Short Chain Fatty Acid Metabolism in Relation to Gut Microbiota and Genetic Variability
Abstract
:1. Introduction
2. Dietary Fibre and Short Chain Fatty Acids
2.1. Dietary Fibre (DF)
2.2. Short Chain Fatty Acids (SCFA)
SCFA | Study (Sample) | Study Design | Tissues Investigated | End-Point Measured | Observed Effects | Reference |
---|---|---|---|---|---|---|
Human interventional studies | ||||||
C2 | H (n =32) | Case-control | Peripheral blood | Immunopharmacological effects of Ringer’s acetate | Increased polyclonal antibody production and NK cell activity in healthy and cancer subjects | [141] |
C3 | H (n = 6) | Cross-over | Serum and stool | Blood lipids and glucose, stool bulk and microbiota | C3 supplementation lowers blood glucose. Lipid changes not significant; increase in stool bulk and Bifidobacteria after 1 week intervention | [142] |
C4 | H (n = 16) | Cross-over | Sigmoid colon biopsies and plasma | Oxidative stress markers in colon; CRP, calprotectin; histological inflammation | Rectal administration significantly reduced uric acid and increased GSH. No significant changes in other parameters | [143] |
Human Observational studies | ||||||
C2-C6 | H (n = 232) | Observation | Stool | Levels of faecal SCFA and BCFA association with BMI and age | BCFA strongly correlated with age, but not with BMI; BCFA negatively associated with fibre consumption; BMI ≥ 40 showed significantly higher production of SCFA, total BCFA, isobutyrate, isovalerate and caproate SCFA production decreases with age | [131] |
Animal (interventional) studies | ||||||
C2, C3 | M (n = 15) | Knock-out | Adipose tissue | Effects of GPCR43 activation | Reduction of lipolysis, reduced plasma free fatty acids levels without flushing associated with GPCR109A | [144] |
C2, C3 | M (n = 12) | Case-control | Adipose, gut, vascular and mesenchymal tissues | GPCR41 and GPCR43 mRNA expression | GPCR43 activation promoted adipose differentiation via PPARγ2. No effects on GPCR41 | [145] |
C2, C3, C4 | S (n = 10) | Case-control | Portal and peripheral blood, liver | Food intake following SCFA infusions | Dose-dependent depression in food intake, explained by C3 content in portal vein, which resolved with portal plexus denervation | [146] |
C3 | R (n = 20) P (n = 12, 60) | Case-control | Portal blood and liver | Cholesterol synthesis and distribution | Supplemented C3 likely absorbed in the stomach Dose-dependent hypocholesterolemic effect likely due to redistribution of cholesterol from plasma to liver, as opposed to synthesis inhibition | [147,148] |
C3 | R (n = 74, 114) | Case-control | Brain, intracerebral ventricles | Behavioural, electrophysiological, neuropathological, and biochemical effects | C3 intraventricular infusion impaired social behaviours, similar to those seen in human ASD; induced neuroinflammation and oxidative stress; Alteration of brain phospholipid and acylcarnitine1 profiles | [149,150] |
C4 | R (n = 22) | Case-control | Duodenum, jejunum, cecum and distal colon | PYY and proglucagon gene expression in gut epithelial cells | Up-regulation of local peptide YY and proglucagon expression via colonocyte sensing following a RS diet in vivo, proved by in vitro incubation with butyrate | [151] |
C4 | M (n = 16–20) | Case-control | Whole-body autopsy | Insulin sensitivity and energy metabolism, mitochondrial function | C4 supplementation prevented diet-induced insulin resistance and reduced adiposity in high-fat model, without reducing food intake. Attributed to enhanced mitochondrial activity and thermogenesis | [152] |
In Vitro Studies | ||||||
C2-C6 | M (n = 18) | N/A | mouse adipocyte cell line and adipose primary culture | Leptin expression | C2-C6 stimulate leptin expression via GPCR41 Acute administration of C3 increased leptin levels | [153] |
C2, C4 | R, B | N/A | Anterior pituitary, fat and liver aspirates | Leptin and leptin-receptor protein expression | C2 and C4 enhanced leptin expression in bovine pituitary and fat cells, however C4 inhibited leptin expression in rat anterior pituitary cells; while C4 suppressed leptin receptor expression in both rat and bovine pituitaries; probable species specific nutrient sensing | [154] |
C2, C3, C4 | R, H | N/A | Colonic stimulation | Effects on colon functions, inc. motility | C3 and C4 induced phasic and tonic contractions of circular muscle via GPCR41 and GPCR43 in mucosae, C2 did not | [155] |
C2, C3, C4 | M (n= 4) H (n= 3) | N/A | Human blood samples, colon cultures (colo320DM) and mice with colitis | Anti-inflammatory properties of SCFA | All SCFA decreased neutrophil TNF-α release without affecting IL-8; all decreased IL-6 release; all inhibited NF-κB activity in colon cells; C4 > C3 > C2 | [156] |
C3 | H (n = 5–9) | N/A | Human umbilical vein endothelial cells (HUVEC) | Expression of endothelial leukocyte adhesion molecules and leukocyte recruitment by cytokine-stimulation | Significant inhibition of TNF-α and NF-κB, reducing expression of VCAM-1 and ICAM-1 in a time- and dose-dependent manner; significantly increased PPARα expression | [157] |
C3 | H (n = 28) | N/A | Omental and subcutaneous adipose tissue | Adipokine expression | Significant leptin induction and secretion; no effect on adiponectin; Reduction of resistin mRNA expression | [158] |
C3 | R, H (n = 1) | N/A | Human blood and rat mesenteric lymph nodes | T and B lymphocyte proliferation and metabolism | Inhibition of lipid synthesis as a possible mechanism leading to reduction of lymphocyte proliferation | [159] |
C3 | R (n = 9) | N/A | Isolated hepatocytes | Hepatic lipidogenesis | Inhibits hepatic cholesterol and fatty acid synthesis in a dose-dependent manner, possibly by competition with C2 | [160] |
3. Inter-Individual Variability
3.1. Enterotypes—SCFA Production and Relation to Disease
- Enterotype 1, or ET-B, presenting Bacteroides as the taxon driver;
- Enterotype 2, or ET-P has Prevotella genus as common denominator—abundance of Prevotella is inversely correlated with Bacteroides;
- Enterotype 3, or ET-F is characterized by an abundance of Firmicutes, namely Ruminococcus.
SCFA(s) | Bacterial Genera (Phylum) | Representative Bacterial Species | Observed Effects | References |
---|---|---|---|---|
Butyrate | Clostridiales cluster I-II (F) | Clostridium histolyticum | Identified as a potential tumour regression therapy (via collagenase production) as well as being associated with gas gangrene in diverticular disease and trauma (via exotoxin) | [199,200] |
Clostridiales XIV, Ruminoccacea (F) | R. bromii | Taxon driver of enterotype 3; Believed to be the main resistant starch fermenter into butyrate, was significantly increased following RS diet in men with obesity | [168,201,202] | |
Clostridiales XIV (F) | Clostridium symbiosum | A SCFA producer, was shown to improve post stroke disability in aged mice | [203] | |
Clostridiales IV, Lachnospiraceae (F) | Roseburia intestinalis Butyrivibrio fibrisolvens | Can rescue intestinal epithelium autophagy and mitochondrial respiration insufficiency, are associated with reduced colorectal cancer; Lachnospiraceae phylotypes increased on an NSP diet with strong cross-feeding interactions | [73,79,202,204] | |
Clostridiales IV (F) | F. prausnitzii | Produce butyrate in 1 step reaction; Influences Muc2 and goblet cell differentiation; depleted in IBD and Crohn’s disease | [52,205,206] | |
Eubacteriae (F) | E. rectale, E. hallii, E. ventriosum | Together with F. prausnitzii, are the major butyrate producers; growth is promoted by low colonic pH, which also inhibits pH-sensitive pathogenic bacteria | [207,208] | |
Propionibacteria (F) | P. acidipropionici | Propionate producer, induces colorectal cancer apoptosis through mitochondrial adenine nucleotide translocator (ANT) | [57,63,209,210] | |
Bacteroides (B) | B. thetaiotaumicron | Driver of enterotype 1; is a mucus-forager with lack of DF B. thetaiotaumicron regenerates NAD+; reduced S-BCAA and alleviated diet-induced weight-gain and obesity in mice. Influences Muc2 and goblet cell differentiation. Produces butyrate via the succinate pathway | [168,196] | |
Propionate | Negativicutes (F) | N. succinicivorans | Produce propionate via succinate pathway | [211,212] |
Veillonellaceae (F) | V. parvula | Produce propionate via acrylate pathway (lactate) and/or acetate. Have been associated with osteomyelitis, hypertension and endocarditis | [211,213,214] | |
Lachnospiraceae (F) | Blautia hydrogenotrophica | Produce propionate via acrylate pathway (lactate) and propanodiol pathways (deoxi-sugars) | [211,213,215,216] | |
Christensenellaceae (F) | C. minuta | Regarded as the most heritable taxon, forming the hub of a co-occurrence network composed of other heritable taxa; is enriched in lean subjects; in mice, reduced adiposity gain in GF model | [217,218,219,220,221] | |
Bacteroides (B) | B. fragilis B. ovatus | Ferment xyloglucans, C3 directly inhibited Salmonella overgrowth by pH modulation in vitro. Bacteroidetes relative abundance has been linked to faecal propionate concentration. Decreased in ASD; in contrast, C3 administration led to ASD behaviour in rodent models via altered mitochondrial metabolism | [218,219] | |
Acetate | Prevotella (B) | P. intestinalis | Driver of Enterotype 2; significant high prevalence of Prevotella in healthy African Americans 50–65 y, while decreased in Western populations. P. intestinalis administration in mice led to reductions of overall SCFA production and increased mucosal inflammation which abated with IL-18 supplementation | [70,168,222,223,224,225,226,227] |
Methanobrevibacter (F) | Methanobrevibacter smithii | Found to be highly inherited, methanogens are inconclusively associated with increased BMI and reduced transit time in humans, as well as with leanness in mice. Metabolizers of formate, which can result in decreased blood pressure. Co-culture with R. intestinalis and B. hydrogenotrophica decreased H2 and produced CH4 and acetate, reducing pH | [196,202,204,217,218,219,220,221,228,229] | |
Bifidobacterium (A) | B. adolescentis | FOS, GOS fermenter. High inheritability LF diet with prebiotic supp. increased Bifidobacteria abundance, which ameliorates the allergic phenotype and inhibited the growth of enteropathogenic bacteria. Bifidobacteria seems to be reduced in obese-derived faecal cultures as well as in ASD; Significantly decreased with a weight-loss diet given to men with obesity | [70,196,202,220,224,225,226,227,228] | |
Lactobacillus (B) | L. johnsonii | Lactobacillus is a lactate producer commonly found in the upper gastrointestinal tract. FOS, GOS fermenter. May protect against diet-induced obesity and reduce asthma incidence in children. However, is increased in ASD. Probiotic supplementation impact revealed to be dependent on basal microbiota between individuals with obesity and normal weight | [70,168,220,222,223,224,225,226,228] |
3.2. Genotypes—Interactions with Gut Microbiota
Metabolic Step (Tissue) | Gene (Protein) | SNP/CNV * | Observed Statistically Significant Association from GWAS | References |
---|---|---|---|---|
Digestion enzyme (gut lumen) | AMY1/2 | CNV rs370981115 | Impacts oral and gut microbiome due to bioavailability of starches; altered blood protein measurements | [251,252] |
LCT | rs4988235, rs1446585, rs2322659, rs35837297 | Lactase persistence allows for dairy product consumption in adult life and increased expression of Bifidobacterium in the gut; altered lung function and leukocyte counts | [253,254,255] | |
Barrier function (colon) | MUC2 | rs4077759, rs10794281, rs35225972 | Modulated by butyrate. Variations associated with decrease gastric cancer progression, enhanced gastric lesion regression, asthma | [61,256,257,258,259] |
FUT2 | rs516246, rs601338, rs679574 | Mucus fucosylation status. Predisposition to Crohn’s disease and dysbiosis; altered blood protein measurements | [252,258,260] | |
Antimicrobial peptides (gut) | DEFA5 | CNV rs2272719 | α-defensins modulate microbial populations; copy number gain identified as pathogenic; altered white blood cell counts; susceptibility to paediatric leukaemia | [249,261] |
MMP7 | rs11568818 | Involved in antimicrobial processes; prostate cancer | ||
SCFA receptor | MCT1 (SLC16A1) | rs147836155 rs4839270 rs773430 | SCFA uptake; variations have been associated to exercise-induced hyperinsulinemia (EIHI); microglial activation, refractive errors of the eye, blood pressure disorders | [262,263,264,265] |
MCT2 (SLC16A7) | rs79297227 | SCFA uptake (hepatocytes); BMI trajectories, development of non-small cell lung carcinoma | [266] | |
MCT3 (SLC16A8) | rs1004763 | Cerebral white matter microstructure; cognitive function | [267] | |
MCT4 (SLC16A3) | rs4239020 | Adipose tissue distribution, BMI | [268] | |
MCT11 (SLC16A11) | rs13342232 | Associated with the risk of paediatric-onset T2D in Mexican families | [269] | |
MCT9 (SLC16A9) | rs7094971 | Carnitine transporter, associated with reversible ASD and mitochondrial abnormalities | [221,270] | |
SMCT1 (SLC5A8) | rs7296340 rs141751904 | SCFA uptake by colonocytes; in absence of microbiota, marked down-regulation of SLC5A8, which acts as a tumour suppressor protein in the presence of butyrate; variation decreases BMI-adjusted waist-hip ratio; decreased IL-2 levels | [271,272,273] | |
SMCT2 (SLC5A12) | rs10835056 | SCFA uptake; decreased MIP-1α levels | [273] | |
Metabolism | GPCR109A (HCAR2) | rs56959712 | Butyrate receptor in enterocytes and MALT, regulating dendritic cell and Treg diff, also present in microglia. Ligand niacin is used to treat dyslipidaemia; variant associate with blood lipid measurements | [274,275] |
GPCR43 (FFFAR2) | rs34536858 | Acetate and propionate receptor, leading to NLRP3 assembly. Regulation of Treg population in colon, ROS production and neutrophil chemotaxis. KO models showed increased arthritis, colitis and allergic disease; regulates adipogenesis and GLP-1 release; associated white and blood cell variance | [72,276] | |
GPCR41 (FFAR3) | rs10407548 | Regulation of SCFA-dependent energy homeostasis. Activation by propionate, butyrate and valerate results in inhibition of NF-κB activation; induce chemokine and cytokine expression; associated with gastrointestinal motility and stool frequency | [277,278] | |
GPCR42 | CNV | Recently reclassified as functioning gene; Propionate affinity; polymorphisms associated with strong pharmacokinetic variation | [279] | |
Metabolism (systemic) | LEP | CNV, rs7799039, rs17151919 | 40–70% estimated heritability for BMI; SNPs associated with CVD and MetS, increased HbA1c, insulin and increased fat mass, among other clinical phenomes. KO mice had higher susceptibility to dysbiosis | [280,281] |
LEPR | CNV, rs1137101, rs9436747 | Same as above, variations associate with blood lipids, proteins, cytokines and cell counts | [280,282,283,284] | |
PLD1 | rs4894707 | Associated with obesity, insulin sensitivity and abundance levels of Akkermansia muciniphila | [285] |
3.3. Phenotypes, Epigenetic Aspects of SCFA
Condition(s) | Increased Bacteria | Decreased Bacteria | Opportunistic spp. or Additional Findings | References |
---|---|---|---|---|
Obesity | ↑Firmicutes:Bacteroidetes ratio, Blautia, Dorea, Proteobacteria, Tenericutes | Akkermansia, F. praustnizii, B. thetaiotaumicron | Ratio seems to be higher in women with ↑BMI Diversity and richness is crucial for responding or not to dietary intervention aiming at improving metabolic parameters (insulin sensitivity, lipid and inflammation markers); increased propionate production compared to normal weight microbiota | [196,202,306,307,308] |
Metabolic Syndrome | ↑Firmicutes:Bacteroidetes ratio, Blautia, Dorea, Methanobacteriaceae | Oscillospira, Rikenellaceae, Bifidobacterium, Christensenellaceae, Akkermansia, Lactobacillus | BCFA are associated with obesity, insulin resistance and development of T2D; Bacteroides spp. may improve the efficiency of BCFA degradation Ass. With ↑faecal SCFA, plasma BCFA, plasma TMAO, plasma total bile acids and plasma LPS. MetS and NAFLD seem to occur via intestinal FXR | [196,309,310] |
Gestational diabetes | Collinsella, Rothia, Desulfovibrio, Faecalibacterium, Anaerotruncus | Clostridium, Veillonella, Akkermansia, Christensenella | Similar findings with obesity enterotype, may remain postpartum P. copri and B. vulgatus identified as the main species leading the biosynthesis of BCFAs and insulin resistance; prebiotic supp. increased Bifidobacteria and led to reduction of faecal SCFA and serum fasting glucose and insulin | [196,306,311] |
T2D | ↑Firmicutes:Bacteroidetes, Dorea, Escherichia, Clostridiales, Lactobacillus | Overall diversity reduced; R. intestinalis, Akkermansia, Streptococcus, Bifidobacteria, F. prausnitzii | Similar findings with obesity and MetS enterotypes, although some studies find ↑Bacteroidetes:Firmicutes ratio. Opportunistic infections with B. caccae, C. hathewayi, C. ramosum, C. symbiosum, E. lenta and E. coli. Butyrate is beneficial for pancreatic B-cell function, whereas propionate has shown to be detrimental. Metformin therapy increases A. muciniphila | [196,308,312,313] |
T1D | ↑Bacteroidetes:Firmicutes, Synergistetes | Clostridium, Prevotella, Bifidobacterium Lachnospiraceae, Veillonellaceae | Opportunistic overgrowth of Ruminococcus gnavus and Streptococcus infantarius. T1D may be related to delivery method, feeding method and antibiotic use in infancy | [314] |
NAFLD | Lactobacillus, Dorea, Streptococcus, Lachnospiraceae | Ruminococcaceae, Prevotella, Flavobacterium, B. vulgatus | Increased intestinal permeability associated with the degree of steatosis, affects up to 70% of patients with T2D and 90% of obese, possibly due to intestinal inflammation and permeability dysfunction, bile acid metabolism (FXR), anaerobic fermentation, and LPS activation of TLR4 leading to insulin resistance | [196,310,315,316] |
Non-alcoholic Steato hepatitis (NASH) | Bacteroidetes, Prevotella, Escherichia | Firmicutes | Prevotella seems to be reduced in advanced stages of NAFLD, i.e., NASH; the levels of serum LPS and TNF-α correlated with disease severity. Synbiotic supp. of B. longum and FOS reduced disease severity of NAFLD and NASH progression | [196] |
Alcoholic Steatohepatitis | E. faecalis, E. coli, Proteobacteria | Bacteroidaceae, Ruminococcaceae, Firmicutes | Only 40% of patients had dysbiosis. E. faecalis correlated with mortality rates in alcohol-induced steatohepatitis; supp. with B. subtilis and E. faecium improved symptoms and microbiome | [317] |
IBD | Proteobacteria | Firmicutes, esp F. prausnitzii; Bacteroides; Clostridium; Peptostreptococcus; Bifidobacterium | Increase in fungal Candida albicans, Aspergillus clavatus, and Cryptococcus neoformans, decreased Saccharomyces cerevisiae. IBD can arise from genetic susceptibility or from disruption of commensal bacteria such as SCFA-producing bacteria, reduction in tryptophan metabolism (promoting mucus barrier function and reduces inflammatory responses), Proteobacteria may represent 20% of overall diversity | [35,258] |
Colorectal cancer | S. bovis, H. Pylori, E. faecalis, E. coli, B. fragilis, F. fucleatum, C. septicum, Fusobacteria, Proteobacteria, Akkermansia | Bifidobacteria, Lactobacilli, Bacteroidetes, Firmicutes, F. prausnitzii, Prevotella, Porphyromonas | S. bovis is increased in neoplastic milieu and may forage tumour metabolites, inducing inflammation. Some bacterial strains may propel CRC development, while others are only found in late stages of CRC, arising as opportunistic pathogens, which may deplete symbionts by substrate competition and lead to tumour survival by immune evasion mechanisms. | [16,50,52,57,210,318,319] |
Psoriatic arthritis | N.A. | Coprococcus, Akkermansia, Ruminococcus, Pseudobutyrivibrio. | Overall reduced microbial diversity, similar to IBD and other autoimmune phenotypes such as skin psoriasis; however, Akkermansia and Ruminococcus were uniquely decreased in psoriatic arthritis. Rheumatoid arthritis presents with increased P. copri | [320] |
Atopy, inc. food allergy, atopic dermatitis and asthma | ↑Firmicutes:Bacteroidetes, C. difficile, Enterobactericeae, E. coli | Bifidobacteria, Lactobacilli, Clostridia, Bacteroides, Actinobacteria, Proteobacteria | Supp. L. rhamnosus GG and L. fermentum to mothers in the prenatal and early postnatal periods or to young children may be effective in reducing symptoms, treatment and prevention of early atopic disease in offspring | [321,322,323] |
Autism Spectrum Disorders | Clostridium, Bacteroidetes, Lactobacillus, Caloramator, Sarcina, Propionibacteria, Desulfovibrio | Bifidobacterium, Prevotella, Firmicutes, Akkermansia | Increased production of propionate due to dysbiosis may be a cause of reversible ASD, also leading to GI symptoms in a majority of cases, which ameliorated by supp. strains of Bifidobacteria and Lactobacilli. Children with ASD show increased levels of opportunistic Candida albicans | [324] |
Cardiovascular Disease (CVD) inc. Atherosclerosis and Hypertension | ↑Firmicutes:Bacteroidetes, Enterobacteriaceae, Clostridia (C. histolyticum, C. perfringens, E. timonensis), Atopobium, Prevotella | microbial richness, diversity and evenness significantly decreased, Odoribacter, Bacteroides | S-TMAO (microbial-derived choline metabolite) levels were dose-dependent associated with CVD outcomes and other indicators such as serum cholesterol, glycaemic indices (HbA1c, fasting plasma glucose), inflammation biomarkers (IL-6, CRP), overall cardiovascular risk, and metabolic syndrome. | [90,196,325,326] |
Odoribacter is a butyrate-producer negatively correlated with systolic blood pressure, like other SCFA producers, although SCFAs increase vascular tone | [308,325,327,328,329,330] | |||
Parkinson Disease | Bifidobacterium, Pasteurella, Enterococcus, Lactobacillus, Verrucomicrobia (A. muciniphila), Bilophila, Christensenella, Dorea, Barnesiellaceae, Tissirellaceae, Ralstonia, Pasteurellaceae. Escherichia, Bacteroidetes | Firmicutes, Brautella, Prevotella, Faecococcus Lachnospiraceae, Paraprevotella, Faecalibacterium, Roseburia, Blautia, C. coccoides, B. fragilis | Paraprevotella mainly decreased in females; Bilophila abundance associated with disease severity; Blautia associated with disease onset/duration; neurotransmitters such as serotonin, dopamine and GABA are produced by microbiota; E. coli producing amyloid protein Curli cross-seeds with α-synuclein and stimulates protein aggregation in gut (present in 65–85% of cases), with gut-to-brain transport demonstrated. Microbial sulphur metabolism is profoundly changed in PD, mainly associated with A. muciniphila and B. wadsworthia | [11,12,31,331,332,333,334,335,336,337,338,339] |
Alzheimer’s Disease (AD) | ↓Firmicutes:Bacteroidetes, E. coli, Shigella, Helicobacter, Odoribacter | Bifidobacteria, Lactobacillus, Firmicutes, Actinobacteria, Verrucomicrobia, Roseburia, Eubacterium, F. prausnitzii | Similar dysbiosis in MCI as in AD; amyloid protein Curli produced by E. coli and S. typhimurium enhances colonization and biofilm development; E. rectale and Shigella taxon in the faecal samples of patients with advanced AD correlated well to the amyloidosis and level of proinflammatory cytokines in the brain. TMAO induced synaptic impairment in AD model with deposition of Aβ plaques and neurofibrillary tangles. Aβ plaques found in gut vessels prior to disease onset, accompanied with systemic inflammation | [11,12,14,90,334,339,340,341,342] |
4. The Holobiont and Short Chain Fatty Acids
4.1. Host-Microbe Interface
4.2. Digestive Enzymes
4.3. Genetic Diversity and Physical Barriers
4.3.1. Mucin
4.3.2. Tight Junction Proteins (TJPs)
4.3.3. Immune Cell Populations in the Gut
4.3.4. Transporter Genetics
4.4. SCFA Metabolism
4.4.1. Colon
4.4.2. Liver and Adipose Tissue
4.4.3. Systemic Metabolism
4.5. Signalling Pathways of Interest
5. Conclusions and Perspectives
5.1. Main Conclusions
5.2. Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Sender, R.; Fuchs, S.; Milo, R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016, 14, e1002533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zilber-Rosenberg, I.; Rosenberg, E. Role of microorganisms in the evolution of animals and plants: The hologenome theory of evolution. FEMS Microbiol. Rev. 2008, 32, 723–735. [Google Scholar] [CrossRef] [PubMed]
- Goodrich, J.K.; Davenport, E.R.; Clark, A.G.; Ley, R.E. The Relationship Between the Human Genome and Microbiome Comes into View. Annu. Rev. Genet. 2017, 51, 413–433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Donaldson, G.P.; Lee, S.M.; Mazmanian, S.K. Gut biogeography of the bacterial microbiota. Nat. Rev. Microbiol. 2016, 14, 20–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sommer, F.; Bäckhed, F. The gut microbiota—Masters of host development and physiology. Nat. Rev. Microbiol. 2013, 11, 227–238. [Google Scholar] [CrossRef]
- Perry, R.J.; Peng, L.; Barry, N.A.; Cline, G.W.; Zhang, D.; Cardone, R.L.; Petersen, K.F.; Kibbey, R.G.; Goodman, A.L.; Shulman, G.I. Acetate mediates a microbiome-brain-β cell axis promoting metabolic syndrome. Nature 2016, 7606, 213–217. [Google Scholar] [CrossRef] [Green Version]
- McRae, M.P. Dietary Fiber is Beneficial for the Prevention of Cardiovascular Disease: An Umbrella Review of Meta-analyses. J. Chiropr. Med. 2017, 16, 289–299. [Google Scholar] [CrossRef]
- Akshintala, V.S.; Talukdar, R.; Singh, V.K.; Goggins, M. The Gut Microbiome in Pancreatic Disease. Clin. Gastroenterol. Hepatol. 2018, 17, 290–295. [Google Scholar] [CrossRef]
- Diamanti, A.P.; Rosado, M.M.; Laganà, B.; D’Amelio, R. Microbiota and chronic inflammatory arthritis: An interwoven link. J. Transl. Med. 2016, 14, 233. [Google Scholar] [CrossRef] [Green Version]
- Kang, L.; Li, P.; Wang, D.; Wang, T.; Hao, D.; Qu, X. Alterations in intestinal microbiota diversity, composition, and function in patients with sarcopenia. Nature 2021, 11, 4628. [Google Scholar] [CrossRef]
- Gentile, F.; Doneddu, P.E.; Riva, N.; Nobile-Orazio, E.; Quattrini, A. Diet, Microbiota and Brain Health: Unraveling the Network Intersecting Metabolism and Neurodegeneration. Int. J. Mol. Sci. 2020, 21, 7471. [Google Scholar] [CrossRef]
- Friedland, R.P. Mechanisms of Molecular Mimicry Involving the Microbiota in Neurodegeneration. J. Alzheimer’s Dis. 2015, 45, 349–352. [Google Scholar] [CrossRef] [Green Version]
- Baldini, F.; Hertel, J.; Sandt, E.; Thinnes, C.C.; Neuberger-Castillo, L.; Pavelka, L.; Betsou, F.; Krüger, R.; Thiele, I. Parkinson’s disease-associated alterations of the gut microbiome predict disease relevant changes in metabolic functions. BMC Biol. 2020, 18, 62. [Google Scholar] [CrossRef]
- Goyal, D.; Ali, S.A.; Singh, R.K. Emerging role of gut microbiota in modulation of neuroinflammation and neurodegeneration with emphasis on Alzheimer’s disease. Prog. Neuropsychopharmacol. Biol. Psychiatry 2021, 106, 9. [Google Scholar] [CrossRef]
- Lazar, V.; Ditu, L.-M.; Pircalabioru, G.G.; Gheorghe, I.; Curutiu, C.; Holban, A.M.; Picu, A.; Petcu, L.; Chifiriuc, M.C. Aspects of Gut Microbiota and Immune System Interactions in Infectious Diseases, Immunopathology, and Cancer. Front. Immunol. 2018, 9, 1830. [Google Scholar] [CrossRef] [Green Version]
- Kunzmann, A.T.; Coleman, H.G.; Huang, W.-Y.; Kitahara, C.M.; Cantwell, M.M.; Berndt, S.I. Dietary fiber intake and risk of colorectal cancer and incident and recurrent adenoma in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. Am. J. Clin. Nutr. 2015, 102, 881–890. [Google Scholar] [CrossRef] [Green Version]
- Kotas, M.E.; Medzhitov, R. Homeostasis, Inflammation, and Disease Susceptibility. Cell 2015, 160, 816–827. [Google Scholar] [CrossRef] [Green Version]
- Cevenini, E.; Monti, D.; Franceschi, C. Inflamm-ageing. Curr. Opin. Clin. Nutr. Metab. Care 2013, 16, 14–20. [Google Scholar] [CrossRef]
- Hotamisligli, G.S. Inflammation and metabolic disorders. Nature 2006, 444, 860–867. [Google Scholar] [CrossRef]
- Hotamisligil, G.S. Inflammation, metaflammation and immunometabolic disorders. Nature 2017, 542, 177–185. [Google Scholar] [CrossRef]
- Kaulmann, A.; Bohn, T. Carotenoids, inflammation, and oxidative stress—Implications of cellular signaling pathways and relation to chronic disease prevention. Nutr. Res. Rev. 2014, 34, 907–929. [Google Scholar] [CrossRef] [PubMed]
- Menzel, A.; Samouda, H.; Dohet, F.; Loap, S.; Ellulu, M.S.; Bohn, T. Common and Novel Markers for Measuring Inflammation and Oxidative Stress Ex Vivo in Research and Clinical Practice—Which to Use Regarding Disease Outcomes? Antioxidants 2021, 10, 414. [Google Scholar] [CrossRef] [PubMed]
- Pan, A.; Lin, X.; Hemler, E.; Hu, F.B. Diet and Cardiovascular Disease: Advances and Challenges in Population-Based Studies. Cell Metab. 2018, 27, 489–496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Christ, A.; Latz, E. The Western lifestyle has lasting effects on metaflammation. Nat. Rev. Immunol. 2019, 19, 267–268. [Google Scholar] [CrossRef] [PubMed]
- Devore, E.E.; Kang, J.H.; Breteler, M.M.B.; Grodstein, F. Dietary intake of berries and flavonoids in relation to cognitive decline. Ann. Neurol. 2012, 72, 135–143. [Google Scholar] [CrossRef]
- Darmadi-Blackberry, I.; Wahlqvist, M.L.; Kouris-Blazos, A.; Steen, B.; Lukito, W.; Horie, Y.; Horie, K. Legumes: The most important dietary predictor of survival in older people of different ethnicities. Asia Pac. J. Clin. Nutr. 2004, 13, 217–220. [Google Scholar]
- Katagiri, R.; Goto, A.; Sawada, N.; Yamaji, T.; Iwasaki, M.; Noda, M.; Iso, H.; Tsugane, S. Dietary fiber intake and total and cause-specific mortality: The Japan Public Health Center-based prospective study. Am. J. Clin. Nutr. 2020, 111, 1027–1035. [Google Scholar] [CrossRef]
- Szic, K.S.V.; Declerck, K.; Vidaković, M.; Berghe, W.V. From inflammaging to healthy aging by dietary lifestyle choices: Is epigenetics the key to personalized nutrition? Clin. Epigenetics 2015, 7, 33. [Google Scholar] [CrossRef] [Green Version]
- Fitzgerald, K.C.; Tyry, T.; Salter, A.; Cofield, S.S.; Cutter, G.; Fox, R.; Marrie, R.A. Diet quality is associated with disability and symptom severity in multiple sclerosis. Neurology 2018, 90, e1–e11. [Google Scholar] [CrossRef]
- Yoo, J.Y.; Kim, S.S. Probiotics and Prebiotics: Present Status and Future Perspectives on Metabolic Disorders. Nutrients 2016, 3, 173. [Google Scholar] [CrossRef] [Green Version]
- Mischley, L.K.; Lau, R.C.; Bennett, R.D. Role of Diet and Nutritional Supplements in Parkinson’s Disease Progression. Oxidative Med. Cell. Longev. 2017, 2017, 6405278. [Google Scholar] [CrossRef]
- Lucas, M.; Chocano-Bedoya, P.; Shulze, M.B.; Mirzaei, F.; O’Reilly, É.J.; Okereke, O.I.; Hu, F.B.; Willett, W.C.; Ascherio, A. Inflammatory dietary pattern and risk of depression among women. Brain Behav. Immun. 2014, 36, 46–53. [Google Scholar] [CrossRef] [Green Version]
- Rayman, M.P. Diet, nutrition and osteoarthritis. BMC Musculoskelet. Disord. 2015, 16, S7. [Google Scholar] [CrossRef] [Green Version]
- Martínez-González, M.A.; Sánchez-Villegas, A. Food patterns and the prevention of depression. Proc. Nutr. Soc. 2016, 75, 139–146. [Google Scholar] [CrossRef] [Green Version]
- Gill, S.K.; Rossi, M.; Bajka, B.; Whelan, K. Dietary fibre in gastrointestinal health and disease. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 101–116. [Google Scholar] [CrossRef]
- Trowell, H. Definition of dietary fiber and hypotheses that it is a protective factor in certain diseases. Am. J. Clin. Nutr. 1976, 29, 417–427. [Google Scholar] [CrossRef]
- Wu, Z.; Xu, Q.; Wang, Q.; Chen, Y.; Lv, L.; Zheng, B.; Yan, R.; Jiang, H.; Shen, J.; Wang, S.; et al. The impact of dietary fibers on Clostridioides difficile infection in a mouse model. Front. Cell. Infect. Microbiol. 2022, 12, 1028267. [Google Scholar] [CrossRef]
- Snauwaert, E.; Paglialonga, F.; Vande Walle, J.; Wan, M.; Desloovere, A.; Polderman, N.; Renken-Terhaerdt, J.; Shaw, V.; Shroff, R. The benefits of dietary fiber: The gastrointestinal tract and beyond. Pediatr. Nephrol. 2022, 2022, 1–10. [Google Scholar] [CrossRef]
- Nakaji, S.; Sugawara, K.; Saito, D.; Yoshioka, Y.; Macauley, D.; Bradley, T.; Kernohan, G.; Baxter, D. Trends in dietary fiber intake in Japan over the last century. Eur. J. Nutr. 2002, 41, 222–227. [Google Scholar] [CrossRef]
- Stephen, A.M.; Champ, M.M.-J.; Cloran, S.J.; Fleith, M.; Lieshout, L.V.; Mejborn, H.; Burley, V.J. Dietary fibre in Europe: Current state of knowledge on definitions, sources, recommendations, intakes and relationships to health. Nutr. Res. Rev. 2017, 30, 149–190. [Google Scholar] [CrossRef] [Green Version]
- Prynne, C.J.; McCarron, A.; Wadsworth, M.E.; Stephen, A.M. Dietary fibre and phytate—A balancing act: Results from three time points in a British birth cohort. Br. J. Nutr. 2010, 103, 274–280. [Google Scholar] [CrossRef] [PubMed]
- Public Health England; Food Standards Agency. National Diet and Nutrition Survey: Results from Years 1, 2, 3 and 4 (Combined) of the Rolling Programme (2008/2009–2011/2012); Public Health England: London, UK, 2014.
- King, D.E.; Mainous, A.G., 3rd; Lambourne, C.A. Trends in dietary fiber intake in the United States, 1999–2008. J. Acad. Nutr. Diet. 2012, 112, 642–648. [Google Scholar] [CrossRef] [PubMed]
- Dingeo, G.; Brito, A.; Samouda, H.; Iddir, M.; Frano, M.R.L.; Bohn, T. Phytochemicals as modifiers of gut microbial communities. Food Funct. 2020, 11, 8444–8471. [Google Scholar] [CrossRef] [PubMed]
- Makki, K.; Deehan, E.C.; Walter, J.; Bäckhed, F. The Impact of Dietary Fiber on Gut Microbiota in Host Health and Disease. Cell Host Microbe 2018, 23, 705–716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jha, S.K.; Singh, H.R.; Prakash, P. Dietary Fiber and Human Health: An Introduction; Elsevier: Amsterdam, The Netherlands, 2017; pp. 1–22. [Google Scholar] [CrossRef]
- Desai, M.S.; Seekatz, A.M.; Koropatkin, N.M.; Kamada, N.; Hickey, C.A.; Wolter, M.; Pudlo, N.A.; Kitamoto, S.; Terrapon, N.; Muller, A.; et al. A Dietary Fiber-Deprived Gut Microbiota Degrades the Colonic Mucus Barrier and Enhances Pathogen Susceptibility. Cell 2016, 167, 1339–1353. [Google Scholar] [CrossRef] [Green Version]
- Tan, J.; McKenzie, C.; Vuillermin, P.J.; Goverse, G.; Vinuesa, C.G.; Mebius, R.E.; Macia, L.; Mackay, C.R. Dietary Fiber and Bacterial SCFA Enhance Oral Tolerance and Protect against Food Allergy through Diverse Cellular Pathways. Cell Rep. 2016, 15, 2809–2824. [Google Scholar] [CrossRef] [Green Version]
- Kuo, S.-M. The interplay between fiber and the intestinal microbiome in the inflammatory response. Adv. Nutr. 2013, 4, 16–28. [Google Scholar] [CrossRef] [Green Version]
- Topping, D.L.; Clifton, P.M. Short-Chain Fatty Acids and Human Colonic Function: Roles of Resistant Starch and Nonstarch Polysaccharides. Physiol. Rev. 2001, 81, 1031–1064. [Google Scholar] [CrossRef] [Green Version]
- Gill, P.A.; Zelm, M.C.V.; Muir, J.G.; Gibson, P.R. Review article: Short chain fatty acids as potential therapeutic agents in human gastrointestinal and inflammatory disorders. Aliment. Pharmacol. Ther. 2018, 48, 15–34. [Google Scholar] [CrossRef] [Green Version]
- Louis, P.; Hold, G.L.; Flint, H.J. The gut microbiota, bacterial metabolites and colorectal cancer. Nat. Rev. Microbiol. 2014, 12, 661–672. [Google Scholar] [CrossRef]
- Singh, N.; Gurav, A.; Sivaprakasam, S.; Brady, E.; Padia, R.; Shi, H.; Thangaraju, M.; Prasad, P.D.; Manicassamy, S.; Munn, D.H.; et al. Activation of Gpr109a, Receptor for Niacin and the Commensal Metabolite Butyrate, Suppresses Colonic Inflammation and Carcinogenesis. Immunity 2014, 40, 128–139. [Google Scholar] [CrossRef] [Green Version]
- Chang, P.V.; Hao, L.; Offermanns, S.; Medzhitov, R. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc. Natl. Acad. Sci. USA 2014, 111, 2247–2252. [Google Scholar] [CrossRef] [Green Version]
- Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef]
- Smith, P.M.; Howitt, M.R.; Panikov, N.; Michaud, M.; Gallini, C.A.; Bohlooly, Y.-M.; Glickman, J.N.; Garrett, W.S. The Microbial Metabolites, Short-Chain Fatty Acids, Regulate Colonic Treg Cell Homeostasis. Science 2013, 341, 569–573. [Google Scholar] [CrossRef] [Green Version]
- Zimmerman, M.A.; Singh, N.; Martin, P.M.; Thangaraju, M.; Ganapathy, V.; Waller, J.L.; Shi, H.; Robertson, K.D.; Munn, D.H.; Liu, K. Butyrate suppresses colonic inflammation through HDAC1-dependent Fas upregulation and Fas-mediated apoptosis of T cells. Am. J. Physiol.-Gastrointest. Liver Physiol. 2012, 302, G1405–G1415. [Google Scholar] [CrossRef]
- Donohoe, D.; Garge, N.; Zhang, X.; Sun, W.; O’Connel, T.; Bunger, M.; Bultman, S. The Microbiome and Butyrate Regulate Energy Metabolism and Autophagy in the Mammalian Colon. Cell Metab. 2011, 13, 517–526. [Google Scholar] [CrossRef] [Green Version]
- Usami, M.; Kishimoto, K.; Ohata, A.; Miyoshi, M.; Aoyama, M.; Fueda, Y.; Kotani, J. Butyrate and trichostatin A attenuate nuclear factor κB activation and tumor necrosis factor α secretion and increase prostaglandin E2 secretion in human peripheral blood mononuclear cells. Nutr. Res. 2008, 28, 321–328. [Google Scholar] [CrossRef]
- Hamer, H.M.; Jonkers, D.; Venema, K.; Vanhoutvin, S.; Troost, F.J.; Brummer, R.J. Review article: The role of butyrate on colonic function. Aliment. Pharmacol. Ther. 2007, 27, 104–119. [Google Scholar] [CrossRef]
- Gaudier, E.; Jarry, A.; Blottière, H.M.; Coppet, P.D.; Buisine, M.P.; Aubert, J.P.; Laboisse, C.; Cherbut, C.; Hoebler, C. Butyrate specifically modulates MUC gene expression in intestinal epithelial goblet cells deprived of glucose. Am. J. Physiol. 2004, 287, G1168–G1174. [Google Scholar] [CrossRef] [Green Version]
- Willemsen, L.E.M. Short chain fatty acids stimulate epithelial mucin 2 expression through differential effects on prostaglandin E1 and E2 production by intestinal myofibroblasts. Gut 2003, 52, 1442–1447. [Google Scholar] [CrossRef]
- Pryde, S.E.; Duncan, S.H.; Hold, G.L.; Stewart, C.S.; Flint, H.J. The microbiology of butyrate formation in the human colon. FEMS Microbiol. Lett. 2002, 217, 133–139. [Google Scholar] [CrossRef] [PubMed]
- Boets, E.; Gomand, S.V.; Deroover, L.; Preston, T.; Vermeulen, K.; de Preter, V.; Hamer, H.M.; van den Mooter, G.; de Vuyst, L.; Courtin, C.M.; et al. Systemic availability and metabolism of colonic-derived short-chain fatty acids in healthy subjects: A stable isotope study. J. Physiol. 2017, 595, 541–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bloemen, J.G.; Venema, K.; van de Poll, M.C.; Olde Damink, S.W.; Buurman, W.A.; Dejong, C.H. Short chain fatty acids exchange across the gut and liver in humans measured at surgery. Clin. Nutr. 2009, 28, 657–661. [Google Scholar] [CrossRef] [PubMed]
- Sakata, T. Pitfalls in short-chain fatty acid research: A methodological review. Anim. Sci. J. 2019, 90, 3–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reichardt, N.; Vollmer, M.; Holtrop, G.; Farquharson, F.M.; Wefers, D.; Bunzel, M.; Duncan, S.H.; Drew, J.E.; Williams, L.M.; Milligan, G.; et al. Specific substrate-driven changes in human faecal microbiota composition contrast with functional redundancy in short-chain fatty acid production. ISME J. 2018, 12, 610–622. [Google Scholar] [CrossRef]
- Durack, J.; Christophersen, C.T. Human Respiratory and Gut Microbiomes—Do They Really Contribute to Respiratory Health? Front. Pediatrics 2020, 8, 528. [Google Scholar] [CrossRef]
- Levan, S.R.; Stamnes, K.A.; Lin, D.L.; Panzer, A.R.; Fukui, E.; McCauley, K.; Fujimura, K.E.; McKean, M.; Ownby, D.R.; Zoratti, E.M.; et al. Elevated faecal 12,13-diHOME concentration in neonates at high risk for asthma is produced by gut bacteria and impedes immune tolerance. Nat. Microbiol. 2019, 4, 1851–1861. [Google Scholar] [CrossRef]
- Durack, J.; Kimes, N.E.; Lin, D.L.; Rauch, M.; McKean, M.; McCauley, K.; Panzer, A.R.; Mar, J.S.; Cabana, M.D.; Lynch, S.V. Delayed gut microbiota development in high-risk for asthma infants is temporarily modifiable by Lactobacillus supplementation. Nat. Commun. 2018, 9, 707. [Google Scholar] [CrossRef] [Green Version]
- Fujimura, K.E.; Sitarik, A.R.; Havstad, S.; Lin, D.L.; Levan, S.; Fadrosh, D.; Panzer, A.R.; LaMere, B.; Rackaityte, E.; Lukacs, N.W.; et al. Neonatal gut microbiota associates with childhood multi– sensitized atopy and T–cell differentiation. Nat. Med. 2016, 22, 1187–1191. [Google Scholar] [CrossRef] [Green Version]
- Maslowski, K.M.; Vieira, A.T.; Ng, A.; Kranich, J.; Sierro, F.; Di, Y.; Schilter, H.C.; Rolph, M.S.; Mackay, F.; Artis, D.; et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 2009, 461, 1282–1286. [Google Scholar] [CrossRef]
- Skelly, A.N.; Sato, Y.; Kearney, S.; Honda, K. Mining the microbiota for microbial and metabolite-based immunotherapies. Nat. Rev. Immunol. 2019, 19, 305–323. [Google Scholar] [CrossRef]
- Tuck, C.J.; Vanner, S.J. Dietary therapies for functional bowel symptoms: Recent advances, challenges, and future directions. Neurogastroenterol. Motil. 2017, 30, e13238. [Google Scholar] [CrossRef]
- Blacher, E.; Levy, M.; Tatirovsky, E.; Elinav, E. Microbiome-Modulated Metabolites at the Interface of Host Immunity. J. Immunol. 2017, 198, 572–580. [Google Scholar] [CrossRef] [Green Version]
- Saura-Calixto, F. Dietary Fiber as a Carrier of Dietary Antioxidants: An Essential Physiological Function. J. Agric. Food Chem. 2011, 1, 43–49. [Google Scholar] [CrossRef]
- Palafox-Carlos, H.; Ayala-Zavala, J.F.; González-Aguilar, G.A. The Role of Dietary Fiber in the Bioaccessibility and Bioavailability of Fruit and Vegetable Antioxidants. J. Food Sci. 2011, 1, R6–R15. [Google Scholar] [CrossRef] [Green Version]
- Luca, S.V.; Macovei, I.; Bujor, A.; Miron, A.; Skalicka-Woźniak, K.; Aprotosoaie, A.C.; Trifa, A. Bioactivity of dietary polyphenols: The role of metabolites. Food Sci. Nutr. 2020, 60, 626–659. [Google Scholar] [CrossRef]
- Dobson, C.C.; Mottawea, W.; Rodrigue, A.; Pereira, B.L.B.; Hammami, R.; Power, K.A.; Bordenave, N. Impact of molecular interactions with phenolic compounds on food polysaccharides functionality. Adv. Food Nutr. Res. 2019, 90, 135–181. [Google Scholar]
- Padayachee, A.; Day, L.; Howell, K.; Gidley, M.J. Complexity and health functionality of plant cell wall fibers from fruits and vegetables. Food Sci. Nutr. 2017, 1, 59–81. [Google Scholar] [CrossRef]
- Bohn, T. Dietary factors affecting polyphenol bioavailability. Nutr. Rev. 2014, 72, 429–452. [Google Scholar] [CrossRef]
- Kaulmann, A.; Bohn, T. Bioactivity of Polyphenols: Preventive and Adjuvant Strategies toward Reducing Inflammatory Bowel Diseases—Promises, Perspectives, and Pitfalls. Oxidative Med. Cell. Longev. 2016, 2016, 9346470. [Google Scholar] [CrossRef] [Green Version]
- Çelik, E.E.; Rubio, J.M.A.; Andersen, M.L.; Gökmen, V. Interactions of dietary fiber bound antioxidants with hydroxycinnamic and hydroxybenzoic acids in aqueous and liposome media. Food Chem. 2019, 278, 294–304. [Google Scholar] [CrossRef] [PubMed]
- Bermúdez-Oria, A.; Rodríguez-Gutiérrez, G.; Fernández-Prior, Á.; Vioque, B.; Fernández-Bolaños, J. Strawberry dietary fiber functionalized with phenolic antioxidants from olives. Interactions between polysaccharides and phenolic compounds. Food Chem. 2019, 280, 310–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Çelik, E.E.; Gökmen, V.; Skibsted, L.H. Synergism between Soluble and Dietary Fiber Bound Antioxidants. J. Agric. Food Chem. 2015, 63, 2338–2343. [Google Scholar] [CrossRef] [PubMed]
- Doğan, E.; Gökmen, V. Mechanism of the interaction between insoluble wheat bran and polyphenols leading to increased antioxidant capacity. Food Res. Int. 2015, 69, 189–193. [Google Scholar] [CrossRef]
- Saura-Calixto, F.; Serrano, J.; Goni, I. Intake and bioaccessibility of total polyphenols in a whole diet. Food Chem. 2007, 101, 492–501. [Google Scholar] [CrossRef] [Green Version]
- Johnson, A.J.; Vangay, P.; Al-Ghalith, G.A.; Hillmann, B.M.; Ward, T.L.; Shields-Cutler, R.R.; Kim, A.D.; Shmagel, A.K.; Syed, A.N.; Students, P.M.C.; et al. Daily Sampling Reveals Personalized Diet-Microbiome Associations in Humans. Cell Host Microbe 2019, 25, 789–802. [Google Scholar] [CrossRef]
- Filippis, F.D.; Pellegrini, N.; Vannini, L.; Jeffery, I.B.; Storia, A.L.; Laghi, L.; Serrazanetti, D.I.; Cagno, R.D.; Ferrocino, I.; Lazzi, C.; et al. High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. BMJ Gut 2016, 65, 1812–1821. [Google Scholar] [CrossRef]
- Martínez, G.P.; Bäuerl, C.; Collado, M.C. Understanding gut microbiota in elderly’s health will enable intervention through probiotics. Benef. Microbes 2014, 3, 235–246. [Google Scholar] [CrossRef]
- Wu, G.D.; Chen, J.; Hoffmann, C.; Bittinger, K.; Chen, Y.-Y.; Keilbaugh, S.A.; Bewtra, M.; Knights, D.; Walters, W.A.; Knight, R.; et al. Linking Long-Term Dietary Patterns with Gut Microbial Enterotypes. Science 2011, 334, 105–109. [Google Scholar] [CrossRef] [Green Version]
- Markiewicz, L.H.; Honke, J.; Haros, M.; Swiaztecka, D.; Wróblewska, B. Diet shapes the ability of human intestinal microbiota to degrade phytate—In vitro studies. J. Appl. Microbiol. 2013, 115, 247–259. [Google Scholar] [CrossRef] [Green Version]
- David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef]
- Merra, G.; Noce, A.; Marrone, G.; Cintoni, M.; Tarsitano, M.G.; Capacci, A.; de Lorenzo, A. Influence of Mediterranean Diet on Human Gut Microbiota. Nutrients 2020, 13, 7. [Google Scholar] [CrossRef]
- Riaz Rajoka, M.S.; Thirumdas, R.; Mehwish, H.M.; Umair, M.; Khurshid, M.; Hayat, H.F.; Phimolsiripol, Y.; Pallarés, N.; Martí-Quijal, F.J.; Barba, F.J. Role of Food Antioxidants in Modulating Gut Microbial Communities: Novel Understandings in Intestinal Oxidative Stress Damage and Their Impact on Host Health. Antioxidants 2021, 10, 1563. [Google Scholar] [CrossRef]
- Esworthy, R.S.; Smith, D.D.; Chu, F.-F. A Strong Impact of Genetic Background on Gut Microflora in Mice. Int. J. Inflamm. 2010, 2010, 986046. [Google Scholar] [CrossRef] [Green Version]
- Nøhr, M.K.; Egerod, K.L.; Christiansen, S.H.; Gille, A.; Offermanns, S.; Schwartz, T.W.; Møller, M. Expression of the short chain fatty acid receptor GPR41/FFAR3 in autonomic and somatic sensory ganglia. Neuroscience 2015, 290, 126–137. [Google Scholar] [CrossRef]
- Poul, E.L.; Loison, C.; Struyf, S.; Springael, J.-Y.; Lannoy, V.; Decobecq, M.-E.; Brezillon, S.; Dupriez, V.; Vassart, G.; Damme, J.V.; et al. Functional Characterization of Human Receptors for Short Chain Fatty Acids and Their Role in Polymorphonuclear Cell Activation. J. Biol. Chem. 2003, 278, 25481–25489. [Google Scholar] [CrossRef] [Green Version]
- Hudson, B.D.; Murdoch, H.; Milligan, G. Minireview: The Effects of Species Ortholog and SNP Variation on Receptors for Free Fatty Acids. Mol. Endocrinol. 2013, 27, 1177–1187. [Google Scholar] [CrossRef] [Green Version]
- Van der Hee, B.; Wells, J.M. Microbial Regulation of Host Physiology by Short-chain Fatty Acids. Trends Microbiol. 2021, 29, 700–712. [Google Scholar] [CrossRef]
- Medicine, P.C.F.R. Dietary Fibre Recommendations. Available online: https://www.pcrm.org/good-nutrition/nutrition-information/fiber (accessed on 22 October 2021).
- EFSA Panel on Dietetic Products, Nutrition and Allergies. Scientific Opinion on Dietary Reference Values for carbohydrates and dietary fibre. EFSA J. 2010, 8, 1462. [Google Scholar] [CrossRef] [Green Version]
- Alkerwi, A.A.; Sauvageot, N.; Donneau, A.-F.; Lair, M.-L.; Couffignal, S.; Beissel, J.; Delagardelle, C.; Wagener, Y.; Albert, A.; Guillaume, M. First nationwide survey on cardiovascular risk factors in Grand-Duchy of Luxembourg (ORISCAV-LUX). BMC Public Health 2010, 10, 468. [Google Scholar] [CrossRef] [Green Version]
- Alkerwi, A.A.; Donneau, A.-F.; Sauvageot, N.; Lair, M.-L.; Albert, A.; Guillaume, M. Dietary, behavioural and socio-economic determinants of the metabolic syndrome among adults in Luxembourg: Findings from the ORISCAV-LUX study. Public Health Nutr. 2012, 15, 849–859. [Google Scholar] [CrossRef] [PubMed]
- Alkerwi, A.A.; Pastore, J.; Sauvageot, N.; Coroller, G.L.; Bocquet, V.; d’Incau, M.; Aguayo, G.; Appenzeller, B.; Bejko, D.; Bohn, T.; et al. Challenges and benefits of integrating diverse sampling strategies in the observation of cardiovascular risk factors (ORISCAV-LUX 2) study. BMC Med. Res. Metholody 2019, 19, 27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vangay, P.; Johnson, A.J.; Ward, T.L.; Al-Ghalith, G.A.; Shields-Cutler, R.R.; Hillmann, B.M.; Lucas, S.K.; Beura, L.K.; Thompson, E.A.; Till, L.M.; et al. U.S. immigration westernizes the human gut microbiome. Cell 2018, 175, 962–972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reynolds, A.; Mann, J.; Cummings, J.; Winter, N.; Mete, E.; Te Morenga, L. Carbohydrate quality and human health: A series of systematic reviews and meta-analyses. Lancet 2019, 393, 434–445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schättin, A.; Gennaro, F.; Egloff, M.; Vogt, S.; de Bruin, E.D. Physical Activity, Nutrition, Cognition, Neurophysiology, and Short-Time Synaptic Plasticity in Healthy Older Adults: A Cross-Sectional Study. Front. Aging Neurosci. 2018, 10, 242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chesnais, J.-C. The Inversion of the Age Pyramid and the Future Population Decline in France: Implications and Policy Responses; United Nations: New York, NY, USA, 2000. [Google Scholar]
- Biagi, E.; Candela, M.; Turroni, S.; Garagnani, P.; Franceschi, C.; Brigidi, P. Ageing and gut microbes: Perspectives for health maintenance and longevity. Pharmacol. Res. 2013, 1, 11–20. [Google Scholar] [CrossRef]
- Franceschi, C.; Capri, M.; Monti, D.; Giunta, S.; Olivieri, F.; Sevini, F.; Panourgia, M.P.; Invidia, L.; Celani, L.; Scurti, M.; et al. Inflammaging and anti-inflammaging: A systemic perspective on aging and longevity emerged from studies in humans. Mech. Ageing Dev. 2006, 128, 92–105. [Google Scholar] [CrossRef]
- Franceschi, C.; Olivieri, F.; Marchegiani, F.; Cardelli, M.; Cavallone, L.; Capri, M.; Salvioli, S.; Valensin, S.; Benedictis, G.D.; Iorio, A.D.; et al. Genes involved in immune response/inflammation, IGF1/insulin pathway and response to oxidative stress play a major role in the genetics of human longevity: The lesson of centenarians. Mech. Ageing Dev. 2005, 126, 351–361. [Google Scholar] [CrossRef]
- Dinan, T.G.; Cryan, J.F. Gut instincts: Microbiota as a key regulator of brain development, ageing and neurodegeneration. J. Physiol. 2016, 595, 489–494. [Google Scholar] [CrossRef] [Green Version]
- Biagi, E.; Nylund, L.; Candela, M.; Ostan, R.; Bucci, L.; Pini, E.; Nikkïla, J.; Monti, D.; Satokari, R.; Franceschi, C.; et al. Through Ageing, and Beyond: Gut Microbiota and Inflammatory Status in Seniors and Centenarians. PLoS ONE 2010, 5, e10667. [Google Scholar] [CrossRef]
- Biagi, E.; Candela, M.; Fairweather-Tait, S.; Franceschi, C.; Brigidi, P. Ageing of the human metaorganism: The microbial counterpart. Age 2012, 34, 247–267. [Google Scholar] [CrossRef]
- Tiihonen, K.; Ouwehand, A.C.; Rautonen, N. Human intestinal microbiota and healthy ageing. Ageing Res. Rev. 2010, 2, 107–116. [Google Scholar] [CrossRef]
- Duncan, S.H.; Flint, H.J. Probiotics and prebiotics and health in ageing populations. Maturitas 2013, 1, 44–50. [Google Scholar] [CrossRef]
- Offringa, L.C.; Hartle, J.C.; Rigdon, J.; Gardner, C.D. Changes in Quantity and Sources of Dietary Fiber from Adopting Healthy Low-Fat vs. Healthy Low-Carb Weight Loss Diets: Secondary Analysis of DIETFITS Weight Loss Diet Study. Nutrients 2021, 13, 3625. [Google Scholar] [CrossRef]
- Mccleary, B.V. Total Dietary Fiber (CODEX Definition) in Foods and Food Ingredients by a Rapid Enzymatic-Gravimetric Method and Liquid Chromatography: Collaborative Study, First Action 2017.16. J. AOAC Int. 2019, 102, 196–207. [Google Scholar] [CrossRef] [Green Version]
- Codex Alimentarius Commission. Report of the 30th Session of the Codex Committee on Nutrition and Foods for Special Dietary Uses; No. ALINORM 02/32/26; FAO: Rome, Italy; WHO: Geneva, Switzerland, 2009. [Google Scholar]
- Kato, N.; Iwami, K. Resistant Protein; Its Existence and Function Beneficial to Health. J. Nutr. Sci. Vitaminol. 2002, 48, 1–5. [Google Scholar] [CrossRef] [Green Version]
- Wang, Z.; Liang, M.; Li, H.; Cai, L.; Yang, L. Rice Protein Exerts Anti-Inflammatory Effect in Growing and Adult Rats via Suppressing NF-κB Pathway. Int. J. Mol. Sci. 2019, 20, 6164. [Google Scholar] [CrossRef] [Green Version]
- Yang, L.; Chen, J.; Xu, T.; Qiu, W.; Zhang, Y.; Zhang, L.; Xu, F.; Liu, H. Rice Protein Extracted by Different Methods Affects Cholesterol Metabolism in Rats Due to Its Lower Digestibility. Int. J. Mol. Sci. 2011, 12, 7594–7608. [Google Scholar] [CrossRef] [Green Version]
- Agence Française de Sécurité Sanitaire des Aliments (AFFSA). Dietary Fibre: Definitions, Analysis and Nutrition Claims; Agence Française de Sécurité Sanitaire des Aliments (AFFSA): Paris, France, 2002. [Google Scholar]
- Martel, J.; Ojcius, D.M.; Ko, Y.-F.; Young, J.D. Phytochemicals as Prebiotics and Biological Stress Inducers. Trends Biochem. Sci. 2020, 45, 462–471. [Google Scholar] [CrossRef]
- Fatima, A.; Khan, M.S.; Ahmad, M.W. Therapeutic Potential of Equol: A Comprehensive Review. Curr. Pharm. Des. 2020, 26, 5837–5843. [Google Scholar] [CrossRef]
- Jones, J.M. CODEX-aligned dietary fiber definitions help to bridge the ‘fiber gap’. Nutr. J. 2014, 13, 34. [Google Scholar] [CrossRef] [PubMed]
- Williams, B.A.; Mikkelsen, D.; Flanagan, B.M.; Gidley, M.J. “Dietary fibre”: Moving beyond the “soluble/insoluble” classification for monogastric nutrition, with an emphasis on humans and pigs. J. Anim. Sci. Biotechnol. 2019, 10, 45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Macfarlane, G.T.; Macfarlane, S. Bacteria, Colonic Fermentation, and Gastrointestinal Health. J. AOAC Int. 2012, 95, 50–60. [Google Scholar] [CrossRef] [PubMed]
- Cummings, J.H.; Gibson, G.R.; Macfarlane, G.T. Quantitative estimates of fermentation in the hind gut of man. Acta Vet. Scand. Suppl. 1989, 86, 76–82. [Google Scholar] [PubMed]
- Rios-Covian, D.; González, S.; Nogacka, A.M.; Arboleya, S.; Salazar, N.; Gueimonde, M.; de Los Reyes-Gavilán, C.G. An Overview on Fecal Branched Short-Chain Fatty Acids Along Human Life and as Related with Body Mass Index: Associated Dietary and Anthropometric Factors. Front. Microbiol. 2020, 11, 973. [Google Scholar] [CrossRef] [PubMed]
- McDonald, J.A.K.; Mullish, B.H.; Pechlivanis, A.; Liu, Z.; Brignardello, J.; Kao, D.; Holmes, E.; Li, J.V.; Clarke, T.B.; Thursz, M.R.; et al. Inhibiting Growth of Clostridioides difficile by Restoring Valerate, Produced by the Intestinal Microbiota. Gastroenterology 2018, 155, 1495–1507.e1415. [Google Scholar] [CrossRef] [Green Version]
- François, I.E.J.A.; Lescroart, O.; Veraverbeke, W.S.; Marzorati, M.; Possemiers, S.; Hamer, H.; Windey, K.; Welling, G.W.; Delcour, J.A.; Courtin, C.M.; et al. Effects of Wheat Bran Extract Containing Arabinoxylan Oligosaccharides on Gastrointestinal Parameters in Healthy Preadolescent Children. J. Pediatric Gastroenterol. Nutr. 2014, 58, 647–653. [Google Scholar] [CrossRef] [Green Version]
- Breit, S.; Kupferberg, A.; Rogler, G.; Hasler, G. Vagus Nerve as Modulator of the Gut-Brain Axis in Psychiatric and Inflammatory Disorders. Front. Psychiatry 2018, 9, 44. [Google Scholar] [CrossRef] [Green Version]
- Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids from Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef] [Green Version]
- Granado-Serrano, A.B.; Martín-Garí, M.; Sánchez, V.; Riart Solans, M.; Berdún, R.; Ludwig, I.A.; Rubió, L.; Vilaprinyó, E.; Portero-Otín, M.; Serrano, J.C.E. Faecal bacterial and short-chain fatty acids signature in hypercholesterolemia. Sci. Rep. 2019, 9, 1772. [Google Scholar] [CrossRef]
- Nataraj, B.H.; Ali, S.A.; Behare, P.V.; Yadav, H. Postbiotics-parabiotics: The new horizons in microbial biotherapy and functional foods. Microb. Cell Factories 2020, 19, 168. [Google Scholar] [CrossRef]
- Dasarathy, S.; Mookerjee, R.P.; Rackayova, V.; Rangroo Thrane, V.; Vairappan, B.; Ott, P.; Rose, C.F. Ammonia toxicity: From head to toe? Metab. Brain Dis. 2017, 32, 529–538. [Google Scholar] [CrossRef]
- Ding, L.; Huang, Z.; Lu, Y.; Liang, L.; Li, N.; Xu, Z.; Zhang, J.; Shi, H.; Hong, M. Toxic effects of ammonia on intestinal health and microbiota in red-eared slider (Trachemys scripta elegans). Chemosphere 2021, 280, 130630. [Google Scholar] [CrossRef]
- Di Masi, A.; Ascenzi, P. H2S: A “double face” molecule in health and disease. BioFactors 2013, 39, 186–196. [Google Scholar] [CrossRef]
- Ishizaka, S.; Kikuchi, E.; Tsujii, T. Effects of acetate on human immune system. Immunopharmacol. Immunotoxicol. 1993, 15, 151–162. [Google Scholar] [CrossRef]
- Todesco, T.; Rao, A.V.; Bosello, O.; Jenkins, D.J. Propionate lowers blood glucose and alters lipid metabolism in healthy subjects. Am. J. Clin. Nutr. 1991, 54, 860–865. [Google Scholar] [CrossRef]
- Hamer, H.M.; Jonkers, D.M.; Bast, A.; Vanhoutvin, S.A.; Fischer, M.A.; Kodde, A.; Troost, F.J.; Venema, K.; Brummer, R.J. Butyrate modulates oxidative stress in the colonic mucosa of healthy humans. Clin. Nutr. 2009, 28, 88–93. [Google Scholar] [CrossRef]
- Ge, H.; Li, X.; Weiszmann, J.; Wang, P.; Baribault, H.; Chen, J.L.; Tian, H.; Li, Y. Activation of G protein-coupled receptor 43 in adipocytes leads to inhibition of lipolysis and suppression of plasma free fatty acids. Endocrinology 2008, 149, 4519–4526. [Google Scholar] [CrossRef] [Green Version]
- Hong, Y.-H.; Nishimura, Y.; Hishikawa, D.; Tsuzuki, H.; Miyahara, H.; Gotoh, C.; Choi, K.-C.; Feng, D.D.; Chen, C.; Lee, H.-G.; et al. Acetate and Propionate Short Chain Fatty Acids Stimulate Adipogenesis via GPCR43. Endocrinology 2005, 146, 5092–5099. [Google Scholar] [CrossRef] [Green Version]
- Anil, M.H.; Forbes, J.M. Feeding in sheep during intraportal infusions of short-chain fatty acids and the effect of liver denervation. J. Physiol. 1980, 298, 407–414. [Google Scholar] [CrossRef]
- Thacker, P.A.; Bell, J.M.; Classen, H.L.; Campbell, G.L.; Rossnagel, B.G. The nutritive value of hulless barley for swine. Anim. Feed Sci. Technol. 1988, 19, 191–196. [Google Scholar] [CrossRef]
- Illman, R.J.; Topping, D.L.; McLntosh, G.H.; Trimble, R.P.; Storer, G.B.; Taylor, M.N.; Cheng, B.Q. Hypocholesterolaemic Effects of Dietary Propionate: Studies in Whole Animals and Perfused Rat Liver. Ann. Nutr. Metab. 1988, 32, 97–107. [Google Scholar] [CrossRef]
- MacFabe, D.F.; Cain, D.P.; Rodriguez-Capote, K.; Franklin, A.E.; Hoffman, J.E.; Boon, F.; Taylor, A.R.; Kavaliers, M.; Ossenkopp, K.P. Neurobiological effects of intraventricular propionic acid in rats: Possible role of short chain fatty acids on the pathogenesis and characteristics of autism spectrum disorders. Behav. Brain Res. 2007, 176, 149–169. [Google Scholar] [CrossRef] [PubMed]
- Shultz, S.R.; MacFabe, D.F.; Ossenkopp, K.P.; Scratch, S.; Whelan, J.; Taylor, R.; Cain, D.P. Intracerebroventricular injection of propionic acid, an enteric bacterial metabolic end-product, impairs social behavior in the rat: Implications for an animal model of autism. Neuropharmacology 2008, 54, 901–911. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.; Hegsted, M.; McCutcheon, K.L.; Keenan, M.J.; Xi, X.; Raggio, A.M.; Martin, R.J. Peptide YY and Proglucagon mRNA Expression Patterns and Regulation in the Gut. Obesity 2006, 14, 683–689. [Google Scholar] [CrossRef] [Green Version]
- Gao, Z.; Yin, J.; Zhang, J.; Ward, R.E.; Martin, R.J.; Lefevre, M.; Cefalu, W.T.; Ye, J. Butyrate Improves Insulin Sensitivity and Increases Energy Expenditure in Mice. Diabetes 2009, 58, 1509–1517. [Google Scholar] [CrossRef] [Green Version]
- Xiong, Y.; Miyamoto, N.; Shibata, K.; Valasek, M.A.; Motoike, T.; Kedzierski, R.M.; Yanagisawa, M. Short-chain fatty acids stimulate leptin production in adipocytes through the G protein-coupled receptor GPR41. Proc. Natl. Acad. Sci. USA 2004, 101, 1045–1050. [Google Scholar] [CrossRef] [Green Version]
- Yonekura, S.; Senoo, T.; Kobayashi, Y.; Yonezawa, T.; Katoh, K.; Obara, Y. Effects of acetate and butyrate on the expression of leptin and short-form leptin receptor in bovine and rat anterior pituitary cells. Gen. Comp. Endocrinol. 2003, 133, 165–172. [Google Scholar] [CrossRef]
- Tazoe, H.; Otomo, Y.; Kaji, I.; Tanaka, R.; Karaki, S.; Kuwahara, A. Roles of short-chain fatty acids receptors, GPR41 and GPR43 on colonic functions. J. Physiol. Pharmacol. 2008, 59, 251–262. [Google Scholar]
- Tedelind, S.; Westberg, F.; Kjerrulf, M.; Vidal, A. Anti-inflammatory properties of the short-chain fatty acids acetate and propionate: A study with relevance to inflammatory bowel disease. World J. Gastroenterol. 2007, 13, 2826–2832. [Google Scholar] [CrossRef]
- Zapolska-Downar, D.; Naruszewicz, M. Propionate reduces the cytokine-induced VCAM-1 and ICAM-1 expression by inhibiting nuclear factor-kappa B (NF-kappaB) activation. J. Physiol. Pharmacol. 2009, 60, 123–131. [Google Scholar]
- Al-Lahham, S.H.; Roelofsen, H.; Priebe, M.; Weening, D.; Dijkstra, M.; Hoek, A.; Rezaee, F.; Venema, K.; Vonk, R.J. Regulation of adipokine production in human adipose tissue by propionic acid. Eur. J. Clin. Investig. 2010, 40, 401–407. [Google Scholar] [CrossRef]
- Curi, R.; Bond, J.A.; Calder, P.C.; Newsholme, E.A. Propionate regulates lymphocyte proliferation and metabolism. Gen. Pharmacol. 1993, 24, 591–597. [Google Scholar] [CrossRef]
- Wright, R.S.; Anderson, J.W.; Bridges, S.R. Propionate Inhibits Hepatocyte Lipid Synthesis. Proc. Soc. Exp. Biol. Med. 1990, 195, 26–29. [Google Scholar] [CrossRef]
- Carlson, J.; Esparza, J.; Swan, J.; Taussig, D.; Combs, J.; Slavin, J. In vitro analysis of partially hydrolyzed guar gum fermentation differences between six individuals. Food Funct. 2016, 7, 1833–1838. [Google Scholar] [CrossRef]
- Potter, T.; Vieira, R.; de Roos, B. Perspective: Application of N-of-1 Methods in Personalized Nutrition Research. Adv. Nutr. 2021, 12, 579–589. [Google Scholar] [CrossRef]
- Jakobsen, J.; Melse-Boonstra, A.; Rychlik, M. Challenges to Quantify Total Vitamin Activity: How to Combine the Contribution of Diverse Vitamers? Curr. Dev. Nutr. 2019, 3, nzz086. [Google Scholar] [CrossRef] [Green Version]
- Yurkovich, J.T.; Tian, Q.; Price, N.D.; Hood, L. A systems approach to clinical oncology uses deep phenotyping to deliver personalized care. Nature 2020, 17, 183–194. [Google Scholar] [CrossRef]
- Subramanian, M.; Wojtusciszyn, A.; Favre, L.; Boughorbel, S.; Shan, J.; Letaief, K.B.; Pitteloud, N.; Chouchane, L. Precision medicine in the era of artificial intelligence: Implications in chronic disease management. J. Transl. Med. 2020, 18, 472. [Google Scholar] [CrossRef]
- Linstow, C.U.V.; Gan-Or, Z.; Brundin, P. Precision medicine in Parkinson’s disease patients with LRRK2 and GBA risk variants—Let’s get even more personal. Transl. Neurodegener. 2020, 9, 39. [Google Scholar] [CrossRef]
- Kumar, M.; Garand, M.; Khodor, S.A. Integrating omics for a better understanding of Inflammatory Bowel Disease: A step towards personalized medicine. J. Transl. Med. 2019, 17, 419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arumugam, M.; Raes, J.; Pelletier, E.; le Paslier, D.; Yamada, T.; Mende, D.R.; Fernandes, G.R.; Tap, J.; Bruls, T.; Batto, J.-M.; et al. Enterotypes of the human gut microbiome. Nature 2011, 473, 174–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Christensen, L.; Roager, H.M.; Astrup, A.; Hjorth, M.F. Microbial enterotypes in personalized nutrition and obesity management. Am. J. Clin. Nutr. 2018, 108, 645–651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zeevi, D.; Korem, T.; Zmora, N.; Israeli, D.; Rothschild, D.; Weinberger, A.; Ben-Yacov, O.; Lador, D.; Avnit-Sagi, T.; Lotan-Pompan, M.; et al. Personalized Nutrition by Prediction of Glycemic Responses. Cell 2015, 163, 1079–1094. [Google Scholar] [CrossRef] [Green Version]
- Wong, J.M.W.; de Souza, R.; Kendall, C.W.C.; Emam, A.; Jenkins, D.J.A. Colonic Health: Fermentation and Short Chain Fatty Acids. J. Clin. Gastroenterol. 2006, 40, 235–243. [Google Scholar] [CrossRef]
- Mitsuhashi, S.; Ballou, S.; Jiang, Z.; Hirsch, W.; Nee, J.; Iturrino, J.; Cheng, V.; Lembo, A. Characterizing Normal Bowel Frequency and Consistency in a Representative Sample of Adults in the United States (NHANES). Am. J. Gastroenterol. 2018, 1, 115–123. [Google Scholar] [CrossRef]
- Sanjoaquin, M.A.; Appleby, P.N.; Spencer, E.A.; Key, T.J. Nutrition and lifestyle in relation to bowel movement frequency: A cross-sectional study of 20 630 men and women in EPIC–Oxford. Public Health Nutr. 2003, 7, 77–83. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.; Zhou, J.; Wang, L. Role and Mechanism of Gut Microbiota in Human Disease. Front. Cell. Infect. Microbiol. 2021, 11, 625913. [Google Scholar] [CrossRef]
- Christodoulides, S.; Dimidi, E.; Fragkos, K.C.; Farmer, A.D.; Whelan, K.; Scott, S.M. Systematic review with meta-analysis: Effect of fibre supplementation on chronic idiopathic constipation in adults. Aliment. Pharmacol. Ther. 2016, 44, 103–116. [Google Scholar] [CrossRef]
- Li, J.; Jia, H.; Cai, X.; Zhong, H.; Feng, Q.; Sunagawa, S.; Arumugam, M.; Kultima, J.R.; Prifti, E.; Nielsen, T.; et al. An integrated catalog of reference genes in the human gut microbiome. Nat. Biotechnol. 2014, 32, 834–841. [Google Scholar] [CrossRef]
- Spor, A.; Koren, O.; Ley, R. Unravelling the effects of the environment and host genotype on the gut microbiome. Nat. Rev. Microbiol. 2011, 9, 279–290. [Google Scholar] [CrossRef]
- Benson, A.K.; Kelly, S.A.; Legge, R.; Ma, F.; Low, S.J.; Kim, J.; Zhang, M.; Oh, P.L.; Nehrenberg, D.; Hua, K.; et al. Individuality in gut microbiota composition is a complex polygenic trait shaped by multiple environmental and host genetic factors. Proc. Natl. Acad. Sci. USA 2010, 107, 18933–18938. [Google Scholar] [CrossRef] [Green Version]
- Rothschild, D.; Weissbrod, O.; Barkan, E.; Kurilshikov, A.; Korem, T.; Zeevi, D.; Costea, P.I.; Godneva, A.; Kalka, I.N.; Bar, N.; et al. Environment dominates over host genetics in shaping human gut microbiota. Nature 2018, 555, 210–215. [Google Scholar] [CrossRef]
- Iebba, V.; Totino, V.; Gagliardi, A.; Santangelo, F.; Cacciotti, F.; Trancassini, M.; Mancini, C.; Cicerone, C.; Corazziari, E.; Pantanella, F.; et al. Eubiosis and dysbiosis: The two sides of the microbiota. New Microbiol. 2016, 39, 1–12. [Google Scholar]
- Goodrich, J.K.; Davenport, E.R.; Waters, J.L.; Clark, A.G.; Ley, R.E. Cross-species comparisons of host genetic associations with the microbiome. Science 2016, 352, 532–535. [Google Scholar] [CrossRef] [Green Version]
- So, D.; Whelan, K.; Rossi, M.; Morrison, M.; Holtmann, G.; Kelly, J.T.; Shanahan, E.R.; Staudacher, H.M.; Campbell, K.L. Dietary fiber intervention on gut microbiota composition in healthy adults: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2018, 107, 965–983. [Google Scholar] [CrossRef] [Green Version]
- Macfarlane, S.; Macfarlane, G.T. Regulation of short-chain fatty acid production. Proc. Nutr. Soc. 2003, 62, 67–72. [Google Scholar] [CrossRef]
- Costea, P.I.; Hildebrand, F.; Arumugam, M.; Bäckhed, F.; Blaser, M.J.; Bushman, F.D.; Willem, M.S.; Fraser, C.M.; Hattori, M.; Huttenhower, C.; et al. Enterotypes in the landscape of gut microbial community composition. Nat. Microbiol. 2018, 3, 8–16. [Google Scholar] [CrossRef] [Green Version]
- Le Chatelier, E.; Nielsen, T.; Qin, J.; Prifti, E.; Hildebrand, F.; Falony, G.; Almeida, M.; Arumugam, M.; Batto, J.M.; Kennedy, S.; et al. Richness of human gut microbiome correlates with metabolic markers. Nature 2013, 500, 541–546. [Google Scholar] [CrossRef]
- Amat, S.; Lantz, H.; Munyaka, P.M.; Willing, B.P. Prevotella in Pigs: The Positive and Negative Associations with Production and Health. Microorganisms 2020, 8, 1584. [Google Scholar] [CrossRef]
- Zheng, L.; Kelly, C.J.; Battista, K.D.; Schaefer, R.; Lanis, J.M.; Alexeev, E.E.; Wang, R.X.; Onyiah, J.C.; Kominsky, D.J.; Colgan, S.P. Microbial-Derived Butyrate Promotes Epithelial Barrier Function through IL-10 Receptor–Dependent Repression of Claudin-2. J. Immunol. 2017, 199, 2976–2984. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Duvallet, C.; Gibbons, S.M.; Gurry, T.; Irizarry, R.A.; Alm, E.J. Meta-analysis of gut microbiome studies identifies disease-specific and shared responses. Nat. Commun. 2017, 8, 1784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gacesa, R.; Kurilshikov, A.; Vich Vila, A.; Sinha, T.; Klaassen, M.A.Y.; Bolte, L.A.; Andreu-Sánchez, S.; Chen, L.; Collij, V.; Hu, S.; et al. Environmental factors shaping the gut microbiome in a Dutch population. Nature 2022, 604, 732–739. [Google Scholar] [CrossRef] [PubMed]
- Derrien, M.; Belzer, C.; de Vos, W.M. Akkermansia muciniphila and its role in regulating host functions. Microb. Pathog. 2017, 106, 171–181. [Google Scholar] [CrossRef] [PubMed]
- Derrien, M.; Vaughan, E.E.; Plugge, C.M.; de Vos, W.M. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int. J. Syst. Evol. Microbiol. 2004, 54, 1469–1476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kashyap, P.C.; Marcobal, A.; Ursell, L.K.; Smits, S.A.; Sonnenburg, E.D.; Costello, E.K.; Higginbottom, S.K.; Domino, S.E.; Holmes, S.P.; Relman, D.A.; et al. Genetically dictated change in host mucus carbohydrate landscape exerts a diet-dependent effect on the gut microbiota. Proc. Natl. Acad. Sci. USA 2013, 110, 17059–17064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, F.; Yu, T.; Huang, G.; Cai, D.; Liang, X.; Su, H.; Zhu, Z.; Li, D.; Yang, Y.; Shen, P.; et al. Gut Microbiota Community and Its Assembly Associated with Age and Diet in Chinese Centenarians. J. Microbiol. Biotechnol. 2015, 25, 1195–1204. [Google Scholar] [CrossRef]
- Bodogai, M.; O’Connell, J.; Kim, K.; Kim, Y.; Moritoh, K.; Chen, C.; Gusev, F.; Vaughan, K.; Shulzhenko, N.; Mattison, J.A.; et al. Commensal bacteria contribute to insulin resistance in aging by activating innate B1a cells. Sci. Transl. Med. 2018, 10, eaat4271. [Google Scholar] [CrossRef]
- Parkin, K.; Christophersen, C.T.; Verhasselt, V.; Cooper, M.N.; Martino, D. Risk Factors for Gut Dysbiosis in Early Life. Microorganisms 2021, 9, 2066. [Google Scholar] [CrossRef]
- Vallianou, N.; Stratigou, T.; Christodoulatos, G.S.; Dalamaga, M. Understanding the Role of the Gut Microbiome and Microbial Metabolites in Obesity and Obesity-Associated Metabolic Disorders: Current Evidence and Perspectives. Curr. Obes. Rep. 2019, 8, 317–332. [Google Scholar] [CrossRef]
- Hiippala, K.; Jouhten, H.; Ronkainen, A.; Hartikainen, A.; Kainulainen, V.; Jalanka, J.; Satokari, R. The Potential of Gut Commensals in Reinforcing Intestinal Barrier Function and Alleviating Inflammation. Nutrients 2018, 10, 988. [Google Scholar] [CrossRef] [Green Version]
- Barash, N.R.; Maloney, J.G.; Singer, S.M.; Dawson, S.C. Giardia Alters Commensal Microbial Diversity throughout the Murine Gut. Infect. Immun. 2017, 85, e00948-16. [Google Scholar] [CrossRef] [Green Version]
- Brunengraber, L.N.; Jayes, F.L.; Leppert, P.C. Injectable Clostridium histolyticum Collagenase as a Potential Treatment for Uterine Fibroids. Reprod. Sci. 2014, 21, 1452–1459. [Google Scholar] [CrossRef] [Green Version]
- Stevens, D.L.; Aldape, M.J.; Bryant, A.E. Life-threatening clostridial infections. Anaerobe 2012, 18, 254–259. [Google Scholar] [CrossRef]
- Ze, X.; Duncan, S.H.; Louis, P.; Flint, H.J. Ruminococcus bromii is a keystone species for the degradation of resistant starch in the human colon. ISME J. 2012, 6, 1535–1543. [Google Scholar] [CrossRef] [Green Version]
- Salonen, A.; Lahti, L.; Salojärvi, J.; Holtrop, G.; Korpela, K.; Duncan, S.H.; Date, P.; Farquharson, F.; Johnstone, A.M.; Lobley, G.E.; et al. Impact of diet and individual variation on intestinal microbiota composition and fermentation products in obese men. ISME J. 2014, 8, 2218–2230. [Google Scholar] [CrossRef]
- Lee, J.; D’Aigle, J.; Atadja, L.; Quaicoe, V.; Honarpisheh, P.; Ganesh, B.P.; Hassan, A.; Graf, J.; Petrosino, J.; Putluri, N.; et al. Gut Microbiota–Derived Short-Chain Fatty Acids Promote Poststroke Recovery in Aged Mice. Circ. Res. 2020, 127, 453–465. [Google Scholar] [CrossRef]
- Chassard, C.; Bernalier-Donadille, A. H2 and acetate transfers during xylan fermentation between a butyrate-producing xylanolytic species and hydrogenotrophic microorganisms from the human gut. FEMS Microbiol. Lett. 2006, 254, 116–122. [Google Scholar] [CrossRef] [Green Version]
- Wrzosek, L.; Miquel, S.; Noordine, M.-L.; Bouet, S.; Chevalier-Curt, M.; Robert, V.; Philippe, C.; Bridonneau, C.; Cherbuy, C.; Robbe-Masselot, C.; et al. Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii influence the production of mucus glycans and the development of goblet cells in the colonic epithelium of a gnotobiotic model rodent. BMC Biol. 2013, 11, 61. [Google Scholar] [CrossRef] [Green Version]
- Quévrain, E.; Maubert, M.A.; Michon, C.; Chain, F.; Marquant, R.; Tailhades, J.; Miquel, S.; Carlier, L.; Bermúdez-Humarán, L.G.; Pigneur, B.; et al. Identification of an anti-inflammatory protein from Faecalibacterium prausnitzii, a commensal bacterium deficient in Crohn’s disease. Gut 2016, 65, 415–425. [Google Scholar] [CrossRef] [Green Version]
- Louis, P.; Young, P.; Holtrop, G.; Flint, H.J. Diversity of human colonic butyrate-producing bacteria revealed by analysis of the butyryl-CoA: Acetate CoA-transferase gene. Environ. Microbiol. 2010, 12, 304–314. [Google Scholar] [CrossRef] [PubMed]
- Cerdó, T.; García-Santos, J.A.; Bermúdez, M.G.; Campoy, C. The Role of Probiotics and Prebiotics in the Prevention and Treatment of Obesity. Nutrients 2019, 11, 635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jan, G.; Belzacq, A.S.; Haouzi, D.; Rouault, A.; Métivier, D.; Kroemer, G.; Brenner, C. Propionibacteria induce apoptosis of colorectal carcinoma cells via short-chain fatty acids acting on mitochondria. Cell Death Differ. 2002, 9, 179–188. [Google Scholar] [CrossRef] [PubMed]
- Aguirre-Portolés, C.; Fernández, L.; Ramírez de Molina, A. Precision Nutrition for Targeting Lipid Metabolism in Colorectal Cancer. Nutrients 2017, 9, 1076. [Google Scholar] [CrossRef] [PubMed]
- Ríos-Covián, D.; Ruas-Madiedo, P.; Margolles, A.; Gueimonde, M.; de los Reyes-Gavilán, C.G.; Salazar, N. Intestinal Short Chain Fatty Acids and their Link with Diet and Human Health. Front. Microbiol. 2016, 7, 185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marchandin, H.; Teyssier, C.; Campos, J.; Jean-Pierre, H.; Roger, F.; Gay, B.; Carlier, J.P.; Jumas-Bilak, E. Negativicoccus succinicivorans gen. nov., sp. nov., isolated from human clinical samples, emended description of the family Veillonellaceae and description of Negativicutes classis nov., Selenomonadales ord. nov. and Acidaminococcaceae fam. nov. in the bacterial phylum Firmicutes. Int. J. Syst. Evol. Microbiol. 2010, 60, 1271–1279. [Google Scholar] [CrossRef] [PubMed]
- Flint, H.J.; Duncan, S.H.; Scott, K.P.; Louis, P. Links between diet, gut microbiota composition and gut metabolism. Proc. Nutr. Soc. 2015, 74, 13–22. [Google Scholar] [CrossRef] [Green Version]
- Pietropaoli, D.; del Pinto, R.; Ferri, C.; Ortu, E.; Monaco, A. Definition of hypertension-associated oral pathogens in NHANES. J. Periodontol. 2019, 90, 866–876. [Google Scholar] [CrossRef]
- Duncan, S.H.; Holtrop, G.; Lobley, G.E.; Calder, A.G.; Stewart, C.S.; Flint, H.J. Contribution of acetate to butyrate formation by human faecal bacteria. Br. J. Nutr. 2004, 91, 915–923. [Google Scholar] [CrossRef] [Green Version]
- Flint, H.J.; Duncan, S.H.; Scott, K.P.; Louis, P. Interactions and competition within the microbial community of the human colon: Links between diet and health. Environ. Microbiol. 2007, 9, 1101–1111. [Google Scholar] [CrossRef]
- Goodrich, J.K.; Davenport, E.R.; Beaumont, M.; Jackson, M.A.; Knight, R.; Ober, C.; Spector, T.D.; Bell, J.T.; Clark, A.G.; Ley, R.E. Genetic Determinants of the Gut Microbiome in UK Twins. Cell Host Microbe 2016, 19, 731–743. [Google Scholar] [CrossRef] [Green Version]
- Jacobson, A.; Lam, L.; Rajendram, M.; Tamburini, F.; Honeycutt, J.; Pham, T.; van Treuren, W.; Pruss, K.; Stabler, S.R.; Lugo, K.; et al. A Gut Commensal-Produced Metabolite Mediates Colonization Resistance to Salmonella Infection. Cell Host Microbe 2018, 24, 296–307.e297. [Google Scholar] [CrossRef] [Green Version]
- Larsbrink, J.; Rogers, T.E.; Hemsworth, G.R.; McKee, L.S.; Tauzin, A.S.; Spadiut, O.; Klinter, S.; Pudlo, N.A.; Urs, K.; Koropatkin, N.M.; et al. A discrete genetic locus confers xyloglucan metabolism in select human gut Bacteroidetes. Nature 2014, 506, 498–502. [Google Scholar] [CrossRef]
- Xu, M.; Xu, X.; Li, J.; Li, F. Association between Gut Microbiota and Autism Spectrum Disorder: A Systematic Review and Meta-Analysis. Front. Psychiatry 2019, 10, 473. [Google Scholar] [CrossRef]
- Macfabe, D.F. Enteric short-chain fatty acids: Microbial messengers of metabolism, mitochondria, and mind: Implications in autism spectrum disorders. Microb. Ecol. Health Dis. 2015, 26, 28177. [Google Scholar] [CrossRef] [Green Version]
- Iljazovic, A.; Roy, U.; Gálvez, E.J.C.; Lesker, T.R.; Zhao, B.; Gronow, A.; Amend, L.; Will, S.E.; Hofmann, J.D.; Pils, M.C.; et al. Perturbation of the gut microbiome by Prevotella spp. enhances host susceptibility to mucosal inflammation. Mucosal Immunol. 2021, 14, 113–124. [Google Scholar] [CrossRef]
- Tett, A.; Huang, K.D.; Asnicar, F.; Fehlner-Peach, H.; Pasolli, E.; Karcher, N.; Armanini, F.; Manghi, P.; Bonham, K.; Zolfo, M.; et al. The Prevotella copri Complex Comprises Four Distinct Clades Underrepresented in Westernized Populations. Cell Host Microbe 2019, 26, 666–679.e667. [Google Scholar] [CrossRef] [Green Version]
- Yang, G.; Hong, E.; Oh, S.; Kim, E. Non-Viable Lactobacillus johnsonii JNU3402 Protects against Diet-Induced Obesity. Foods 2020, 10, 1494. [Google Scholar] [CrossRef]
- Fonseca, W.; Lucey, K.; Jang, S.; Fujimura, K.E.; Rasky, A.; Ting, H.; Petersen, J.; Johnson, C.C.; Boushey, H.A.; Zoratti, E.; et al. Lactobacillus johnsonii Supplementation Attenuates Respiratory Viral Infection via Metabolic Reprogramming and Immune Cell Modulation. Mucosal Immunol. 2017, 10, 1569–1580. [Google Scholar] [CrossRef] [Green Version]
- Goh, Y.J.; Klaenhammer, T.R. Genetic Mechanisms of Prebiotic Oligosaccharide Metabolism in Probiotic Microbes. Annu. Rev. Food Sci. Technol. 2015, 6, 137–156. [Google Scholar] [CrossRef]
- Fukuda, S.; Toh, H.; Hase, K.; Oshima, K.; Nakanishi, Y.; Yoshimura, K.; Tobe, T.; Clarke, J.M.; Topping, D.L.; Suzuki, T.; et al. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature 2011, 469, 543–547. [Google Scholar] [CrossRef] [PubMed]
- Nogacka, A.M.; de los Reyes-Gavilán, C.G.; Arboleya, S.; Ruas-Madiedo, P.; Martínez-Faedo, C.; Suarez, A.; He, F.; Harata, G.; Endo, A.; Salazar, N.; et al. In vitro Selection of Probiotics for Microbiota Modulation in Normal-Weight and Severely Obese Individuals: Focus on Gas Production and Interaction with Intestinal Epithelial Cells. Front. Microbiol. 2021, 12, 630572. [Google Scholar] [CrossRef] [PubMed]
- Basseri, R.J.; Basseri, B.; Pimentel, M.; Chong, K.; Youdim, A.; Low, K.; Hwang, L.; Soffer, E.; Chang, C.; Mathur, R. Intestinal methane production in obese individuals is associated with a higher body mass index. Gastroenterol. Hepatol. 2012, 8, 22–28. [Google Scholar]
- Bordenstein, S.R.; Theis, K.R. Host Biology in Light of the Microbiome: Ten Principles of Holobionts and Hologenomes. PLoS Biol. 2015, 13, e1002226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Asnicar, F.; Berry, S.E.; Valdes, A.M.; Nguyen, L.H.; Piccinno, G.; Drew, D.A.; Leeming, E.; Gibson, R.; le Roy, C.; Khatib, H.A.; et al. Microbiome connections with host metabolism and habitual diet from 1098 deeply phenotyped individuals. Nat. Med. 2021, 27, 321–332. [Google Scholar] [CrossRef] [PubMed]
- Bataineh, M.T.A.; Dash, N.R.; Elkhazendar, M.; Alnusairat, D.A.M.H.; Darwish, I.M.I.; Al-Hajjaj, M.S.; Hamid, Q. Revealing oral microbiota composition and functionality associated with heavy cigarette smoking. J. Transl. Med. 2020, 18, 421. [Google Scholar] [CrossRef]
- Lee, S.H.; Yun, Y.; Kim, S.J.; Lee, E.-J.; Chang, Y.; Ryu, S.; Shin, H.; Kim, H.-L.; Kim, H.-N.; Lee, J.H. Association between Cigarette Smoking Status and Composition of Gut Microbiota: Population-Based Cross-Sectional Study. J. Clin. Med. 2018, 7, 282. [Google Scholar] [CrossRef] [Green Version]
- Telenti, A.; Jiang, X. Treating medical data as a durable asset. Nat. Genet. 2020, 52, 1005–1010. [Google Scholar] [CrossRef]
- Wainschtein, P.; Jain, D.; Zheng, Z.; Cupples, L.A.; Shadyab, A.H.; McKnight, B.; Shoemaker, B.M.; Mitchell, B.D.; Psaty, B.M.; Kooperberg, C.; et al. Recovery of trait heritability from whole genome sequence data. bioRxiv 2019. [Google Scholar] [CrossRef]
- Billingsley, K.J.; Barbosa, I.A.; Bandrés-Ciga, S.; Quinn, J.P.; Bubb, V.J.; Deshpande, C.; Botia, J.A.; Reynolds, R.H.; Zhang, D.; Simpson, M.A.; et al. Mitochondria function associated genes contribute to Parkinson’s Disease risk and later age at onset. NPJ Parkinson’s Dis. 2019, 5, 8. [Google Scholar] [CrossRef] [Green Version]
- Natarajan, P.; Young, R.; Stitziel, N.O.; Padmanabhan, S.; Baber, U.; Mehran, R.; Sartori, S.; Fuster, V.; Reilly, D.F.; Butterworth, A.; et al. Polygenic Risk Score Identifies Subgroup with Higher Burden of Atherosclerosis and Greater Relative Benefit from Statin Therapy in the Primary Prevention Setting. Circulation 2017, 135, 2091–2101. [Google Scholar] [CrossRef] [Green Version]
- Levy, M.; Thaiss, C.A.; Elinav, E. Metabolites: Messengers between the microbiota and the immune system. Genes Dev. 2016, 30, 1589–1597. [Google Scholar] [CrossRef] [Green Version]
- Choi, S.W.; Mak, T.S.-H.; O’Reilly, P.F. Tutorial: A guide to performing polygenic risk score analyses. Nat. Protoc. 2020, 15, 2759–2772. [Google Scholar] [CrossRef]
- Blekhman, R.; Goodrich, J.K.; Huang, K.; Sun, Q.; Bukowski, R.; Bell, J.T.; Spector, T.D.; Keinan, A.; Ley, R.E.; Gevers, D.; et al. Host genetic variation impacts microbiome composition across human body sites. Genome Biol. 2015, 16, 191. [Google Scholar] [CrossRef] [Green Version]
- Kawamoto, S.; Tran, T.H.; Maruya, M.; Suzuki, K.; Doi, Y.; Tsutsui, Y.; Kato, L.M.; Fagarasan, S. The Inhibitory Receptor PD-1 Regulates IgA Selection and Bacterial Composition in the Gut. Science 2012, 336, 485–489. [Google Scholar] [CrossRef]
- Donaldson, G.P.; Ladinsky, M.S.; Yu, K.B.; Sanders, J.G.; Yoo, B.B.; Chou, W.C.; Conner, M.E.; Earl, A.M.; Knight, R.; Bjorkman, P.J.; et al. Gut microbiota utilize immunoglobulin A for mucosal colonization. Science 2018, 360, 795–800. [Google Scholar] [CrossRef] [Green Version]
- Nakajima, A.; Vogelzang, A.; Maruya, M.; Miyajima, M.; Murata, M.; Son, A.; Kuwahara, T.; Tsuruyama, T.; Yamada, S.; Matsuura, M.; et al. IgA regulates the composition and metabolic function of gut microbiota by promoting symbiosis between bacteria. J. Exp. Med. 2018, 215, 2019–2034. [Google Scholar] [CrossRef] [Green Version]
- Takeuchi, T.; Miyauchi, E.; Kanaya, T.; Kato, T.; Nakanishi, Y.; Watanabe, T.; Kitami, T.; Taida, T.; Sasaki, T.; Negishi, H.; et al. Acetate differentially regulates IgA reactivity to commensal bacteria. Nature 2021, 595, 560–564. [Google Scholar] [CrossRef]
- Liang, S.C.; Tan, X.-Y.; Luxenberg, D.P.; Karim, R.; Dunussi-Joannopoulos, K.; Collins, M.; Fouser, L.A. Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J. Exp. Med. 2006, 203, 2271–2279. [Google Scholar] [CrossRef] [Green Version]
- Kawai, T.; Adachi, O.; Ogawa, T.; Takeda, K.; Akira, S. Unresponsiveness of MyD88-deficient mice to endotoxin. Immunity 1999, 11, 115–122. [Google Scholar] [CrossRef] [Green Version]
- Petnicki-Ocwieja, T.; Hrncir, T.; Liu, Y.-J.; Biswas, A.; Hudcovic, T.; Tlaskalova-Hogenova, H.; Kobayashi, K.S. Nod2 is required for the regulation of commensal microbiota in the intestine. Proc. Natl. Acad. Sci. USA 2009, 106, 15813–15818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Knights, D.; Silverberg, M.S.; Weersma, R.K.; Gevers, D.; Dijkstra, G.; Huang, H.; Tyler, A.D.; van Sommeren, S.; Imhann, F.; Stempak, J.M.; et al. Complex host genetics influence the microbiome in inflammatory bowel disease. Genome Med. 2014, 6, 107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salzman, N.H.; Hung, K.; Haribhai, D.; Chu, H.; Karlsson-Sjöberg, J.; Amir, E.; Teggatz, P.; Barman, M.; Hayward, M.; Eastwood, D.; et al. Enteric defensins are essential regulators of intestinal microbial ecology. Nat. Immunol. 2010, 11, 76–82. [Google Scholar] [CrossRef]
- Khachatryan, Z.A.; Ktsoyan, Z.A.; Manukyan, G.P.; Kelly, D.; Ghazaryan, K.A.; Aminov, R.I. Predominant Role of Host Genetics in Controlling the Composition of Gut Microbiota. PLoS ONE 2008, 3, e3064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Poole, A.C.; Goodrich, J.K.; Youngblut, N.D.; Luque, G.G.; Ruaud, A.; Sutter, J.L.; Waters, J.L.; Shi, Q.; El-Hadidi, M.; Johnson, L.M.; et al. Human Salivary Amylase Gene Copy Number Impacts Oral and Gut Microbiomes. Cell Host Microbe 2019, 25, 553–564.e557. [Google Scholar] [CrossRef]
- Sun, B.B.; Maranville, J.C.; Peters, J.E.; Stacey, D.; Staley, J.R.; Blackshaw, J.; Burgess, S.; Jiang, T.; Paige, E.; Surendran, P.; et al. Genomic atlas of the human plasma proteome. Nature 2018, 558, 73–79. [Google Scholar] [CrossRef]
- Troelsen, J.T. Adult-type hypolactasia and regulation of lactase expression. Biochim. Biophys. Acta BBA Gen. Subj. 2005, 1723, 19–32. [Google Scholar] [CrossRef]
- Jackson, V.E.; Latourelle, J.C.; Wain, L.V.; Smith, A.V.; Grove, M.L.; Bartz, T.M.; Obeidat, M.; Province, M.A.; Gao, W.; Qaiser, B.; et al. Meta-analysis of exome array data identifies six novel genetic loci for lung function. Wellcome Open Res 2018, 3, 4. [Google Scholar] [CrossRef] [Green Version]
- Wojcik, G.L.; Graff, M.; Nishimura, K.K.; Tao, R.; Haessler, J.; Gignoux, C.R.; Highland, H.M.; Patel, Y.M.; Sorokin, E.P.; Avery, C.L.; et al. Genetic analyses of diverse populations improves discovery for complex traits. Nature 2019, 570, 514–518. [Google Scholar] [CrossRef]
- Gaudier, E.; Rival, M.; Buisine, M.P.; Robineau, I.; Hoebler, C. Butyrate enemas upregulate Muc genes expression but decrease adherent mucus thickness in mice colon. Physiol. Res. 2009, 58, 111–119. [Google Scholar] [CrossRef]
- Marin, F.; Bonet, C.; Munoz, X.; Garcia, N.; Pardo, M.L.; Ruiz-Liso, J.M.; Alonso, P.; Capella, G.; Sanz-Anquela, J.M.; Gonzalez, C.A.; et al. Genetic variation in MUC1, MUC2 and MUC6 genes and evolution of gastric cancer precursor lesions in a long-term follow-up in a high-risk area in Spain. Carcinogenesis 2012, 33, 1072–1080. [Google Scholar] [CrossRef] [Green Version]
- Caruso, R.; Lo, B.C.; Núñez, G. Host-microbiota interactions in inflammatory bowel disease. Nat. Rev. Immunol. 2020, 20, 411–426. [Google Scholar] [CrossRef]
- Valette, K.; Li, Z.; Bon-Baret, V.; Chignon, A.; Bérubé, J.C.; Eslami, A.; Lamothe, J.; Gaudreault, N.; Joubert, P.; Obeidat, M.; et al. Prioritization of candidate causal genes for asthma in susceptibility loci derived from UK Biobank. Commun. Biol. 2021, 4, 700. [Google Scholar] [CrossRef]
- Tong, M.; McHardy, I.; Ruegger, P.; Goudarzi, M.; Kashyap, P.C.; Haritunians, T.; Li, X.; Graeber, T.G.; Schwager, E.; Huttenhower, C.; et al. Reprograming of gut microbiome energy metabolism by the FUT2 Crohn’s disease risk polymorphism. ISME J. 2014, 8, 2193–2206. [Google Scholar] [CrossRef] [Green Version]
- Kachuri, L.; Jeon, S.; DeWan, A.T.; Metayer, C.; Ma, X.; Witte, J.S.; Chiang, C.W.K.; Wiemels, J.L.; de Smith, A.J. Genetic determinants of blood-cell traits influence susceptibility to childhood acute lymphoblastic leukemia. Am. J. Hum. Genet. 2021, 108, 1823–1835. [Google Scholar] [CrossRef]
- Otonkoski, T.; Jiao, H.; Kaminen-Ahola, N.; Tapia-Paez, I.; Ullah, M.S.; Parton, L.E.; Schuit, F.; Quintens, R.; Sipilä, I.; Mayatepek, E.; et al. Physical Exercise–Induced Hypoglycemia Caused by Failed Silencing of Monocarboxylate Transporter 1 in Pancreatic β Cells. Am. J. Hum. Genet. 2007, 81, 467–474. [Google Scholar] [CrossRef] [Green Version]
- Felsky, D.; Roostaei, T.; Nho, K.; Risacher, S.L.; Bradshaw, E.M.; Petyuk, V.; Schneider, J.A.; Saykin, A.; Bennett, D.A.; de Jager, P.L. Neuropathological correlates and genetic architecture of microglial activation in elderly human brain. Nat. Commun. 2019, 10, 409. [Google Scholar] [CrossRef] [Green Version]
- Hysi, P.G.; Choquet, H.; Khawaja, A.P.; Wojciechowski, R.; Tedja, M.S.; Yin, J.; Simcoe, M.J.; Patasova, K.; Mahroo, O.A.; Thai, K.K.; et al. Meta-analysis of 542,934 subjects of European ancestry identifies new genes and mechanisms predisposing to refractive error and myopia. Nat. Genet. 2020, 52, 401–407. [Google Scholar] [CrossRef]
- Kichaev, G.; Bhatia, G.; Loh, P.R.; Gazal, S.; Burch, K.; Freund, M.K.; Schoech, A.; Pasaniuc, B.; Price, A.L. Leveraging Polygenic Functional Enrichment to Improve GWAS Power. Am. J. Hum. Genet. 2019, 104, 65–75. [Google Scholar] [CrossRef] [Green Version]
- You, D.; Wang, D.; Wu, Y.; Chen, X.; Shao, F.; Wei, Y.; Zhang, R.; Lange, T.; Ma, H.; Xu, H.; et al. Associations of genetic risk, BMI trajectories, and the risk of non-small cell lung cancer: A population-based cohort study. BMC Med. 2022, 20, 203. [Google Scholar] [CrossRef]
- Smith, S.M.; Douaud, G.; Chen, W.; Hanayik, T.; Alfaro-Almagro, F.; Sharp, K.; Elliott, L.T. An expanded set of genome-wide association studies of brain imaging phenotypes in UK Biobank. Nat. Neurosci. 2021, 24, 737–745. [Google Scholar] [CrossRef] [PubMed]
- Pulit, S.L.; Stoneman, C.; Morris, A.P.; Wood, A.R.; Glastonbury, C.A.; Tyrrell, J.; Yengo, L.; Ferreira, T.; Marouli, E.; Ji, Y.; et al. Meta-analysis of genome-wide association studies for body fat distribution in 694 649 individuals of European ancestry. Hum. Mol. Genet. 2019, 28, 166–174. [Google Scholar] [CrossRef] [Green Version]
- Miranda-Lora, A.L.; Cruz, M.; Molina-Díaz, M.; Gutiérrez, J.; Flores-Huerta, S.; Klünder-Klünder, M. Associations of common variants in the SLC16A11, TCF7L2, and ABCA1 genes with pediatric-onset type 2 diabetes and related glycemic traits in families: A case-control and case-parent trio study. Pediatric Diabetes 2017, 18, 824–831. [Google Scholar] [CrossRef] [PubMed]
- Suhre, K.; Shin, S.-Y.; Petersen, A.-K.; Mohney, R.P.; Meredith, D.; Wägele, B.; Altmaier, E.; Deloukas, P.; Erdmann, J.; Grundberg, E.; et al. Human metabolic individuality in biomedical and pharmaceutical research. Nature 2011, 477, 54–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ganapathy, V.; Thangaraju, M.; Gopal, E.; Martin, P.M.; Itagaki, S.; Miyauchi, S.; Prasad, P.D. Sodium-coupled Monocarboxylate Transporters in Normal Tissues and in Cancer. AAPS J. 2008, 10, 193–199. [Google Scholar] [CrossRef]
- Christakoudi, S.; Evangelou, E.; Riboli, E.; Tsilidis, K.K. GWAS of allometric body-shape indices in UK Biobank identifies loci suggesting associations with morphogenesis, organogenesis, adrenal cell renewal and cancer. Sci. Rep. 2021, 11, 10688. [Google Scholar] [CrossRef]
- Ahola-Olli, A.V.; Würtz, P.; Havulinna, A.S.; Aalto, K.; Pitkänen, N.; Lehtimäki, T.; Kähönen, M.; Lyytikäinen, L.-P.; Raitoharju, E.; Seppälä, I.; et al. Genome-wide Association Study Identifies 27 Loci Influencing Concentrations of Circulating Cytokines and Growth Factors. Am. J. Hum. Genet. 2017, 100, 40–50. [Google Scholar] [CrossRef] [Green Version]
- Chai, J.T.; Digby, J.E.; Choudhury, R.P. GPR109A and Vascular Inflammation. Curr. Atheroscler. Rep. 2013, 15, 325. [Google Scholar] [CrossRef] [Green Version]
- Richardson, T.G.; Leyden, G.M.; Wang, Q.; Bell, J.A.; Elsworth, B.; Davey Smith, G.; Holmes, M.V. Characterising metabolomic signatures of lipid-modifying therapies through drug target mendelian randomisation. PLoS Biol. 2022, 20, e3001547. [Google Scholar] [CrossRef]
- Vuckovic, D.; Bao, E.L.; Akbari, P.; Lareau, C.A.; Mousas, A.; Jiang, T.; Chen, M.-H.; Raffield, L.M.; Tardaguila, M.; Huffman, J.E.; et al. The Polygenic and Monogenic Basis of Blood Traits and Diseases. Cell 2020, 182, 1214–1231.e1211. [Google Scholar] [CrossRef]
- Lührs, H.; Gerke, T.; Müller, J.G.; Melcher, R.; Schauber, J.; Boxberge, F.; Scheppach, W.; Menzel, T. Butyrate inhibits NF-kappaB activation in lamina propria macrophages of patients with ulcerative colitis. Scand. J. Gastroenterol. 2002, 37, 458–466. [Google Scholar] [CrossRef]
- Bonfiglio, F.; Liu, X.; Smillie, C.; Pandit, A.; Kurilshikov, A.; Bacigalupe, R.; Zheng, T.; Nim, H.; Garcia-Etxebarria, K.; Bujanda, L.; et al. GWAS of stool frequency provides insights into gastrointestinal motility and irritable bowel syndrome. Cell Genom. 2021, 1, 100069. [Google Scholar] [CrossRef]
- Puhl, H.L., III; Won, Y.-J.; Lu, V.B.; Ikeda, S.R. Human GPR42 is a transcribed multisite variant that exhibits copy number polymorphism and is functional when heterologously expressed. Sci. Rep. 2015, 5, 12880. [Google Scholar] [CrossRef] [Green Version]
- De Faria, A.P.; Ritter, A.M.; Sabbatini, A.R.; Modolo, R.; Moreno, H. Effects of leptin and leptin receptor SNPs on clinical- and metabolic-related traits in apparent treatment-resistant hypertension. Blood Press. 2017, 26, 74–80. [Google Scholar] [CrossRef]
- Yaghootkar, H.; Zhang, Y.; Spracklen, C.N.; Karaderi, T.; Huang, L.O.; Bradfield, J.; Schurmann, C.; Fine, R.S.; Preuss, M.H.; Kutalik, Z.; et al. Genetic Studies of Leptin Concentrations Implicate Leptin in the Regulation of Early Adiposity. Diabetes 2020, 69, 2806–2818. [Google Scholar] [CrossRef]
- Park, K.S.; Shin, H.D.; Park, B.L.; Cheong, H.S.; Cho, Y.M.; Lee, H.K.; Lee, J.-Y.; Lee, J.-K.; Oh, B.; Kimm, K. Polymorphisms in the leptin receptor (LEPR)—Putative association with obesity and T2DM. J. Hum. Genet. 2006, 51, 85–91. [Google Scholar] [CrossRef] [Green Version]
- Jeon, J.-P.; Shim, S.-M.; Nam, H.-Y.; Ryu, G.-M.; Hong, E.-J.; Kim, H.-L.; Han, B.-G. Copy number variation at leptin receptor gene locus associated with metabolic traits and the risk of type 2 diabetes mellitus. BMC Genom. 2010, 11, 426. [Google Scholar] [CrossRef] [Green Version]
- Astle, W.J.; Elding, H.; Jiang, T.; Allen, D.; Ruklisa, D.; Mann, A.L.; Mead, D.; Bouman, H.; Riveros-Mckay, F.; Kostadima, M.A.; et al. The Allelic Landscape of Human Blood Cell Trait Variation and Links to Common Complex Disease. Cell 2016, 167, 1415–1429.e1419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davenport, E.R.; Cusanovich, D.A.; Michelini, K.; Barreiro, L.B.; Ober, C.; Gilad, Y. Genome-Wide Association Studies of the Human Gut Microbiota. PLoS ONE 2015, 10, e0140301. [Google Scholar] [CrossRef]
- Park, J.; Kim, M.; Kang, S.G.; Jannasch, A.H.; Cooper, B.; Patterson, J.; Kim, C.H. Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR–S6K pathway. Mucosal Immunol. 2015, 8, 80–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tan, J.; McKenzie, C.; Potamitis, M.; Thorburn, A.N.; Mackay, C.R.; Macia, L. The Role of Short-Chain Fatty Acids in Health and Disease. In Advances in Immunology; Elsevier: Amsterdam, The Netherlands, 2014; Volume 121, pp. 91–112. [Google Scholar]
- Broek, T.J.V.D.; Bakker, G.C.M.; Rubingh, C.M.; Bijlsma, S.; Stroeve, J.H.M.; Ommen, B.V.; Erk, M.J.V.; Wopereis, S. Ranges of phenotypic flexibility in healthy subjects. Genes Nutr. 2017, 12, 32. [Google Scholar] [CrossRef] [PubMed]
- Stroeve, J.H.M.; Wietmarschen, H.V.; Kremer, B.H.A.; Ommen, B.V.; Wopereis, S. Phenotypic flexibility as a measure of health: The optimal nutritional stress response test. Genes Nutr. 2015, 10, 12–33. [Google Scholar] [CrossRef] [Green Version]
- Van Ommen, B.; van der Greef, J.; Ordovas, J.M.; Daniel, H. Phenotypic flexibility as key factor in the human nutrition and health relationship. Genes Nutr. 2014, 9, 423. [Google Scholar] [CrossRef] [Green Version]
- Zhu, Z.; Cao, F.; Li, X. Epigenetic Programming and Fetal Metabolic Programming. Front. Endocrinol. 2019, 10, 00764. [Google Scholar] [CrossRef] [Green Version]
- Lumey, L.; Stein, A.D.; Kahn, H.S.; van der Pal-De Bruin, K.M.; Blauw, G.; Zybert, P.A.; Susser, E.S. Cohort Profile: The Dutch Hunger Winter Families Study. Int. J. Epidemiol. 2007, 36, 1196–1204. [Google Scholar] [CrossRef]
- Duncan, B.B.; Schmidt, M.I.S.; Pankow, J.S.; Ballantyne, C.M.; Couper, D.; Vigo, A.; Hoogeveen, R.; Folsom, A.R.; Heiss, G. Low-Grade Systemic Inflammation and the Development of Type 2 Diabetes. Diabetes 2003, 52, 1799–1805. [Google Scholar] [CrossRef] [Green Version]
- Wacklin, P.; Tuimala, J.; Nikkilä, J.; Sebastian, T.; Mäkivuokko, H.; Alakulppi, N.; Laine, P.; Rajilic-Stojanovic, M.; Paulin, L.; de Vos, W.M.; et al. Faecal Microbiota Composition in Adults Is Associated with the FUT2 Gene Determining the Secretor Status. PLoS ONE 2014, 9, e94863. [Google Scholar] [CrossRef] [Green Version]
- Barzilai, N.; Huffman, D.M.; Muzumdar, R.H.; Bartke, A. The Critical Role of Metabolic Pathways in Aging. Diabetes 2012, 61, 1315–1322. [Google Scholar] [CrossRef] [Green Version]
- Henry, C.J.; Kaur, B.; Quek, R.Y.C. Chrononutrition in the management of diabetes. Nutr. Diabetes 2020, 10, 6. [Google Scholar] [CrossRef] [Green Version]
- Papakonstantinou, E.; Oikonomou, C.; Nychas, G.; Dimitriadis, G.D. Effects of Diet, Lifestyle, Chrononutrition and Alternative Dietary Interventions on Postprandial Glycemia and Insulin Resistance. Nutrients 2022, 14, 823. [Google Scholar] [CrossRef]
- Katsi, V.; Papakonstantinou, I.P.; Soulaidopoulos, S.; Katsiki, N.; Tsioufis, K. Chrononutrition in Cardiometabolic Health. J. Clin. Med. 2022, 11, 296. [Google Scholar] [CrossRef] [PubMed]
- Mohd Azmi, N.A.S.; Juliana, N.; Mohd Fahmi Teng, N.I.; Azmani, S.; Das, S.; Effendy, N. Consequences of Circadian Disruption in Shift Workers on Chrononutrition and their Psychosocial Well-Being. Int. J. Environ. Res. Public Health 2020, 17, 2043. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, H.-K.; Chijiki, H.; Nanba, T.; Ozaki, M.; Sasaki, H.; Takahashi, M.; Shibata, S. Ingestion of Helianthus tuberosus at Breakfast Rather Than at Dinner is More Effective for Suppressing Glucose Levels and Improving the Intestinal Microbiota in Older Adults. Nutrients 2020, 12, 3035. [Google Scholar] [CrossRef] [PubMed]
- Sarkar, A.; Kuehl, M.N.; Alman, A.C.; Burkhardt, B.R. Linking the oral microbiome and salivary cytokine abundance to circadian oscillations. Sci. Rep. 2021, 11, 2658. [Google Scholar] [CrossRef] [PubMed]
- Barnard, N.D.; Kahleova, H.; Holtz, D.N.; del Aguila, F.; Neola, M.; Crosby, L.M.; Holubkov, R. The Women’s Study for the Alleviation of Vasomotor Symptoms (WAVS): A randomized, controlled trial of a plant-based diet and whole soybeans for postmenopausal women. Menopause 2021, 28, 1150–1156. [Google Scholar] [CrossRef]
- Zhao, H.; Yang, A.; Mao, L.; Quan, Y.; Cui, J.; Sun, Y. Association Between Dietary Fiber Intake and Non-alcoholic Fatty Liver Disease in Adults. Front. Nutr. 2020, 7, 593735. [Google Scholar] [CrossRef]
- Nova, E.; Gómez-Martinez, S.; González-Soltero, R. The Influence of Dietary Factors on the Gut Microbiota. Microorganisms 2022, 10, 1368. [Google Scholar] [CrossRef]
- Delgado, S.; Ruas-Madiedo, P.; Suárez, A.; Mayo, B. Interindividual Differences in Microbial Counts and Biochemical-Associated Variables in the Feces of Healthy Spanish Adults. Dig. Dis. Sci. 2006, 51, 737–743. [Google Scholar] [CrossRef] [Green Version]
- Salazar, N.; Dewulf, E.M.; Neyrinck, A.M.; Bindels, L.B.; Cani, P.D.; Mahillon, J.; de Vos, W.M.; Thissen, J.-P.; Gueimonde, M.; de los Reyes-Gavilán, C.G.; et al. Inulin-type fructans modulate intestinal Bifidobacterium species populations and decrease fecal short-chain fatty acids in obese women. Clin. Nutr. 2015, 34, 501–507. [Google Scholar] [CrossRef]
- Yang, J.; Keshavarzian, A.; Rose, D.J. Impact of dietary fiber fermentation from cereal grains on metabolite production by the fecal microbiota from normal weight and obese individuals. J. Med. Food 2013, 16, 862–867. [Google Scholar] [CrossRef]
- Miele, L.; Giorgio, V.; Alberelli, M.A.; de Candia, E.; Gasbarrini, A.; Grieco, A. Impact of Gut Microbiota on Obesity, Diabetes, and Cardiovascular Disease Risk. Curr. Cardiol. Rep. 2015, 17, 120. [Google Scholar] [CrossRef]
- Gonzalez, F.J.; Jiang, C.; Patterson, A.D. An Intestinal Microbiota-Farnesoid X Receptor Axis Modulates Metabolic Disease. Gastroenterology 2016, 151, 845–859. [Google Scholar] [CrossRef] [Green Version]
- Jiang, C.; Xie, C.; Li, F.; Zhang, L.; Nichols, R.G.; Krausz, K.W.; Cai, J.; Qi, Y.; Fang, Z.Z.; Takahashi, S.; et al. Intestinal farnesoid X receptor signaling promotes nonalcoholic fatty liver disease. J. Clin. Investig. 2015, 125, 386–402. [Google Scholar] [CrossRef] [Green Version]
- Crusell, M.K.W.; Hansen, T.H.; Nielsen, T.; Allin, K.H.; Rühlemann, M.C.; Damm, P.; Vestergaard, H.; Rørbye, C.; Jørgensen, N.R.; Christiansen, O.B.; et al. Gestational diabetes is associated with change in the gut microbiota composition in third trimester of pregnancy and postpartum. Microbiome 2018, 6, 89. [Google Scholar] [CrossRef]
- Li, Q.; Chang, Y.; Zhang, K.; Chen, H.; Tao, S.; Zhang, Z. Implication of the gut microbiome composition of type 2 diabetic patients from northern China. Sci. Rep. 2020, 10, 5450. [Google Scholar] [CrossRef]
- Cunningham, A.L.; Stephens, J.W.; Harris, D.A. Gut microbiota influence in type 2 diabetes mellitus (T2DM). Gut Pathog. 2021, 13, 50. [Google Scholar] [CrossRef]
- Torun, A.; Hupalowska, A.; Trzonkowski, P.; Kierkus, J.; Pyrzynska, B. Intestinal Microbiota in Common Chronic Inflammatory Disorders Affecting Children. Front. Immunol. 2021, 12, 642166. [Google Scholar] [CrossRef]
- Kelly, C.J.; Zheng, L.; Campbell, E.L.; Saeedi, B.; Scholz, C.C.; Bayless, A.J.; Wilson, K.E.; Glover, L.E.; Kominsky, D.J.; Magnuson, A.; et al. Crosstalk between Microbiota-Derived Short-Chain Fatty Acids and Intestinal Epithelial HIF Augments Tissue Barrier Function. Cell Host Microbe 2015, 17, 662–671. [Google Scholar] [CrossRef] [Green Version]
- Raman, M.; Ahmed, I.; Gillevet, P.M.; Probert, C.S.; Ratcliffe, N.M.; Smith, S.; Greenwood, R.; Sikaroodi, M.; Lam, V.; Crotty, P.; et al. Fecal microbiome and volatile organic compound metabolome in obese humans with nonalcoholic fatty liver disease. Clin. Gastroenterol. Hepatol. 2013, 11, 868–875.e3. [Google Scholar] [CrossRef]
- Bajaj, J.S. Alcohol, liver disease and the gut microbiota. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 235–246. [Google Scholar] [CrossRef]
- Fernández, J.; Redondo-Blanco, S.; Gutiérrez-del-Río, I.; Miguélez, E.M.; Villar, C.J.; Lombó, F. Colon microbiota fermentation of dietary prebiotics towards short-chain fatty acids and their roles as anti-inflammatory and antitumour agents: A review. J. Funct. Foods 2016, 25, 511–522. [Google Scholar] [CrossRef]
- Gagnière, J.; Raisch, J.; Veziant, J.; Barnich, N.; Bonnet, R.; Buc, E.; Bringer, M.A.; Pezet, D.; Bonnet, M. Gut microbiota imbalance and colorectal cancer. World J. Gastroenterol. 2016, 22, 501–518. [Google Scholar] [CrossRef] [PubMed]
- Scher, J.U.; Ubeda, C.; Artacho, A.; Attur, M.; Isaac, S.; Reddy, S.M.; Marmon, S.; Neimann, A.; Brusca, S.; Patel, T.; et al. Decreased bacterial diversity characterizes the altered gut microbiota in patients with psoriatic arthritis, resembling dysbiosis in inflammatory bowel disease. Arthritis Rheumatol. 2015, 67, 128–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kalliomäki, M.; Salminen, S.; Arvilommi, H.; Kero, P.; Koskinen, P.; Isolauri, E. Probiotics in primary prevention of atopic disease: A randomised placebo-controlled trial. Lancet 2001, 357, 1076–1079. [Google Scholar] [CrossRef] [PubMed]
- Weston, S.; Halbert, A.; Richmond, P.; Prescott, S.L. Effects of probiotics on atopic dermatitis: A randomised controlled trial. Arch. Dis. Child. 2005, 90, 892–897. [Google Scholar] [CrossRef]
- Łoś-Rychalska, E.; Gołębiewski, M.; Grzybowski, T.; Rogalla-Ładniak, U.; Krogulska, A. The microbiome and its impact on food allergy and atopic dermatitis in children. Adv. Dermatol. Allergol. 2020, 37, 641–650. [Google Scholar] [CrossRef]
- Garcia-Gutierrez, E.; Narbad, A.; Rodríguez, J.M. Autism Spectrum Disorder Associated with Gut Microbiota at Immune, Metabolomic, and Neuroactive Level. Front. Neurosci. 2020, 14, 578666. [Google Scholar] [CrossRef]
- Sanchez-Rodriguez, E.; Egea-Zorrilla, A.; Plaza-Díaz, J.; Aragón-Vela, J.; Muñoz-Quezada, S.; Tercedor-Sánchez, L.; Abadia-Molina, F. The Gut Microbiota and Its Implication in the Development of Atherosclerosis and Related Cardiovascular Diseases. Nutrients 2020, 12, 605. [Google Scholar] [CrossRef] [Green Version]
- Georgia-Eirini, D.; Athina, S.; Wim, V.B.; Christos, K.; Theodoros, C. Natural Products from Mediterranean Diet: From Anti-hyperlipidemic Agents to Dietary Epigenetic Modulators. Curr. Pharm. Biotechnol. 2019, 20, 825–844. [Google Scholar] [CrossRef]
- Nie, J.; Xie, L.; Zhao, B.X.; Li, Y.; Qiu, B.; Zhu, F.; Li, G.F.; He, M.; Wang, Y.; Wang, B.; et al. Serum Trimethylamine N-Oxide Concentration Is Positively Associated with First Stroke in Hypertensive Patients. Stroke 2018, 49, 2021–2028. [Google Scholar] [CrossRef]
- Koeth, R.A.; Wang, Z.; Levison, B.S.; Buffa, J.A.; Org, E.; Sheehy, B.T.; Britt, E.B.; Fu, X.; Wu, Y.; Li, L.; et al. Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat. Med. 2013, 19, 576–585. [Google Scholar] [CrossRef] [Green Version]
- Koeth, R.A.; Lam-Galvez, B.R.; Kirsop, J.; Wang, Z.; Levison, B.S.; Gu, X.; Copeland, M.F.; Bartlett, D.; Cody, D.B.; Dai, H.J.; et al. l-Carnitine in omnivorous diets induces an atherogenic gut microbial pathway in humans. J. Clin. Investig. 2018, 129, 373–387. [Google Scholar] [CrossRef] [Green Version]
- Ascher, S.; Reinhardt, C. The gut microbiota: An emerging risk factor for cardiovascular and cerebrovascular disease. Eur. J. Immunol. 2018, 48, 564–575. [Google Scholar] [CrossRef] [Green Version]
- Elfil, M.; Kamel, S.; Kandil, M.; Koo, B.B.; Schaefer, S.M. Implications of the Gut Microbiome in Parkinson’s Disease. Mov. Disord. 2020, 35, 921–933. [Google Scholar] [CrossRef]
- Nag, N.; Jelinek, G.A. More Research Is Needed on Lifestyle Behaviors That Influence Progression of Parkinson’s Disease. Front. Neurol. 2019, 10, 452. [Google Scholar] [CrossRef]
- Hertel, J.; Harms, A.C.; Heinken, A.; Baldini, F.; Thinnes, C.C.; Glaab, E.; Vasco, D.A.; Pietzner, M.; Stewart, I.D.; Wareham, N.J.; et al. Integrated Analyses of Microbiome and Longitudinal Metabolome Data Reveal Microbial-Host Interactions on Sulfur Metabolism in Parkinson’s Disease. Cell Rep. 2019, 29, 1767–1777. [Google Scholar] [CrossRef] [Green Version]
- González-Sanmiguel, J.; Schuh, C.M.A.P.; Muñoz-Montesino, C.; Contreras-Kallens, P.; Aguayo, L.G.; Aguayo, S. Complex Interaction between Resident Microbiota and Misfolded Proteins: Role in Neuroinflammation and Neurodegeneration. Cells 2020, 9, 2476. [Google Scholar] [CrossRef] [PubMed]
- Keshavarzian, A.; Green, S.J.; Engen, P.A.; Voigt, R.M.; Naqib, A.; Forsyth, C.B.; Mutlu, E.; Shannon, K.M. Colonic bacterial composition in Parkinson’s disease. Mov. Disord. 2015, 30, 1351–1360. [Google Scholar] [CrossRef] [PubMed]
- O’Donovan, S.M.; Crowley, E.K.; Brown, J.R.M.; O’Sullivan, O.; O’Leary, O.F.; Timmons, S.; Nolan, Y.M.; Clarke, D.J.; Hyland, N.P.; Joyce, S.A.; et al. Nigral overexpression of α-synuclein in a rat Parkinson’s disease model indicates alterations in the enteric nervous system and the gut microbiome. Neurogastroenterol. Motil. 2020, 32, e13726. [Google Scholar] [CrossRef] [PubMed]
- Singh, Y.; Trautwein, C.; Dhariwal, A.; Salker, M.S.; Alauddin, M.; Zizmare, L.; Pelzl, L.; Feger, M.; Admard, J.; Casadei, N.; et al. DJ-1 (Park7) affects the gut microbiome, metabolites and the development of innate lymphoid cells (ILCs). Sci. Rep. 2020, 10, 16131. [Google Scholar] [CrossRef] [PubMed]
- Seidl, S.E.; Santiago, J.A.; Bilyk, H.; Potashkin, J.A. The emerging role of nutrition in Parkinson’s disease. Front. Aging Neurosci. 2014, 6, 36. [Google Scholar] [CrossRef] [Green Version]
- Govindarajulu, M.; Pinky, P.D.; Steinke, I.; Bloemer, J.; Ramesh, S.; Kariharan, T.; Rella, R.T.; Bhattacharya, S.; Dhanasekaran, M.; Suppiramaniam, V.; et al. Gut Metabolite TMAO Induces Synaptic Plasticity Deficits by Promoting Endoplasmic Reticulum Stress. Front. Mol. Neurosci. 2020, 13, 138. [Google Scholar] [CrossRef]
- Kwon, H.S.; Koh, S.-H. Neuroinflammation in neurodegenerative disorders: The roles of microglia and astrocytes. Transl. Neurodegener. 2020, 9, 42. [Google Scholar] [CrossRef]
- Milne, G.L.; Musiek, E.S.; Morrow, J.D. F2-isoprostanes as markers of oxidative stress in vivo: An overview. Biomarkers 2005, 10 (Suppl. 1), S10–S23. [Google Scholar] [CrossRef]
- Zhang, C.; Nestorova, G.; Rissman, R.A.; Feng, J. Detection and quantification of 8-hydroxy-2′-deoxyguanosine in Alzheimer’s transgenic mouse urine using capillary electrophoresis. Electrophoresis 2013, 34, 2268–2274. [Google Scholar] [CrossRef]
- Greenwood-Van Meerveld, B.; Johnson, A.C.; Grundy, D. Gastrointestinal Physiology and Function; Springer International Publishing: Berlin/Heidelberg, Germany, 2017; pp. 1–16. [Google Scholar] [CrossRef]
- Recio, C.; Lucy, D.; Iveson, P.; Iqbal, A.J.; Valaris, S.; Wynne, G.; Russell, A.J.; Choudhury, R.P.; O’Callaghan, C.; Monaco, C.; et al. The Role of Metabolite-Sensing G Protein-Coupled Receptors in Inflammation and Metabolic Disease. Antioxid. Redox Signal. 2018, 29, 237–256. [Google Scholar] [CrossRef]
- Fallingborg, J. Intraluminal pH of the human gastrointestinal tract. Dan. Med. Bull. 1999, 46, 183–196. [Google Scholar]
- Belenguer, A.; Duncan, S.H.; Calder, A.G.; Holtrop, G.; Louis, P.; Lobley, G.E.; Flint, H.J. Two Routes of Metabolic Cross-Feeding between Bifidobacterium adolescentis and Butyrate-Producing Anaerobes from the Human Gut. Appl. Environ. Microbiol. 2006, 72, 3593–3599. [Google Scholar] [CrossRef] [Green Version]
- Heinken, A.; Thiele, I. Anoxic Conditions Promote Species-Specific Mutualism between Gut Microbes In Silico. Appl. Environ. Microbiol. 2015, 81, 4049–4061. [Google Scholar] [CrossRef] [Green Version]
- Den Besten, G.; Lange, K.; Havinga, R.; van Dijk, T.H.; Gerding, A.; van Eunen, K.; Müller, M.; Groen, A.K.; Hooiveld, G.J.; Bakker, B.M.; et al. Gut-derived short-chain fatty acids are vividly assimilated into host carbohydrates and lipids. Am. J. Physiol. Gastrointest. Liver Physiol. 2013, 305, G900–G910. [Google Scholar] [CrossRef]
- Jung, T.-H.; Park, J.H.; Jeon, W.-M.; Han, K.-S. Butyrate modulates bacterial adherence on LS174T human colorectal cells by stimulating mucin secretion and MAPK signaling pathway. Nutr. Res. Pract. 2015, 9, 343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peng, L.; Li, Z.-R.; Green, R.S.; Holzman, I.R.; Lin, J. Butyrate Enhances the Intestinal Barrier by Facilitating Tight Junction Assembly via Activation of AMP-Activated Protein Kinase in Caco-2 Cell Monolayers. J. Nutr. 2009, 139, 1619–1625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Panpetch, W.; Hiengrach, P.; Nilgate, S.; Tumwasorn, S.; Somboonna, N.; Wilantho, A.; Chatthanathon, P.; Prueksapanich, P.; Leelahavanichkul, A. Additional Candida albicans administration enhances the severity of dextran sulfate solution induced colitis mouse model through leaky gut-enhanced systemic inflammation and gut-dysbiosis but attenuated by Lactobacillus rhamnosus L34. Gut Microbes 2020, 11, 465–480. [Google Scholar] [CrossRef]
- Dias, A.M.M.; Douhard, R.; Hermetet, F.; Regimbeau, M.; Lopez, T.E.; Gonzalez, D.; Masson, S.; Marcion, G.; Chaumonnot, K.; Uyanik, B.; et al. Lactobacillus stress protein GroEL prevents colonic inflammation. J. Gastroenterol. 2021, 56, 442–455. [Google Scholar] [CrossRef] [PubMed]
- Ichikawa, H.; Sakata, T. Effect of L-Lactic Acid, Short-Chain Fatty Acids, and pH in Cecal Infusate on Morphometric and Cell Kinetic Parameters of Rat Cecum. Dig. Dis. Sci. 1997, 42, 1598–1610. [Google Scholar] [CrossRef]
- Frugé, A.D.; Smith, K.S.; Riviere, A.J.; Tenpenny-Chigas, R.; Demark-Wahnefried, W.; Arthur, A.E.; Murrah, W.M.; van der Pol, W.J.; Jasper, S.L.; Morrow, C.D.; et al. A Dietary Intervention High in Green Leafy Vegetables Reduces Oxidative DNA Damage in Adults at Increased Risk of Colorectal Cancer: Biological Outcomes of the Randomized Controlled Meat and Three Greens (M3G) Feasibility Trial. Nutrients 2021, 13, 1220. [Google Scholar] [CrossRef]
- Lupton, J.R. Microbial degradation products influence colon cancer risk: The butyrate controversy. J. Nutr. 2004, 134, 479–482. [Google Scholar] [CrossRef] [Green Version]
- Donohoe, D.R.; Collins, L.B.; Wali, A.; Bigler, R.; Sun, W.; Bultman, S.J. The Warburg effect dictates the mechanism of butyrate-mediated histone acetylation and cell proliferation. Mol. Cell 2012, 48, 612–626. [Google Scholar] [CrossRef] [Green Version]
- Ternes, D.; Tsenkova, M.; Pozdeev, V.I.; Meyers, M.; Koncina, E.; Atatri, S.; Schmitz, M.; Karta, J.; Schmoetten, M.; Heinken, A.; et al. The gut microbial metabolite formate exacerbates colorectal cancer progression. Nat. Metab. 2022, 4, 458–475. [Google Scholar] [CrossRef]
- Venkatapoorna, C.; Ayine, P.; Parra, E.; Koenigs, T.; Phillips, M.; Babu, J.; Sandey, M.; Geetha, T. Association of Salivary Amylase (AMY1) Gene Copy Number with Obesity in Alabama Elementary School Children. Nutrients 2019, 11, 1379. [Google Scholar] [CrossRef] [Green Version]
- Lopera-Maya, E.A.; Kurilshikov, A.; van der Graaf, A.; Hu, S.; Andreu-Sánchez, S.; Chen, L.; Vila, A.V.; Gacesa, R.; Sinha, T.; Collij, V.; et al. Effect of host genetics on the gut microbiome in 7738 participants of the Dutch Microbiome Project. Nat. Genet. 2022, 54, 143–151. [Google Scholar] [CrossRef]
- Wen, R.; Gao, F.; Zhou, C.-J.; Jia, Y.-B. Polymorphisms in mucin genes in the development of gastric cancer. World J. Gastrointest. Oncol. 2015, 7, 328. [Google Scholar] [CrossRef]
- Braniste, V.; Al-Asmakh, M.; Kowal, C.; Anuar, F.; Abbaspour, A.; Toth, M.; Korecka, A.; Bakocevic, N.; Ng, L.G.; Kundu, P.; et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 2014, 6, 263ra158. [Google Scholar] [CrossRef] [Green Version]
- Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in Gut Microbiota Control Metabolic Endotoxemia-Induced Inflammation in High-Fat Diet–Induced Obesity and Diabetes in Mice. Diabetes 2008, 57, 1470–1481. [Google Scholar] [CrossRef] [Green Version]
- 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, 721. [Google Scholar] [CrossRef]
- Banan, A.; Fields, J.Z.; Farhadi, A.; Talmage, D.A.; Zhang, L.; Keshavarzian, A. Activation of delta-isoform of protein kinase C is required for oxidant-induced disruption of both the microtubule cytoskeleton and permeability barrier of intestinal epithelia. J. Pharmacol. Exp. Ther. 2002, 303, 17–28. [Google Scholar] [CrossRef]
- Jain, S.; Suzuki, T.; Seth, A.; Samak, G.; Rao, R. Protein kinase Cζ phosphorylates occludin and promotes assembly of epithelial tight junctions. Biochem. J. 2011, 437, 289–299. [Google Scholar] [CrossRef] [Green Version]
- Morrison, D.J.; Preston, T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 2016, 7, 189–200. [Google Scholar] [CrossRef] [Green Version]
- Bordin, M.; D’Atri, F.; Guillemot, L.; Citi, S. Histone deacetylase inhibitors up-regulate the expression of tight junction proteins. Mol. Cancer Res. 2004, 2, 692–701. [Google Scholar] [CrossRef]
- Sturgeon, C.; Fasano, A. Zonulin, a regulator of epithelial and endothelial barrier functions, and its involvement in chronic inflammatory diseases. Tissue Barriers 2016, 4, e1251384. [Google Scholar] [CrossRef] [Green Version]
- Feng, Y.; Wang, Y.; Wang, P.; Huang, Y.; Wang, F. Short-Chain Fatty Acids Manifest Stimulative and Protective Effects on Intestinal Barrier Function Through the Inhibition of NLRP3 Inflammasome and Autophagy. Cell. Physiol. Biochem. 2018, 49, 190–205. [Google Scholar] [CrossRef] [PubMed]
- König, J.; Wells, J.; Cani, P.D.; García-Ródenas, C.L.; MacDonald, T.; Mercenier, A.; Whyte, J.; Troost, F.; Brummer, R.J. Human Intestinal Barrier Function in Health and Disease. Clin. Transl. Gastroenterol. 2016, 7, e196. [Google Scholar] [CrossRef] [PubMed]
- Tajik, N.; Frech, M.; Schulz, O.; Schälter, F.; Lucas, S.; Azizov, V.; Dürholz, K.; Steffen, F.; Omata, Y.; Rings, A.; et al. Targeting zonulin and intestinal epithelial barrier function to prevent onset of arthritis. Nat. Commun. 2020, 11, 1995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sanos, S.L.; Bui, V.L.; Mortha, A.; Oberle, K.; Heners, C.; Johner, C.; Diefenbach, A. RORγt and commensal microflora are required for the differentiation of mucosal interleukin 22–producing NKp46+ cells. Nat. Immunol. 2009, 10, 83–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zheng, Y.; Valdez, P.A.; Danilenko, D.M.; Hu, Y.; Sa, S.M.; Gong, Q.; Abbas, A.R.; Modrusan, Z.; Ghilardi, N.; de Sauvage, F.J.; et al. Interleukin-22 mediates early host defense against attaching and effacing bacterial pathogens. Nat. Med. 2008, 14, 282–289. [Google Scholar] [CrossRef]
- Van Kaer, L.; Parekh, V.V.; Wu, L. Invariant natural killer T cells: Bridging innate and adaptive immunity. Cell Tissue Res. 2011, 343, 43–55. [Google Scholar] [CrossRef] [Green Version]
- Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; Deroos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef] [Green Version]
- Risnes, K.R.; Belanger, K.; Murk, W.; Bracken, M.B. Antibiotic Exposure by 6 Months and Asthma and Allergy at 6 Years: Findings in a Cohort of 1,401 US Children. Am. J. Epidemiol. 2011, 173, 310–318. [Google Scholar] [CrossRef] [Green Version]
- Ege, M.J. The Hygiene Hypothesis in the Age of the Microbiome. Ann. Am. Thorac. Soc. 2017, 14, S348–S353. [Google Scholar] [CrossRef]
- Vulevic, J.; Drakoularakou, A.; Yaqoob, P.; Tzortzis, G.; Gibson, G.R. Modulation of the fecal microflora profile and immune function by a novel trans-galactooligosaccharide mixture (B-GOS) in healthy elderly volunteers. Am. J. Clin. Nutr. 2008, 88, 1438–1446. [Google Scholar] [CrossRef]
- Enerson, B.E.; Drewes, L.R. Molecular features, regulation, and function of monocarboxylate transporters: Implications for drug delivery. J. Pharm. Sci. 2003, 92, 1531–1544. [Google Scholar] [CrossRef]
- Vijay, N.; Morris, M. Role of Monocarboxylate Transporters in Drug Delivery to the Brain. Curr. Pharm. Des. 2014, 20, 1487–1498. [Google Scholar] [CrossRef] [Green Version]
- Felmlee, M.A.; Jones, R.S.; Rodriguez-Cruz, V.; Follman, K.E.; Morris, M.E. Monocarboxylate Transporters (SLC16): Function, Regulation, and Role in Health and Disease. Pharmacol. Rev. 2020, 72, 466–485. [Google Scholar] [CrossRef]
- Rodriguez, J.E.R.; Garcia-Perdomo, H.A. Role of monocarboxylate transporters in the diagnosis, progression, prognosis, and treatment of prostate cancer. Turk. J. Urol. 2020, 46, 413–418. [Google Scholar] [CrossRef]
- Pinheiro, C.; Penna, V.; Morais-Santos, F.; Abrahão-Machado, L.F.; Ribeiro, G.; Curcelli, E.C.; Olivieri, M.V.; Morini, S.; Valença, I.; Ribeiro, D.; et al. Characterization of monocarboxylate transporters (MCTs) expression in soft tissue sarcomas: Distinct prognostic impact of MCT1 sub-cellular localization. J. Transl. Med. 2014, 12, 118. [Google Scholar] [CrossRef]
- Yu, S.; Wu, Y.; Li, C.; Qu, Z.; Lou, G.; Guo, X.; Ji, J.; Li, N.; Guo, M.; Zhang, M.; et al. Comprehensive analysis of the SLC16A gene family in pancreatic cancer via integrated bioinformatics. Sci. Rep. 2020, 10, 7315. [Google Scholar] [CrossRef]
- Gallagher-Colombo, S.; Maminishkis, A.; Tate, S.; Grunwald, G.B.; Philp, N.J. Modulation of MCT3 Expression during Wound Healing of the Retinal Pigment Epithelium. Investig. Opthalmology Vis. Sci. 2010, 51, 5343. [Google Scholar] [CrossRef]
- Halestrap, A.P. Monocarboxylic acid transport. Compr. Physiol. 2013, 3, 1611–1643. [Google Scholar] [CrossRef]
- Guan, X.; Rodriguez-Cruz, V.; Morris, M.E. Cellular Uptake of MCT1 Inhibitors AR-C155858 and AZD3965 and Their Effects on MCT-Mediated Transport of L-Lactate in Murine 4T1 Breast Tumor Cancer Cells. AAPS J. 2019, 21, 13. [Google Scholar] [CrossRef]
- Hinnebusch, B.F.; Meng, S.; Wu, J.T.; Archer, S.Y.; Hodin, R.A. The effects of short-chain fatty acids on human colon cancer cell phenotype are associated with histone hyperacetylation. J. Nutr. 2002, 132, 1012–1017. [Google Scholar] [CrossRef] [Green Version]
- Halestrap, A.P.; Wilson, M.C. The monocarboxylate transporter family-Role and regulation. IUBMB Life 2012, 64, 109–119. [Google Scholar] [CrossRef] [PubMed]
- Hooper, P.L.; Hooper, P.L.; Tytell, M.; Vígh, L. Xenohormesis: Health benefits from an eon of plant stress response evolution. Cell Stress Chaperones 2010, 15, 761–770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Halestrap, A.P. The SLC16 gene family—Structure, role and regulation in health and disease. Mol. Asp. Med. 2013, 34, 337–349. [Google Scholar] [CrossRef] [PubMed]
- Schutkowski, A.; Wege, N.; Stangl, G.I.; König, B. Tissue-Specific Expression of Monocarboxylate Transporters during Fasting in Mice. PLoS ONE 2014, 9, e112118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ullah, M.S.; Davies, A.J.; Halestrap, A.P. The plasma membrane lactate transporter MCT4, but not MCT1, is up-regulated by hypoxia through a HIF-1alpha-dependent mechanism. J. Biol. Chem. 2006, 281, 9030–9037. [Google Scholar] [CrossRef]
- Bergman, E.N. Energy contributions of volatile fatty acids from the gastrointestinal tract in various species. Physiol. Rev. 1990, 70, 567–590. [Google Scholar] [CrossRef] [Green Version]
- De Smit, S.M.; de Leeuw, K.D.; Buisman, C.J.N.; Strik, D.P.B.T.B. Continuous n-valerate formation from propionate and methanol in an anaerobic chain elongation open-culture bioreactor. Biotechnol. Biofuels 2019, 12, 132. [Google Scholar] [CrossRef]
- Zhu, X.; Zhou, Y.; Wang, Y.; Wu, T.; Li, X.; Li, D.; Tao, Y. Production of high-concentration n-caproic acid from lactate through fermentation using a newly isolated Ruminococcaceae bacterium CPB6. Biotechnol. Biofuels 2017, 10, 102. [Google Scholar] [CrossRef] [Green Version]
- Jørgensen, J.; Mortensen, P.B. Utilization of short-chain fatty acids by colonic mucosal tissue strips. A new method of assessing colonic mucosal metabolism. Scand. J. Gastroenterol. 2000, 35, 659–666. [Google Scholar] [CrossRef]
- Den Besten, G.; van Eunen, K.; Groen, A.K.; Venema, K.; Reijngoud, D.-J.; Bakker, B.M. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J. Lipid Res. 2013, 54, 2325–2340. [Google Scholar] [CrossRef] [Green Version]
- Freeman, H.J. Effects of differing concentrations of sodium butyrate on 1,2-dimethylhydrazine-induced rat intestinal neoplasia. Gastroenterology 1986, 91, 596–602. [Google Scholar] [CrossRef]
- McIntyre, A.; Gibson, P.R.; Young, G.P. Butyrate production from dietary fibre and protection against large bowel cancer in a rat model. Gut 1993, 34, 386–391. [Google Scholar] [CrossRef] [Green Version]
- Morris, B.J.; Willcox, D.C.; Donlon, T.A.; Willcox, B.J. FOXO3: A Major Gene for Human Longevity—A Mini-Review. Gerontology 2015, 61, 515–525. [Google Scholar] [CrossRef] [Green Version]
- Lee, G. The Balance of Th17 versus Treg Cells in Autoimmunity. Int. J. Mol. Sci. 2018, 19, 730. [Google Scholar] [CrossRef] [Green Version]
- Lu, L.; Barbi, J.; Pan, F. The regulation of immune tolerance by FOXP3. Nat. Rev. Immunol. 2017, 17, 703–717. [Google Scholar] [CrossRef]
- Janson, W.; Brandner, G.; Siegel, J. Butyrate modulates DNA-damage-induced p53 response by induction of p53-independent differentiation and apoptosis. Oncogene 1997, 15, 1395–1406. [Google Scholar] [CrossRef] [Green Version]
- Liu, B.; Qian, J.; Wang, Q.; Wang, F.; Ma, Z.; Qiao, Y. Butyrate protects rat liver against total hepatic ischemia reperfusion injury with bowel congestion. PLoS ONE 2014, 9, e106184. [Google Scholar] [CrossRef] [Green Version]
- Tsukahara, T.; Matsukawa, N.; Tomonaga, S.; Inoue, R.; Ushida, K.; Ochiai, K. High-sensitivity detection of short-chain fatty acids in porcine ileal, cecal, portal and abdominal blood by gas chromatography-mass spectrometry. Anim. Sci. J. 2014, 85, 494–498. [Google Scholar] [CrossRef]
- Yuan, F.; Tan, W.; Ren, H.; Yan, L.; Wang, Y.; Luo, H. The Effects of Short-Chain Fatty Acids on Rat Colonic Hypermotility Induced by Water Avoidance Stress. Drug Des. Dev. Ther. 2020, 14, 4671–4684. [Google Scholar] [CrossRef]
- Le Blay, G.; Blottière, H.M.; Ferrier, L.; le Foll, E.; Bonnet, C.; Galmiche, J.P.; Cherbut, C. Short-chain fatty acids induce cytoskeletal and extracellular protein modifications associated with modulation of proliferation on primary culture of rat intestinal smooth muscle cells. Dig. Dis. Sci. 2000, 45, 1623–1630. [Google Scholar] [CrossRef]
- Fei, F.; Guo, X.; Chen, Y.; Liu, X.; Tu, J.; Xing, J.; Chen, Z.; Ji, J.; He, X. Polymorphisms of monocarboxylate transporter genes are associated with clinical outcomes in patients with colorectal cancer. J. Cancer Res. Clin. Oncol. 2015, 141, 1095–1102. [Google Scholar] [CrossRef] [PubMed]
- De Vadder, F.; Kovatcheva-Datchary, P.; Goncalves, D.; Vinera, J.; Zitoun, C.; Duchampt, A.; Bäckhed, F.; Mithieux, G. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell 2014, 156, 84–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Soty, M.; Penhoat, A.; Amigo-Correig, M.; Vinera, J.; Sardella, A.; Vullin-Bouilloux, F.; Zitoun, C.; Houberdon, I.; Mithieux, G. A gut-brain neural circuit controlled by intestinal gluconeogenesis is crucial in metabolic health. Mol. Metab. 2015, 4, 106–117. [Google Scholar] [CrossRef] [PubMed]
- Poll, B.G.; Xu, J.; Jun, S.; Sanchez, J.; Zaidman, N.A.; He, X.; Lester, L.; Berkowitz, D.E.; Paolocci, N.; Gao, W.D.; et al. Acetate, a Short-Chain Fatty Acid, Acutely Lowers Heart Rate and Cardiac Contractility Along with Blood Pressure. J. Pharmacol. Exp. Ther. 2021, 377, 39–50. [Google Scholar] [CrossRef]
- Rajpal, A.; Dube, S.; Carvalho, F.; Simoes, A.R.; Figueiredo, A.; Basu, A.; Jones, J.; Basu, R. Effects of transaldolase exchange on estimates of gluconeogenesis in type 2 diabetes. Am. J. Physiol. Endocrinol. Metab. 2013, 305, E465–E474. [Google Scholar] [CrossRef] [PubMed]
- Chambers, E.S.; Viardot, A.; Psichas, A.; Morrison, D.J.; Murphy, K.G.; Zac-Varghese, S.E.; MacDougall, K.; Preston, T.; Tedford, C.; Finlayson, G.S.; et al. Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut 2015, 64, 1744–1754. [Google Scholar] [CrossRef] [Green Version]
- Wolever, T.M.; Brighenti, F.; Royall, D.; Jenkins, A.L.; Jenkins, D.J. Effect of rectal infusion of short chain fatty acids in human subjects. Am. J. Gastroenterol. 1989, 84, 1027–1033. [Google Scholar]
- Den Besten, G.; Bleeker, A.; Gerding, A.; van Eunen, K.; Havinga, R.; van Dijk, T.H.; Oosterveer, M.H.; Jonker, J.W.; Groen, A.K.; Reijngoud, D.-J.; et al. Short-Chain Fatty Acids Protect Against High-Fat Diet–Induced Obesity via a PPARγ-Dependent Switch From Lipogenesis to Fat Oxidation. Diabetes 2015, 64, 2398–2408. [Google Scholar] [CrossRef] [Green Version]
- Stienstra, R.; Duval, C.; Müller, M.; Kersten, S. PPARs, Obesity, and Inflammation. PPAR Res. 2007, 2007, 095974. [Google Scholar] [CrossRef] [Green Version]
- Kadowaki, T.; Hara, K.; Kubota, N.; Tobe, K.; Terauchi, Y.; Yamauchi, T.; Eto, K.; Kadowaki, H.; Noda, M.; Hagura, R.; et al. The role of PPARgamma in high-fat diet-induced obesity and insulin resistance. J. Diabetes Complicat. 2002, 16, 41–45. [Google Scholar] [CrossRef]
- Kurilshikov, A.; van den Munckhof, I.C.L.; Chen, L.; Bonder, M.J.; Schraa, K.; Rutten, J.H.W.; Riksen, N.P.; de Graaf, J.; Oosting, M.; Sanna, S.; et al. Gut Microbial Associations to Plasma Metabolites Linked to Cardiovascular Phenotypes and Risk. Circ. Res. 2019, 124, 1808–1820. [Google Scholar] [CrossRef]
- Müller, M.; Hernández, M.A.G.; Goossens, G.H.; Reijnders, D.; Holst, J.J.; Jocken, J.W.E.; van Eijk, H.; Canfora, E.E.; Blaak, E.E. Circulating but not faecal short-chain fatty acids are related to insulin sensitivity, lipolysis and GLP-1 concentrations in humans. Sci. Rep. 2019, 9, 12515. [Google Scholar] [CrossRef] [Green Version]
- Cummings, J.H.; Pomare, E.W.; Branch, W.J.; Naylor, C.P.; Macfarlane, G.T. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut 1987, 28, 1221–1227. [Google Scholar] [CrossRef] [Green Version]
- Peters, S.G.; Pomare, E.W.; Fisher, C.A. Portal and peripheral blood short chain fatty acid concentrations after caecal lactulose instillation at surgery. Gut 1992, 33, 1249–1252. [Google Scholar] [CrossRef] [Green Version]
- Wang, D.; Liu, C.D.; Tian, M.L.; Tan, C.Q.; Shu, G.; Jiang, Q.Y.; Zhang, L.; Yin, Y. Propionate promotes intestinal lipolysis and metabolic benefits via AMPK/LSD1 pathway in mice. J. Endocrinol. 2019, 243, 187–197. [Google Scholar] [CrossRef]
- Kong, D.; Schipper, L.; van Dijk, G. Distinct Effects of Short Chain Fatty Acids on Host Energy Balance and Fuel Homeostasis with Focus on Route of Administration and Host Species. Front. Neurosci. 2021, 15, 755845. [Google Scholar] [CrossRef]
- Trompette, A.; Gollwitzer, E.S.; Pattaroni, C.; Lopez-Mejia, I.C.; Riva, E.; Pernot, J.; Ubags, N.; Fajas, L.; Nicod, L.P.; Marsland, B.J. Dietary Fiber Confers Protection against Flu by Shaping Ly6c− Patrolling Monocyte Hematopoiesis and CD8+ T Cell Metabolism. Immunity 2018, 48, 992–1005.e8. [Google Scholar] [CrossRef] [Green Version]
- Todesco, T.; Zamboni, M.; Armellini, F.; Bissoli, L.; Turcato, E.; Piemonte, G.; Rao, A.V.; Jenkins, D.J.; Bosello, O. Plasma acetate levels in a group of obese diabetic, obese normoglycemic, and control subjects and their relationships with other blood parameters. Am. J. Gastroenterol. 1993, 88, 751–755. [Google Scholar]
- Dehghan, P.; Pourghassem Gargari, B.; Asghari Jafar-Abadi, M. Oligofructose-enriched inulin improves some inflammatory markers and metabolic endotoxemia in women with type 2 diabetes mellitus: A randomized controlled clinical trial. Nutrition 2014, 30, 418–423. [Google Scholar] [CrossRef]
- Samuel, B.S.; Shaito, A.; Motoike, T.; Rey, F.E.; Backhed, F.; Manchester, J.K.; Hammer, R.E.; Williams, S.C.; Crowley, J.; Yanagisawa, M.; et al. Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proc. Natl. Acad. Sci. USA 2008, 105, 16767–16772. [Google Scholar] [CrossRef] [Green Version]
- Song, S.Y.; Woo, H.J.; Bae, C.H.; Kim, Y.W.; Kim, Y.D. Expression of leptin receptor in nasal polyps: Leptin as a mucosecretagogue. Laryngoscope 2010, 120, 1046–1050. [Google Scholar] [CrossRef] [PubMed]
- Tadokoro, S.; Ide, S.; Tokuyama, R.; Umeki, H.; Tatehara, S.; Kataoka, S.; Satomura, K. Leptin promotes wound healing in the skin. PLoS ONE 2015, 10, e0121242. [Google Scholar] [CrossRef] [PubMed]
- Cerman, A.A.; Bozkurt, S.; Sav, A.; Tulunay, A.; Elbaşi, M.O.; Ergun, T. Serum leptin levels, skin leptin and leptin receptor expression in psoriasis. Br. J. Dermatol. 2008, 159, 820–826. [Google Scholar] [CrossRef] [PubMed]
- Zhu, K.J.; Zhang, C.; Li, M.; Zhu, C.Y.; Shi, G.; Fan, Y.M. Leptin levels in patients with psoriasis: A meta-analysis. Clin. Exp. Dermatol. 2013, 38, 478–483. [Google Scholar] [CrossRef] [PubMed]
- Sittipo, P.; Shim, J.-W.; Lee, Y. Microbial Metabolites Determine Host Health and the Status of Some Diseases. Int. J. Mol. Sci. 2019, 20, 5296. [Google Scholar] [CrossRef] [Green Version]
- Parada Venegas, D.; de la Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front. Immunol. 2019, 10, 277. [Google Scholar] [CrossRef] [Green Version]
- Vinolo, M.A.R.; Rodrigues, H.G.; Hatanaka, E.; Sato, F.T.; Sampaio, S.C.; Curi, R. Suppressive effect of short-chain fatty acids on production of proinflammatory mediators by neutrophils. J. Nutr. Biochem. 2011, 22, 849–855. [Google Scholar] [CrossRef]
- Vogt, J.A.; Wolever, T.M. Fecal acetate is inversely related to acetate absorption from the human rectum and distal colon. J. Nutr. 2003, 133, 3145–3148. [Google Scholar] [CrossRef] [Green Version]
- Consortium, T.H.M.P. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [Google Scholar] [CrossRef] [Green Version]
- Turnbaugh, P.J.; Ley, R.E.; Hamady, M.; Fraser-Liggett, C.M.; Knight, R.; Gordon, J.I. The Human Microbiome Project. Nature 2007, 449, 804–810. [Google Scholar] [CrossRef] [Green Version]
- Sivaprakasama, S.; Prasad, P.D.; Singh, N. Benefits of short-chain fatty acids and their receptors in inflammation and carcinogenesis. Pharmacol. Ther. 2016, 164, 144–151. [Google Scholar] [CrossRef] [Green Version]
- Wakade, C.; Chong, R.; Bradley, E.; Thomas, B.; Morgan, J. Upregulation of GPR109A in Parkinson’s Disease. PLoS ONE 2014, 9, e109818. [Google Scholar] [CrossRef] [Green Version]
- Bonder, M.J.; Kurilshikov, A.; Tigchelaar, E.F.; Mujagic, Z.; Imhann, F.; Vila, A.V.; Deelen, P.; Vatanen, T.; Schirmer, M.; Smeekens, S.P.; et al. The effect of host genetics on the gut microbiome. Nat. Genet. 2016, 48, 1407–1412. [Google Scholar] [CrossRef]
- Sánchez-Tapia, M.; Tovar, A.R.; Torres, N. Diet as Regulator of Gut Microbiota and its Role in Health and Disease. Arch. Med. Res. 2019, 50, 259–268. [Google Scholar] [CrossRef]
- Nicolas, G.R.; Chang, P.V. Deciphering the Chemical Lexicon of Host–Gut Microbiota Interactions. Trends Pharmacol. Sci. 2019, 40, 430–445. [Google Scholar] [CrossRef] [Green Version]
- Wolter, M.; Grant, E.T.; Boudaud, M.; Steimle, A.; Pereira, G.V.; Martens, E.C.; Desai, M.S. Leveraging diet to engineer the gut microbiome. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 885–902. [Google Scholar] [CrossRef]
- Sigman, M. Personalized medicine: What is it and what are the challenges? Fertil. Steril. 2018, 109, 944–945. [Google Scholar] [CrossRef]
- Frueh, F.W. Back to the future: Why randomized controlled trials cannot be the answer to pharmacogenomics and personalized medicine. Pharmacogenomics 2009, 10, 1077–1081. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Ramos Meyers, G.; Samouda, H.; Bohn, T. Short Chain Fatty Acid Metabolism in Relation to Gut Microbiota and Genetic Variability. Nutrients 2022, 14, 5361. https://doi.org/10.3390/nu14245361
Ramos Meyers G, Samouda H, Bohn T. Short Chain Fatty Acid Metabolism in Relation to Gut Microbiota and Genetic Variability. Nutrients. 2022; 14(24):5361. https://doi.org/10.3390/nu14245361
Chicago/Turabian StyleRamos Meyers, Guilherme, Hanen Samouda, and Torsten Bohn. 2022. "Short Chain Fatty Acid Metabolism in Relation to Gut Microbiota and Genetic Variability" Nutrients 14, no. 24: 5361. https://doi.org/10.3390/nu14245361
APA StyleRamos Meyers, G., Samouda, H., & Bohn, T. (2022). Short Chain Fatty Acid Metabolism in Relation to Gut Microbiota and Genetic Variability. Nutrients, 14(24), 5361. https://doi.org/10.3390/nu14245361