Sodium butyrate regulates Th17/Treg cell balance to ameliorate uveitis via the Nrf2/HO-1 pathway. Biochem. Pharmacol. 142, 111–119 (2017).

192.      Kim, S. Y. et al. Sodium butyrate inhibits high cholesterol-induced neuronal amyloidogenesis by modulating NRF2 stabilization-mediated ROS levels: involvement of NOX2 and SOD1. Cell Death Dis. 11, 469 (2020).

193.      Braniste, V. et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 6, 263ra158 (2014).

194.      Hoyles, L. et al. Microbiome-host systems interactions: protective effects of propionate upon the blood-brain barrier. Microbiome 6, 55 (2018).

195.      Soto-Martin, E. C. et al. Vitamin Biosynthesis by Human Gut Butyrate-Producing Bacteria and Cross-Feeding in Synthetic Microbial Communities. MBio 11, (2020).

196.      Kinoshita, M. et al. Dietary folic acid promotes survival of Foxp3+ regulatory T cells in the colon. J. Immunol. 189, 2869–78 (2012).

197.      Mölzer, C., Wilson, H. M., Kuffova, L. & Forrester, J. V. A Role for Folate in Microbiome-Linked Control of Autoimmunity. J. Immunol. Res. 2021, 9998200 (2021).

198.      Chriett, S. et al. Prominent action of butyrate over β-hydroxybutyrate as histone deacetylase inhibitor, transcriptional modulator and anti-inflammatory molecule. Sci. Rep. 9, 742 (2019).

199.      Goldberg, E. L. et al. β-Hydroxybutyrate Deactivates Neutrophil NLRP3 Inflammasome to Relieve Gout Flares. Cell Rep. 18, 2077–2087 (2017).

200.      Robles-Vera, I. et al. Protective Effects of Short-Chain Fatty Acids on Endothelial Dysfunction Induced by Angiotensin II. Front. Physiol. 11, 277 (2020).

201.      Kimura, I. et al. Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41). Proc. Natl. Acad. Sci. U. S. A. 108, 8030–5 (2011).

202.      Tao, J. et al. Ketogenic Diet Suppressed T-Regulatory Cells and Promoted Cardiac Fibrosis via Reducing Mitochondria-Associated Membranes and Inhibiting Mitochondrial Function. Oxid. Med. Cell. Longev. 2021, 5512322 (2021).

203.      Li, Y. et al. Ketogenic Diets Induced Glucose Intolerance and Lipid Accumulation in Mice with Alterations in Gut Microbiota and Metabolites. MBio 12, (2021).

204.      Goldberg, E. L. et al. Ketogenesis activates metabolically protective γδ T cells in visceral adipose tissue. Nat. Metab. 2, 50–61 (2020).

205.      González-Sarrías, A., Espín, J. C. & Tomás-Barberán, F. A. Non-extractable polyphenols produce gut microbiota metabolites that persist in circulation and show anti-inflammatory and free radical-scavenging effects. Trends Food Sci. Technol. 69, 281–288 (2017).

206.      Kay, C. D., Pereira-Caro, G., Ludwig, I. A., Clifford, M. N. & Crozier, A. Anthocyanins and Flavanones Are More Bioavailable than Previously Perceived: A Review of Recent Evidence. Annu. Rev. Food Sci. Technol. 8, 155–180 (2017).

207.      Gasperotti, M. et al. Fate of microbial metabolites of dietary polyphenols in rats: is the brain their target destination? ACS Chem. Neurosci. 6, 1341–52 (2015).

208.      Liu, C., Boeren, S., Miro Estruch, I. & Rietjens, I. M. C. M. The Gut Microbial Metabolite Pyrogallol Is a More Potent Inducer of Nrf2-Associated Gene Expression Than Its Parent Compound Green Tea (-)-Epigallocatechin Gallate. Nutrients 14, (2022).

209.      Gao, Z. et al. Urolithin A protects against acetaminophen-induced liver injury in mice via sustained activation of Nrf2. Int. J. Biol. Sci. 18, 2146–2162 (2022).

210.      Chen, P. et al. Gut bacterial metabolite Urolithin A inhibits myocardial fibrosis through activation of Nrf2 pathway in vitro and in vivo. Mol. Med. 28, 19 (2022).

211.      Lee, G., Park, J.-S., Lee, E.-J., Ahn, J.-H. & Kim, H.-S. Anti-inflammatory and antioxidant mechanisms of urolithin B in activated microglia. Phytomedicine 55, 50–57 (2019).

212.      Xu, Z. et al. Urolithin A ameliorates diabetic retinopathy via activation of the Nrf2/HO-1 pathway. Endocr. J. (2022). doi:10.1507/endocrj.EJ21-0490

213.      Del Bo’, C. et al. A polyphenol-rich dietary pattern improves intestinal permeability, evaluated as serum zonulin levels, in older subjects: The MaPLE randomised controlled trial. Clin. Nutr. 40, 3006–3018 (2021).

214.      Peron, G. et al. Crosstalk among intestinal barrier, gut microbiota and serum metabolome after a polyphenol-rich diet in older subjects with ‘leaky gut’: The MaPLE trial. Clin. Nutr. 40, 5288–5297 (2021).

215.      Zhao, Y. & Jiang, Q. Roles of the Polyphenol-Gut Microbiota Interaction in Alleviating Colitis and Preventing Colitis-Associated Colorectal Cancer. Adv. Nutr. 12, 546–565 (2021).

216.      Wu, Z. et al. Gut microbiota from green tea polyphenol-dosed mice improves intestinal epithelial homeostasis and ameliorates experimental colitis. Microbiome 9, 184 (2021).

217.      Shi, T. et al. Quercetin improves gut dysbiosis in antibiotic-treated mice. Food Funct. 11, 8003–8013 (2020).

218.      Ma, Y. et al. Resveratrol alleviates temporomandibular joint inflammatory pain by recovering disturbed gut microbiota. Brain. Behav. Immun. 87, 455–464 (2020).

219.      de Mello, V. D. et al. Indolepropionic acid and novel lipid metabolites are associated with a lower risk of type 2 diabetes in the Finnish Diabetes Prevention Study. Sci. Rep. 7, 46337 (2017).

220.      Koay, Y. C. et al. Ingestion of resistant starch by mice markedly increases microbiome-derived metabolites. FASEB J. 33, 8033–8042 (2019).

221.      Peron, G. et al. A Polyphenol-Rich Diet Increases the Gut Microbiota Metabolite Indole 3-Propionic Acid in Older Adults with Preserved Kidney Function. Mol. Nutr. Food Res. e2100349 (2022). doi:10.1002/mnfr.202100349

222.      Yang, J. et al. Pomegranate Metabolites Impact Tryptophan Metabolism in Humans and Mice. Curr. Dev. Nutr. 4, nzaa165 (2020).

223.      Lundberg, J. O., Weitzberg, E. & Gladwin, M. T. The nitrate-nitrite-nitric oxide pathway in physiology and therapeutics. Nat. Rev. Drug Discov. 7, 156–67 (2008).

224.      Cutler, C. et al. Post-exercise hypotension and skeletal muscle oxygenation is regulated by nitrate-reducing activity of oral bacteria. Free Radic. Biol. Med. 143, 252–259 (2019).

225.      Vanhatalo, A. et al. Nitrate-responsive oral microbiome modulates nitric oxide homeostasis and blood pressure in humans. Free Radic. Biol. Med. 124, 21–30 (2018).

226.      Bahadoran, Z., Mirmiran, P., Kabir, A., Azizi, F. & Ghasemi, A. The Nitrate-Independent Blood Pressure-Lowering Effect of Beetroot Juice: A Systematic Review and Meta-Analysis. Adv. Nutr. 8, 830–838 (2017).

227.      Rocha, B. S., Nunes, C., Pereira, C., Barbosa, R. M. & Laranjinha, J. A shortcut to wide-ranging biological actions of dietary polyphenols: modulation of the nitrate-nitrite-nitric oxide pathway in the gut. Food Funct. 5, 1646–52 (2014).

228.      Zhang, Y., Xu, J. & Yang, H. Hydrogen: An Endogenous Regulator of Liver Homeostasis. Front. Pharmacol. 11, 1–8 (2020).

229.      Ishida, Y., Hino, S., Morita, T., Ikeda, S. & Nishimura, N. Hydrogen produced in rat colon improves in vivo reduction-oxidation balance due to induced regeneration of α-tocopherol. Br. J. Nutr. 123, 537–544 (2020).

230.      Chaoqun, L., Yuqi, Z., Shi, Z., Zhenghui, Y. & Li, W. A Comparison of the Antioxidant Effects Between Hydrogen Gas Inhalation and Vitamin C Supplementation in Response to a 60-Min Treadmill Exercise in Rat Gastrocnemius Muscle. Front. Physiol. 12, 745194 (2021).

231.      Shen, X. et al. Microbial regulation of host hydrogen sulfide bioavailability and metabolism. Free Radic. Biol. Med. 60, 195–200 (2013).

232.      Hourihan, J. M., Kenna, J. G. & Hayes, J. D. The gasotransmitter hydrogen sulfide induces nrf2-target genes by inactivating the keap1 ubiquitin ligase substrate adaptor through formation of a disulfide bond between cys-226 and cys-613. Antioxid. Redox Signal. 19, 465–81 (2013).

233.      Ghosh, S. et al. Elevated muscle TLR4 expression and metabolic endotoxemia in human aging. J. Gerontol. A. Biol. Sci. Med. Sci. 70, 232–46 (2015).

234.      Kavanagh, K. et al. Biomarkers of leaky gut are related to inflammation and reduced physical function in older adults with cardiometabolic disease and mobility limitations. GeroScience 41, 923–933 (2019).

235.      Stehle, J. R. et al. Lipopolysaccharide-binding protein, a surrogate marker of microbial translocation, is associated with physical function in healthy older adults. J. Gerontol. A. Biol. Sci. Med. Sci. 67, 1212–8 (2012).

236.      Zhan, X., Stamova, B. & Sharp, F. R. Lipopolysaccharide Associates with Amyloid Plaques, Neurons and Oligodendrocytes in Alzheimer’s Disease Brain: A Review. Front. Aging Neurosci. 10, 42 (2018).

237.      Loffredo, L. et al. Oxidative Stress and Gut-Derived Lipopolysaccharides in Neurodegenerative Disease: Role of NOX2. Oxid. Med. Cell. Longev. 2020, 8630275 (2020).

238.      West,  a P. et al. TLR signalling augments macrophage bactericidal activity through mitochondrial ROS. Nature 472, 476–80 (2011).

239.      Wu, M.-L., Yang, X.-Q., Xue, L., Duan, W. & Du, J.-R. Age-related cognitive decline is associated with microbiota-gut-brain axis disorders and neuroinflammation in mice. Behav. Brain Res. 402, 113125 (2021).

240.      Matt, S. M. et al. Butyrate and Dietary Soluble Fiber Improve Neuroinflammation Associated With Aging in Mice. Front. Immunol. 9, 1832 (2018).

241.      Wang, H. et al. Gut microbiota is causally associated with poststroke cognitive impairment through lipopolysaccharide and butyrate. J. Neuroinflammation 19, 76 (2022).

242.      Allen, J. M. et al. Psychological stress disrupts intestinal epithelial cell function and mucosal integrity through microbe and host-directed processes. Gut Microbes 14, 2035661 (2022).

 

No comments:

Post a Comment