Search
2024 Volume 3
Article Contents
REVIEW   Open Access    

Gut microbiota-associated metabolites in metabolic diseases and their impact from food processing

More Information
  • Gut microbiota-associated metabolites can be synthesized endogenously or derived from dietary nutrients and host compounds. Among them, alkaloids, terpenes, and flavones originating from edible and medicinal foods have attracted remarkable interest recently and play crucial roles in metabolic diseases. The efficacy of these metabolites is susceptible to dietary intervention, especially after food processing. Therefore, this review comprehensively summarizes the different sources of common gut microbial metabolites, including microbial self-synthesis, biodegradation of exogenous substances (mainly dietary nutrients), and participation in host metabolism. In addition, the latest studies on novel metabolites such as alkaloids, terpenoids, and flavonoids are discussed, and their action mechanisms on metabolic diseases are elaborated. How food processing impacts dietary nutrients and their metabolites is carefully examined, as well as their effects on disease modification. These insights could contribute to a deeper understanding of the mechanisms by which diet efficacy helps prevent metabolic diseases, particularly through gut microbial metabolites.
  • 加载中
  • [1]

    Liu Z, Yan C, Lin X, Ai C, Dong X, et al. 2022. Responses of the gut microbiota and metabolite profiles to sulfated polysaccharides from sea cucumber in humanized microbiota mice. Food & Function 13(7):4171−83

    doi: 10.1039/D1FO04443E

    CrossRef   Google Scholar

    [2]

    Chen J, Xiao Y, Li D, Zhang S, Wu Y, et al. 2023. New insights into the mechanisms of high-fat diet mediated gut microbiota in chronic diseases. iMeta 2(1):e69

    doi: 10.1002/imt2.69

    CrossRef   Google Scholar

    [3]

    Liu L, Wang H, Chen X, Zhang Y, Zhang H, et al. 2023. Gut microbiota and its metabolites in depression: From pathogenesis to treatment. EBioMedicine 90:104527

    doi: 10.1016/j.ebiom.2023.104527

    CrossRef   Google Scholar

    [4]

    Bui TPN, De Vos WM. 2021. Next-generation therapeutic bacteria for treatment of obesity, diabetes, and other endocrine diseases. Best Practice & Research Clinical Endocrinology Metabolism 35(3):101504

    doi: 10.1016/j.beem.2021.101504

    CrossRef   Google Scholar

    [5]

    Krautkramer KA, Fan J, Bäckhed F. 2021. Gut microbial metabolites as multi-kingdom intermediates. Nature Reviews Microbiology 19(2):77−94

    doi: 10.1038/s41579-020-0438-4

    CrossRef   Google Scholar

    [6]

    Schirmer M, Garner A, Vlamakis H, Xavier R J. 2019. Microbial genes and pathways in inflammatory bowel disease. Nature Reviews Microbiology 17(8):497−511

    doi: 10.1038/s41579-019-0213-6

    CrossRef   Google Scholar

    [7]

    Shaffer M, Thurimella K, Quinn K, Doenges K, Zhang X, et al. 2019. AMON: annotation of metabolite origins via networks to integrate microbiome and metabolome data. BMC Bioinformatics 20(1):614

    doi: 10.1186/s12859-019-3176-8

    CrossRef   Google Scholar

    [8]

    Shepherd ES, DeLoache WC, Pruss KM, Whitaker WR, Sonnenburg JL. 2018. An exclusive metabolic niche enables strain engraftment in the gut microbiota. Nature 557(7705):434−38

    doi: 10.1038/s41586-018-0092-4

    CrossRef   Google Scholar

    [9]

    Agus A, Clément K, Sokol H. 2021. Gut microbiota-derived metabolites as central regulators in metabolic disorders. Gut 70(6):1174−82

    doi: 10.1136/gutjnl-2020-323071

    CrossRef   Google Scholar

    [10]

    Rodríguez-Daza MC, Pulido-Mateos EC, Lupien-Meilleur J, Guyonnet D, Desjardins Y, et al. 2021. Polyphenol-mediated gut microbiota modulation: toward prebiotics and further. Frontiers in Nutrition 8:689456

    doi: 10.3389/fnut.2021.689456

    CrossRef   Google Scholar

    [11]

    Wang J, Feng W, Tang F, Ao H, Peng C. 2019. Gut microbial transformation, a potential improving factor in the therapeutic activities of four groups of natural compounds isolated from herbal medicines. Fitoterapia 138:104293

    doi: 10.1016/j.fitote.2019.104293

    CrossRef   Google Scholar

    [12]

    Ansari MHR, Saher S, Parveen R, Khan W, Khan IA, et al. 2022. Role of gut microbiota metabolism and biotransformation on dietary natural products to human health implications with special reference to biochemoinformatics approach. Journal of Traditional and Complementary Medicine 13(2):150−60

    doi: 10.1016/j.jtcme.2022.03.005

    CrossRef   Google Scholar

    [13]

    Dai H, Han J, Wang T, Yin WB, Chen Y, et al. 2023. Recent advances in gut microbiota-associated natural products: structures, bioactivities, and mechanisms. Natural Product Reports 40(6):1078−93

    doi: 10.1039/D2NP00075J

    CrossRef   Google Scholar

    [14]

    Sá AGA, Moreno YMF, Carciofi BAM. 2020. Food processing for the improvement of plant proteins digestibility. Critical Reviews in Food Science and Nutrition 60(20):3367−86

    doi: 10.1080/10408398.2019.1688249

    CrossRef   Google Scholar

    [15]

    Lavelle A, Sokol H. 2020. Gut microbiota-derived metabolites as key actors in inflammatory bowel disease. Nature Reviews Gastroenterology & Hepatology 17(4):223−37

    Google Scholar

    [16]

    Tarracchini C, Lugli GA, Mancabelli L, Van Sinderen D, Turroni F, et al. 2024. Exploring the vitamin biosynthesis landscape of the human gut microbiota. mSystems 9(10):e0092924

    doi: 10.1128/msystems.00929-24

    CrossRef   Google Scholar

    [17]

    Zabolotneva AA, Kolesnikova IM, Vasiliev IY, Grigoryeva TV, Roumiantsev SA, et al. 2024. The obesogenic gut microbiota as a crucial factor defining the depletion of predicted enzyme abundance for vitamin B12 synthesis in the mouse intestine. Biomedicines 12(6):1280

    doi: 10.3390/biomedicines12061280

    CrossRef   Google Scholar

    [18]

    Qi X, Zhang Y, Zhang Y, Luo F, Song K, et al. 2023. Vitamin B12 produced by Cetobacterium somerae improves host resistance against pathogen infection through strengthening the interactions within gut microbiota. Microbiome 11(1):135

    doi: 10.1186/s40168-023-01574-2

    CrossRef   Google Scholar

    [19]

    Ribeiro M, Maciel C, Cruz P, Darmancier H, Nogueira T, et al. 2023. Exploiting potential probiotic lactic acid bacteria isolated from Chlorella vulgaris photobioreactors as promising vitamin B12 producers. Foods 12(17):3277

    doi: 10.3390/foods12173277

    CrossRef   Google Scholar

    [20]

    Uebanso T, Yoshimoto A, Aizawa S, Nakamura M, Masuda R, et al. 2020. Glycolate is a novel marker of vitamin B2 deficiency involved in gut microbe metabolism in mice. Nutrients 12(3):736

    doi: 10.3390/nu12030736

    CrossRef   Google Scholar

    [21]

    Rodionov DA, Arzamasov AA, Khoroshkin MS, Iablokov SN, Leyn SA, et al. 2019. Micronutrient requirements and sharing capabilities of the human gut microbiome. Frontiers in Microbiology 10:1316

    doi: 10.3389/fmicb.2019.01316

    CrossRef   Google Scholar

    [22]

    Atasoglu C, Valdés C, Walker ND, Newbold CJ, Wallace RJ. 1998. De novo synthesis of amino acids by the ruminal bacteria Prevotella bryantii B14, Selenomonas ruminantium HD4, and Streptococcus bovis ES1. Applied and Environmental Microbiology 64(8):2836−43

    doi: 10.1128/AEM.64.8.2836-2843.1998

    CrossRef   Google Scholar

    [23]

    Brown EM, Ke X, Hitchcock D, Jeanfavre S, Avila-Pacheco J, et al. 2019. Bacteroides-derived sphingolipids are critical for maintaining intestinal homeostasis and symbiosis. Cell Host & Microbe 25(5):668−80

    doi: 10.1016/j.chom.2019.04.002

    CrossRef   Google Scholar

    [24]

    Liang D, Zhang L, Chen H, Zhang H, Hu H, et al. 2021. Potato resistant starch inhibits diet-induced obesity by modifying the composition of intestinal microbiota and their metabolites in obese mice. International Journal of Biological Macromolecules 180:458−69

    doi: 10.1016/j.ijbiomac.2021.02.209

    CrossRef   Google Scholar

    [25]

    Mao G, Li S, Orfila C, Shen X, Zhou S, et al. 2019. Depolymerized RG-I-enriched pectin from citrus segment membranes modulates gut microbiota, increases SCFA production, and promotes the growth of Bifidobacterium spp., Lactobacillus spp. and Faecalibaculum spp. Food & Function 10(12):7828−43

    doi: 10.1039/c9fo01534e

    CrossRef   Google Scholar

    [26]

    Guo J, Zhang M, Wang H, Li N, Lu Z, et al. 2022. Gut microbiota and short chain fatty acids partially mediate the beneficial effects of inulin on metabolic disorders in obese ob/ob mice. Journal of Food Biochemistry 46(5):e14063

    doi: 10.1111/jfbc.14063

    CrossRef   Google Scholar

    [27]

    Meenu M, Xu B. 2019. A critical review on anti-diabetic and anti-obesity effects of dietary resistant starch. Critical Reviews in Food Science and Nutrition 59(18):3019−31

    doi: 10.1080/10408398.2018.1481360

    CrossRef   Google Scholar

    [28]

    Brandsma E, Kloosterhuis NJ, Koster M, Dekker DC, Gijbels MJJ, et al. 2019. A proinflammatory gut microbiota increases systemic inflammation and accelerates atherosclerosis. Circulation Research 124(1):94−100

    doi: 10.1161/CIRCRESAHA.118.313234

    CrossRef   Google Scholar

    [29]

    Wu J, Ma N, Johnston LJ, Ma X. 2020. Dietary nutrients mediate intestinal host defense peptide expression. Advances in Ntrition 11(1):92−102

    doi: 10.1093/advances/nmz057

    CrossRef   Google Scholar

    [30]

    Ma N, He T, Johnston LJ, Ma X. 2020. Host-microbiome interactions: the aryl hydrocarbon receptor as a critical node in tryptophan metabolites to brain signaling. Gut Microbes 11(5):1203−19

    doi: 10.1080/19490976.2020.1758008

    CrossRef   Google Scholar

    [31]

    Araújo JR, Tazi A, Burlen-Defranoux O, Vichier-Guerre S, Nigro G, et al. 2020. Fermentation products of commensal bacteria alter enterocyte lipid metabolism. Cell Host & Microbe 27(3):358−375.e7

    doi: 10.1016/j.chom.2020.01.028

    CrossRef   Google Scholar

    [32]

    Wu J, Zhao Y, Wang X, Kong L, Johnston LJ, et al. 2022. Dietary nutrients shape gut microbes and intestinal mucosa via epigenetic modifications. Critical Reviews in Food Science and Nutrition 62(3):783−97

    doi: 10.1080/10408398.2020.1828813

    CrossRef   Google Scholar

    [33]

    Dong F, Hao F, Murray IA, Smith PB, Koo I, et al. 2020. Intestinal microbiota-derived tryptophan metabolites are predictive of Ah receptor activity. Gut Microbes 12(1):e1788899

    doi: 10.1080/19490976.2020.1788899

    CrossRef   Google Scholar

    [34]

    Du L, Qi R, Wang J, Liu Z, Wu Z. 2021. Indole-3-propionic acid, a functional metabolite of Clostridium sporogenes, promotes muscle tissue development and reduces muscle cell inflammation. International Journal of Molecular Sciences 22(22):12435

    doi: 10.3390/ijms222212435

    CrossRef   Google Scholar

    [35]

    Cui W, Guo M, Liu D, Xiao P, Yang C, et al. 2024. Gut microbial metabolite facilitates colorectal cancer development via ferroptosis inhibition. Nature Cell Biology 26(1):124−37

    doi: 10.1038/s41556-023-01314-6

    CrossRef   Google Scholar

    [36]

    Fang Z, Pan T, Li L, Wang H, Zhu J, et al. 2022. Bifidobacterium longum mediated tryptophan metabolism to improve atopic dermatitis via the gut-skin axis. Gut Microbes 14(1):2044723

    doi: 10.1080/19490976.2022.2044723

    CrossRef   Google Scholar

    [37]

    Kitada Y, Muramatsu K, Toju H, Kibe R, Benno Y, et al. 2018. Bioactive polyamine production by a novel hybrid system comprising multiple indigenous gut bacterial strategies. Science Advances 4(6):eaat0062

    doi: 10.1126/sciadv.aat0062

    CrossRef   Google Scholar

    [38]

    Anwar S, Bhandari U, Panda BP, Dubey K, Khan W, et al. 2018. Trigonelline inhibits intestinal microbial metabolism of choline and its associated cardiovascular risk. Journal of Pharmaceutical and Biomedical Analysis 159:100−12

    doi: 10.1016/j.jpba.2018.06.027

    CrossRef   Google Scholar

    [39]

    Dehghan P, Farhangi MA, Nikniaz L, Nikniaz Z, Asghari-Jafarabadi M. 2020. Gut microbiota-derived metabolite trimethylamine N-oxide (TMAO) potentially increases the risk of obesity in adults: An exploratory systematic review and dose-response meta-analysis. Obesity Reviews 21(5):e12993

    doi: 10.1111/obr.12993

    CrossRef   Google Scholar

    [40]

    Li R, Zheng M, Zheng M, Cai R, Cui X, et al. 2022. Metagenomic analysis reveals the linkages between bacteria and the functional enzymes responsible for potential ammonia and biogenic amine production in alfalfa silage. Journal of Applied Microbiology 132(4):2594−604

    doi: 10.1111/jam.15411

    CrossRef   Google Scholar

    [41]

    Sugiyama Y, Nara M, Sakanaka M, Gotoh A, Kitakata A, et al. 2017. Comprehensive analysis of polyamine transport and biosynthesis in the dominant human gut bacteria: Potential presence of novel polyamine metabolism and transport genes. The International Journal of Biochemistry & Cell Biology 93:52−61

    doi: 10.1016/j.biocel.2017.10.01

    CrossRef   Google Scholar

    [42]

    Sakanaka M, Sugiyama Y, Kitakata A, Katayama T, Kurihara S. 2016. Carboxyspermidine decarboxylase of the prominent intestinal microbiota species Bacteroides thetaiotaomicron is required for spermidine biosynthesis and contributes to normal growth. Amino Acids 48(10):2443−51

    doi: 10.1007/s00726-016-2233-0

    CrossRef   Google Scholar

    [43]

    Li D, Feng Y, Tian M, Ji J, Hu X, et al. 2021. Gut microbiota-derived inosine from dietary barley leaf supplementation attenuates colitis through PPARγ signaling activation. Microbiome 9(1):83

    doi: 10.1186/s40168-021-01028-7

    CrossRef   Google Scholar

    [44]

    Huang Y, Wang YF, Ruan XZ, Lau CW, Wang L, et al. 2024. The role of KLF2 in regulating hepatic lipogenesis and blood cholesterol homeostasis via the SCAP/SREBP pathway. Journal of Lipid Research 65(1):100472

    doi: 10.1016/j.jlr.2023.100472

    CrossRef   Google Scholar

    [45]

    Li Y, Dong P, Dai L, Wang S. 2023. Untargeted and targeted metabolomics reveal the active peptide of eupolyphaga sinensis walker against hyperlipidemia by modulating imbalance in amino acid metabolism. Molecules 28(20):7049

    doi: 10.3390/molecules28207049

    CrossRef   Google Scholar

    [46]

    Kenny DJ, Plichta DR, Shungin D, Koppel N, Hall AB, et al. 2020. Cholesterol metabolism by uncultured human gut bacteria influences host cholesterol level. Cell Host & Microbe 28(2):245−257.e6

    doi: 10.1016/j.chom.2020.05.013

    CrossRef   Google Scholar

    [47]

    Yao L, D'Agostino GD, Park J, Hang S, Adhikari AA, et al. 2022. A biosynthetic pathway for the selective sulfonation of steroidal metabolites by human gut bacteria. Nature Microbiology 7(9):1404−18

    doi: 10.1038/s41564-022-01176-y

    CrossRef   Google Scholar

    [48]

    Deng C, Pan J, Zhu H, Chen ZY. 2023. Effect of gut microbiota on blood cholesterol: A review on mechanisms. Foods 12(23):4308

    doi: 10.3390/foods12234308

    CrossRef   Google Scholar

    [49]

    Guzior DV, Quinn RA. 2021. Review: microbial transformations of human bile acids. Microbiome 9(1):140

    doi: 10.1186/s40168-021-01101-1

    CrossRef   Google Scholar

    [50]

    Pruss KM, Chen H, Liu Y, Van Treuren W, Higginbottom SK, et al. 2023. Host-microbe co-metabolism via MCAD generates circulating metabolites including hippuric acid. Nature Communications 14(1):512

    doi: 10.1038/s41467-023-36138-3

    CrossRef   Google Scholar

    [51]

    Brial F, Chilloux J, Nielsen T, Vieira-Silva S, Falony G, et al. 2021. Human and preclinical studies of the host-gut microbiome co-metabolite hippurate as a marker and mediator of metabolic health. Gut 70(11):2105−14

    doi: 10.1136/gutjnl-2020-323314

    CrossRef   Google Scholar

    [52]

    Zhang L, Ma XG. 2024. A comprehensive review on biotransformation, interaction, and health of gut microbiota and bioactive components. Combinatorial Chemistry & High Throughput Screening 27(11):1551−65

    doi: 10.2174/0113862073257733231011072004

    CrossRef   Google Scholar

    [53]

    Cheng H, Liu J, Tan Y, Feng W, Peng C. 2022. Interactions between gut microbiota and berberine, a necessary procedure to understand the mechanisms of berberine. Journal of Pharmaceutical Analysis 12(4):541−55

    doi: 10.1016/j.jpha.2021.10.003

    CrossRef   Google Scholar

    [54]

    Li C, Ai G, Wang Y, Lu Q, Luo C, et al. 2020. Oxyberberine, a novel gut microbiota-mediated metabolite of berberine, possesses superior anti-colitis effect: Impact on intestinal epithelial barrier, gut microbiota profile and TLR4-MyD88-NF-κB pathway. Pharmacological Research 152:104603

    doi: 10.1016/j.phrs.2019.104603

    CrossRef   Google Scholar

    [55]

    Kamble SH, Sharma A, King TI, Berthold EC, León F, et al. 2020. Exploration of cytochrome P450 inhibition mediated drug-drug interaction potential of kratom alkaloids. Toxicology Letters 319:148−54

    doi: 10.1016/j.toxlet.2019.11.005

    CrossRef   Google Scholar

    [56]

    Zhang ZW, Cong L, Peng R, Han P, Ma SR, et al. 2021. Transformation of berberine to its demethylated metabolites by the CYP51 enzyme in the gut microbiota. Journal of Pharmaceutical Analysis 11(5):628−37

    doi: 10.1016/j.jpha.2020.10.001

    CrossRef   Google Scholar

    [57]

    Su Y, Huang P, Wu Z, Dai W, Zhang Y, et al. 2024. Effect of dietary supplementation with sanguinarine on meat quality and lipid metabolism of broilers. Poultry Science 103(8):103925

    doi: 10.1016/j.psj.2024.103925

    CrossRef   Google Scholar

    [58]

    Yue SJ, Liu J, Wang WX, Wang AT, Yang XY, et al. 2019. Berberine treatment-emergent mild diarrhea associated with gut microbiota dysbiosis. Biomedicine & Pharmacotherapy 116:109002

    doi: 10.1016/j.biopha.2019.109002

    CrossRef   Google Scholar

    [59]

    Wu H, Chen Q, Liu J, Chen X, Luo H, et al. 2021. Microbiome analysis reveals gut microbiota alteration in mice with the effect of matrine. Microbial Pathogenesis 156:104926

    doi: 10.1016/j.micpath.2021.104926

    CrossRef   Google Scholar

    [60]

    Wang Y, Zhang Z, Du M, Ji X, Liu X, et al. 2024. Berberine alleviates ETEC-induced intestinal inflammation and oxidative stress damage by optimizing intestinal microbial composition in a weaned piglet model. Frontiers in Immunology 15:1460127

    doi: 10.3389/fimmu.2024.1460127

    CrossRef   Google Scholar

    [61]

    Liu Q, Liu S, Cao H, Ji W, Li C, et al. 2021. Ramulus mori (Sangzhi) alkaloids (SZ-A) ameliorate glucose metabolism accompanied by the modulation of gut microbiota and ileal inflammatory damage in type 2 diabetic KKAy mice. Frontiers in Pharmacology 12:642400

    doi: 10.3389/fphar.2021.642400

    CrossRef   Google Scholar

    [62]

    Shu X, Li M, Cao Y, Li C, Zhou W, et al. 2021. Berberine alleviates non-alcoholic steatohepatitis through modulating gut microbiota mediated intestinal FXR activation. Frontiers in Pharmacology 12:750826

    doi: 10.3389/fphar.2021.750826

    CrossRef   Google Scholar

    [63]

    Fusco W, Lorenzo MB, Cintoni M, Porcari S, Rinninella E, et al. 2023. Short-chain fatty-acid-producing bacteria: Key components of the human gut microbiota. Nutrients 15(9):2211

    doi: 10.3390/nu15092211

    CrossRef   Google Scholar

    [64]

    Wolf PG, Devendran S, Doden HL, Ly LK, Moore T, et al. 2021. Berberine alters gut microbial function through modulation of bile acids. BMC Microbiology 21(1):24

    doi: 10.1186/s12866-020-02020-1

    CrossRef   Google Scholar

    [65]

    Mitchelson KAJ, O'Connell F, O'Sullivan J, Roche HM. 2024. Obesity, dietary fats, and gastrointestinal cancer risk-potential mechanisms relating to lipid metabolism and inflammation. Metabolites 14(1):42

    doi: 10.3390/metabo14010042

    CrossRef   Google Scholar

    [66]

    Xiang D, Yang J, Liu L, Yu H, Gong X, et al. 2023. The regulation of tissue-specific farnesoid X receptor on genes and diseases involved in bile acid homeostasis. Biomedicine & Pharmacotherapy 168:115606

    doi: 10.1016/j.biopha.2023.115606

    CrossRef   Google Scholar

    [67]

    Liu Z, Liu J, Tang R, Zhang Z, Tian S. 2024. Procyanidin B1 and coumaric acid from highland barley alleviated high-fat-diet-induced hyperlipidemia by regulating PPARα-mediated hepatic lipid metabolism and gut microbiota in diabetic C57BL/6J mice. Foods 13(12):1843

    doi: 10.3390/foods13121843

    CrossRef   Google Scholar

    [68]

    Ning Y, Xu F, Xin R, Yao F. 2020. Palmatine regulates bile acid cycle metabolism and maintains intestinal flora balance to maintain stable intestinal barrier. Life Sciences 262:118405

    doi: 10.1016/j.lfs.2020.118405

    CrossRef   Google Scholar

    [69]

    Li Z, Jiang JD, Kong WJ. 2014. Berberine up-regulates hepatic low-density lipoprotein receptor through Ras-independent but AMP-activated protein kinase-dependent Raf-1 activation. Biological & Pharmaceutical Bulletin 37(11):1766−75

    doi: 10.1248/bpb.b14-00412

    CrossRef   Google Scholar

    [70]

    Zhang C, Deng J, Liu D, Tuo X, Xiao L, et al. 2018. Nuciferine ameliorates hepatic steatosis in high-fat diet/streptozocin-induced diabetic mice through a PPARα/PPARγ coactivator-1α pathway. British Journal of Pharmacology 175(22):4218−28

    doi: 10.1111/bph.14482

    CrossRef   Google Scholar

    [71]

    Dou Y, Huang R, Li Q, Liu Y, Li Y, et al. 2021. Oxyberberine, an absorbed metabolite of berberine, possess superior hypoglycemic effect via regulating the PI3K/Akt and Nrf2 signaling pathways. Biomedicine & Pharmacotherapy 137:111312

    doi: 10.1016/j.biopha.2021.111312

    CrossRef   Google Scholar

    [72]

    Wu YS, Li ZM, Chen YT, Dai SJ, Zhou XJ, et al. 2020. Berberine improves inflammatory responses of diabetes mellitus in Zucker diabetic fatty rats and insulin-resistant HepG2 cells through the PPM1B pathway. Journal of Immunology Research 2020:2141508

    doi: 10.1155/2020/2141508

    CrossRef   Google Scholar

    [73]

    Guo B, Yang B, Pang X, Chen T, Chen F, et al. 2019. Fucoxanthin modulates cecal and fecal microbiota differently based on diet. Food & Function 10(9):5644−55

    doi: 10.1039/c9fo01018a

    CrossRef   Google Scholar

    [74]

    Sell LB, Ramelow CC, Kohl HM, Hoffman K, Bains JK, et al. 2022. Farnesol induces protection against murine CNS inflammatory demyelination and modifies gut microbiome. Clinical Immunology 235:108766

    doi: 10.1016/j.clim.2021.108766

    CrossRef   Google Scholar

    [75]

    Han C, Wu X, Zou N, Zhang Y, Yuan J, et al. 2021. Cichorium pumilum Jacq extract inhibits LPS-induced inflammation via MAPK signaling pathway and protects rats from hepatic fibrosis caused by abnormalities in the gut-liver axis. Frontiers in Pharmacology 12:683613

    doi: 10.3389/fphar.2021.683613

    CrossRef   Google Scholar

    [76]

    Ricci C, Rizzello F, Valerii MC, Spisni E, Gionchetti P, et al. 2022. Geraniol treatment for irritable bowel syndrome: A double-blind randomized clinical trial. Nutrients 14(19):4208

    doi: 10.3390/nu14194208

    CrossRef   Google Scholar

    [77]

    Quan LH, Zhang C, Dong M, Jiang J, Xu H, et al. 2020. Myristoleic acid produced by enterococci reduces obesity through brown adipose tissue activation. Gut 69(7):1239−47

    doi: 10.1136/gutjnl-2019-319114

    CrossRef   Google Scholar

    [78]

    Zhang Y, Peng Y, Zhao L, Zhou G, Li X. 2021. Regulating the gut microbiota and SCFAs in the faeces of T2DM rats should be one of antidiabetic mechanisms of mogrosides in the fruits of Siraitia grosvenorii. Journal of Ethnopharmacology 274:114033

    doi: 10.1016/j.jep.2021.114033

    CrossRef   Google Scholar

    [79]

    Xu Z, Huang J, Wen M, Zhang X, Lyu D, et al. 2024. Gentiopicroside ameliorates glucose and lipid metabolism in T2DM via targeting FGFR1. Phytomedicine 132:155780

    doi: 10.1016/j.phymed.2024.155780

    CrossRef   Google Scholar

    [80]

    Fu J, Yu H, Guo Q, Wang Y, Xu H, et al. 2023. Metabolic transformation of gentiopicrin, a liver protective active ingredient, based on intestinal bacteria. Molecules 28(22):7575

    doi: 10.3390/molecules28227575

    CrossRef   Google Scholar

    [81]

    Li YX, Lv L, Li SL, Qian HH. 2024. Gentianine alleviates dextran sulfate sodium-induced ulcerative colitis via inhibition of TLR4/NLRP3-mediated pyroptosis. International immunopharmacology 126:111214

    doi: 10.1016/j.intimp.2023.111214

    CrossRef   Google Scholar

    [82]

    Zhang F, He F, Li L, Guo L, Zhang B, et al. 2020. Bioavailability based on the gut microbiota: A new perspective. Microbiology and Molecular Biology Reviews 84(2):e00072-19

    doi: 10.1128/MMBR.00072-19

    CrossRef   Google Scholar

    [83]

    Yang H, Li Y, Huo P, Li XO, Kong D, et al. 2015. Protective effect of Jolkinolide B on LPS-induced mouse acute lung injury. International Immunopharmacology 26(1):119−24

    doi: 10.1016/j.intimp.2015.03.021

    CrossRef   Google Scholar

    [84]

    Liu S, Zhang S, Lv X, Lu J, Ren C, et al. 2019. Limonin ameliorates ulcerative colitis by regulating STAT3/miR-214 signaling pathway. International Immunopharmacology 75:105768

    doi: 10.1016/j.intimp.2019.105768

    CrossRef   Google Scholar

    [85]

    Ben Ammar R, Zahra HA, Abu Zahra AM, Alfwuaires M, Abdulaziz Alamer S, et al. 2023. Protective effect of fucoxanthin on zearalenone-induced hepatic damage through Nrf2 mediated by PI3K/AKT signaling. Marine Drugs 21(7):391

    doi: 10.3390/md21070391

    CrossRef   Google Scholar

    [86]

    Suwanmanee G, Tantrawatpan C, Kheolamai P, Paraoan L, Manochantr S. 2023. Fucoxanthin diminishes oxidative stress damage in human placenta-derived mesenchymal stem cells through the PI3K/Akt/Nrf-2 pathway. Scientific Reports 13(1):22974

    doi: 10.1038/s41598-023-49751-5

    CrossRef   Google Scholar

    [87]

    Yang S, Li J, Yan L, Wu Y, Zhang L, et al. 2024. Molecular mechanisms of fucoxanthin in alleviating lipid deposition in metabolic associated fatty liver disease. Journal of Agricultural and Food Chemistry 72(18):10391−405

    doi: 10.1021/acs.jafc.4c00590

    CrossRef   Google Scholar

    [88]

    Chang YH, Chen YL, Huang WC, Liou CJ. 2018. Fucoxanthin attenuates fatty acid-induced lipid accumulation in FL83B hepatocytes through regulated Sirt1/AMPK signaling pathway. Biochemical and Biophysical Research Communications 495(1):197−203

    doi: 10.1016/j.bbrc.2017.11.022

    CrossRef   Google Scholar

    [89]

    Ou HC, Chou WC, Chu PM, Hsieh PL, Hung CH, et al. 2019. Fucoxanthin protects against oxLDL-induced endothelial damage via activating the AMPK-Akt-CREB-PGC1α pathway. Molecular Nutrition & Food Research 63(10):e1801353

    doi: 10.1002/mnfr.201801353

    CrossRef   Google Scholar

    [90]

    Ding LL, Matsumura M, Obitsu T, Sugino T. 2021. Phytol supplementation alters plasma concentrations of formate, amino acids, and lipid metabolites in sheep. Animal 15(3):100174

    doi: 10.1016/j.animal.2021.100174

    CrossRef   Google Scholar

    [91]

    Nakanishi T, Kagamizono K, Yokoyama S, Suzuki R, Sakakibara H, et al. 2020. Effects of dietary phytol on tissue accumulation of phytanic acid and pristanic acid and on the tissue lipid profiles in mice. Animal Science Journal 91(1):e13424

    doi: 10.1111/asj.13424

    CrossRef   Google Scholar

    [92]

    Torequl Islam M, Shimul Bhuia M, Paulo Martins de Lima J, Paulo Araujo Maia F, Beatriz Herminia Ducati A, et al. 2023. Phytanic acid, an inconclusive phytol metabolite: A review. Current Research in Toxicology 5:100120

    doi: 10.1016/j.crtox.2023.100120

    CrossRef   Google Scholar

    [93]

    Kasahara N, Imi Y, Amano R, Shinohara M, Okada K, et al. 2023. A gut microbial metabolite of linoleic acid ameliorates liver fibrosis by inhibiting TGF-β signaling in hepatic stellate cells. Scientific Reports 13(1):18983

    doi: 10.1038/s41598-023-46404-5

    CrossRef   Google Scholar

    [94]

    Zhang Z, Diao P, Zhang X, Nakajima T, Kimura T, et al. 2022. Clinically relevant dose of pemafibrate, a novel selective peroxisome proliferator-activated receptor α modulator (SPPARMα), lowers serum triglyceride levels by targeting hepatic PPARα in mice. Biomedicines 10(7):1667

    doi: 10.3390/biomedicines10071667

    CrossRef   Google Scholar

    [95]

    Zhu SS, Liu JW, Yan YM, Liu Y, Mao Z, et al. 2020. Terpenoids from Resina commiphora regulating lipid metabolism via activating PPARα and CPT1 expression. Organic Letters 22(9):3428−32

    doi: 10.1021/acs.orglett.0c00898

    CrossRef   Google Scholar

    [96]

    Kuroyanagi K, Kang MS, Goto T, Hirai S, Ohyama K, et al. 2008. Citrus auraptene acts as an agonist for PPARs and enhances adiponectin production and MCP-1 reduction in 3T3-L1 adipocytes. Biochemical and Biophysical Research Communications 366(1):219−25

    doi: 10.1016/j.bbrc.2007.11.119

    CrossRef   Google Scholar

    [97]

    Alseekh S, Perez de Souza L, Benina M, Fernie AR. 2020. The style and substance of plant flavonoid decoration; towards defining both structure and function. Phytochemistry 174:112347

    doi: 10.1016/j.phytochem.2020.112347

    CrossRef   Google Scholar

    [98]

    Dias MC, Pinto DCGA, Silva AMS. 2021. Plant flavonoids: Chemical characteristics and biological activity. Molecules 26(17):5377

    doi: 10.3390/molecules26175377

    CrossRef   Google Scholar

    [99]

    Wang M, Yu F, Zhang Y, Chang W, Zhou M. 2022. The effects and mechanisms of flavonoids on cancer prevention and therapy: Focus on gut microbiota. International Journal of Biological Sciences 18(4):1451−75

    doi: 10.7150/ijbs.68170

    CrossRef   Google Scholar

    [100]

    Santangelo R, Silvestrini A, Mancuso C. 2019. Ginsenosides, catechins, quercetin and gut microbiota: Current evidence of challenging interactions. Food and Chemical Toxicology 123:42−49

    doi: 10.1016/j.fct.2018.10.042

    CrossRef   Google Scholar

    [101]

    Di Pede G, Bresciani L, Calani L, Petrangolini G, Riva A, et al. 2020. The human microbial metabolism of quercetin in different formulations: An in vitro evaluation. Foods 9(8):1121

    doi: 10.3390/foods9081121

    CrossRef   Google Scholar

    [102]

    Tan J, Li Y, Hou DX, Wu S. 2019. The effects and mechanisms of cyanidin-3-glucoside and its phenolic metabolites in maintaining intestinal integrity. Antioxidants 8(10):479

    doi: 10.3390/antiox8100479

    CrossRef   Google Scholar

    [103]

    Li J, Liu W, Jiao W, Lian Y, Mi S, Chitrakar B, et al. 2024. Effect of ferulic acid and p-coumaric acid on lowering uric acid through network pharmacology and in vitro studies. Food Safety and Health 2(1):133−44

    doi: 10.1002/fsh3.12027

    CrossRef   Google Scholar

    [104]

    Tan S, Rupasinghe TW, Tull DL, Boughton B, Oliver C, et al. 2014. Degradation of curcuminoids by in vitro pure culture fermentation. Journal of Agricultural and Food Chemistry 62(45):11005−15

    doi: 10.1021/jf5031168

    CrossRef   Google Scholar

    [105]

    Ahn HJ, You HJ, Park MS, Li Z, Choe D, et al. 2020. Microbial biocatalysis of quercetin-3-glucoside and isorhamnetin-3-glucoside in Salicornia herbacea and their contribution to improved anti-inflammatory activity. RSC Advances 10(9):5339−50

    doi: 10.1039/C9RA08059G

    CrossRef   Google Scholar

    [106]

    Su T, Huang C, Yang C, Jiang T, Su J, et al. 2020. Apigenin inhibits STAT3/CD36 signaling axis and reduces visceral obesity. Pharmacological Research 152:104586

    doi: 10.1016/j.phrs.2019.104586

    CrossRef   Google Scholar

    [107]

    Li H, Zhang M, Wang Y, Gong K, Yan T, et al. 2022. Daidzein alleviates doxorubicin-induced heart failure via the SIRT3/FOXO3a signaling pathway. Food & Function 13(18):9576−88

    doi: 10.1039/d2fo00772j

    CrossRef   Google Scholar

    [108]

    Luo T, Miranda-Garcia O, Sasaki G, Wang J, Shay NF. 2018. Genistein and daidzein decrease food intake and body weight gain in mice, and alter LXR signaling in vivo and in vitro. Food & Function 9(12):6257−67

    doi: 10.1039/c8fo01718b

    CrossRef   Google Scholar

    [109]

    Ronis MJ, Chen Y, Badeaux J, Badger TM. 2009. Dietary soy protein isolate attenuates metabolic syndrome in rats via effects on PPAR, LXR, and SREBP signaling. The Journal of Nutrition 139(8):1431−38

    doi: 10.3945/jn.109.107029

    CrossRef   Google Scholar

    [110]

    Zhang H, Chi M, Chen L, Sun X, Wan L, et al. 2021. Daidzein alleviates cisplatin-induced muscle atrophy by regulating Glut4/AMPK/FoxO pathway. Phytotherapy Research 35(8):4363−76

    doi: 10.1002/ptr.7132

    CrossRef   Google Scholar

    [111]

    Han S, You L, Hu Y, Wei S, Liu T, et al. 2023. Ginsenoside F2 enhances glucose metabolism by modulating insulin signal transduction in human hepatocarcinoma cells. Journal of Ginseng Research 47(3):420−28

    doi: 10.1016/j.jgr.2022.10.003

    CrossRef   Google Scholar

    [112]

    Mokashi P, Khanna A, Pandita N. 2017. Flavonoids from Enicostema littorale blume enhances glucose uptake of cells in insulin resistant human liver cancer (HepG2) cell line via IRS-1/PI3K/Akt pathway. Biomedicine & Pharmacotherapy 90:268−77

    doi: 10.1016/j.biopha.2017.03.047

    CrossRef   Google Scholar

    [113]

    Zhou Y, Wang S, Wan T, Huang Y, Pang N, et al. 2020. Cyanidin-3-O-β-glucoside inactivates NLRP3 inflammasome and alleviates alcoholic steatohepatitis via SirT1/NF-κB signaling pathway. Free Radical Biology & Medicine 160:334−41

    doi: 10.1016/j.freeradbiomed.2020.08.006

    CrossRef   Google Scholar

    [114]

    Yue B, Ren J, Yu Z, Luo X, Ren Y, et al. 2020. Pinocembrin alleviates ulcerative colitis in mice via regulating gut microbiota, suppressing TLR4/MD2/NF-κB pathway and promoting intestinal barrier. Bioscience Reports 40(7):BSR20200986

    doi: 10.1042/BSR20200986

    CrossRef   Google Scholar

    [115]

    Ju S, Ge Y, Li P, Tian X, Wang H, et al. 2018. Dietary quercetin ameliorates experimental colitis in mouse by remodeling the function of colonic macrophages via a heme oxygenase-1-dependent pathway. Cell Cycle 17(1):53−63

    doi: 10.1080/15384101.2017.1387701

    CrossRef   Google Scholar

    [116]

    Hussain T, Yuan D, Tan B, Murtaza G, Rahu N, et al. 2020. Eucommia ulmoides flavones (EUF) abrogated enterocyte damage induced by LPS involved in NF-κB signaling pathway. Toxicology in Vitro 62:104674

    doi: 10.1016/j.tiv.2019.104674

    CrossRef   Google Scholar

    [117]

    Wu S, Hu R, Tan J, He Z, Liu M, et al. 2019. Cyanidin 3-glucoside and its metabolites protect against nonalcoholic fatty liver disease: Crosstalk between serum lipids, inflammatory cytokines and MAPK/ERK pathway. Stroke 50:AWP534

    doi: 10.1161/str.50.suppl_1.wp534

    CrossRef   Google Scholar

    [118]

    Jeong HJ, Nam SY, Kim HY, Jin MH, Kim MH, et al. 2018. Anti-allergic inflammatory effect of vanillic acid through regulating thymic stromal lymphopoietin secretion from activated mast cells. Natural Product Research 32(24):2945−49

    doi: 10.1080/14786419.2017.1389938

    CrossRef   Google Scholar

    [119]

    Forde CG, Decker EA. 2022. The importance of food processing and eating behavior in promoting healthy and sustainable diets. Annual Review of Nutrition 42:377−99

    doi: 10.1146/annurev-nutr-062220-030123

    CrossRef   Google Scholar

    [120]

    Meléndez-Martínez AJ, Esquivel P, Rodriguez-Amaya DB. 2023. Comprehensive review on carotenoid composition: Transformations during processing and storage of foods. Food Research International 169:112773

    doi: 10.1016/j.foodres.2023.112773

    CrossRef   Google Scholar

    [121]

    Castro-Barquero S, Ruiz-León AM, Sierra-Pérez M, Estruch R, Casas R. 2020. Dietary strategies for metabolic syndrome: A comprehensive review. Nutrients 12(10):2983

    doi: 10.3390/nu12102983

    CrossRef   Google Scholar

    [122]

    Bhat ZF, Morton JD, Bekhit AEDA, Kumar S, Bhat HF. 2021. Thermal processing implications on the digestibility of meat, fish and seafood proteins. Comprehensive Reviews in Food Science and Food Safety 20(5):4511−48

    doi: 10.1111/1541-4337.12802

    CrossRef   Google Scholar

    [123]

    Chupeerach C, Aursalung A, Watcharachaisoponsiri T, Whanmek K, Thiyajai P, et al. 2021. The effect of steaming and fermentation on nutritive values, antioxidant activities, and inhibitory properties of tea leaves. Foods 10(1):117

    doi: 10.3390/foods10010117

    CrossRef   Google Scholar

    [124]

    Zheng B, Zhong S, Tang Y, Chen L. 2020. Understanding the nutritional functions of thermally-processed whole grain highland barley in vitro and in vivo. Food Chemistry 310:125979

    doi: 10.1016/j.foodchem.2019.125979

    CrossRef   Google Scholar

    [125]

    Jain T, Grover K, Kaur G. 2016. Effect of processing on nutrients and fatty acid composition of garden cress (Lepidium sativum) seeds. Food Chemistry 213:806−12

    doi: 10.1016/j.foodchem.2016.07.034

    CrossRef   Google Scholar

    [126]

    Xu A, Zhao Y, Shi Y, Zuo X, Yang Y, et al. 2022. Effects of oxidation-based tea processing on the characteristics of the derived polysaccharide conjugates and their regulation of intestinal homeostasis in DSS-induced colitis mice. International Journal of Biological Macromolecules 214:402−13

    doi: 10.1016/j.ijbiomac.2022.06.115

    CrossRef   Google Scholar

    [127]

    Li T, Rui Z, Mao L, Chang Y, Shao J, et al. 2023. Eight weeks of Bifidobacterium lactis BL-99 supplementation improves lipid metabolism and sports performance through short-chain fatty acids in cross-country skiers: A preliminary study. Nutrients 15(21):4554

    doi: 10.3390/nu15214554

    CrossRef   Google Scholar

    [128]

    Ji J, Zhang S, Yuan M, Zhang M, Tang L, et al. 2022. Fermented Rosa roxburghii tratt juice alleviates high-fat diet-induced hyperlipidemia in rats by modulating gut microbiota and metabolites. Frontiers in Pharmacology 13:883629

    doi: 10.3389/fphar.2022.883629

    CrossRef   Google Scholar

    [129]

    Zhang Y, Dong L, Zhang J, Shi J, Wang Y, et al. 2021. Adverse effects of thermal food processing on the structural, nutritional, and biological properties of proteins. Annual Review of Food Science and Technology 12:259−86

    doi: 10.1146/annurev-food-062320-012215

    CrossRef   Google Scholar

    [130]

    Yang S, Mi L, Wang K, Wang X, Wu J, et al. 2023. Comparative metabolomics analysis in the clean label ingredient of NFC spine grape juice processed by mild heating vs high pressure processing. Food Innovation and Advances 2(2):95−105

    doi: 10.48130/FIA-2023-0011

    CrossRef   Google Scholar

    [131]

    Wang X, Dong L, Ma C, Wang Z, Hu X, et al. 2023. Impact of high-hydrostatic pressure and thermal processing on the antioxidant profiles and capacity of tomato juice during storage. Food Innovation and Advances 2(2):124−34

    doi: 10.48130/FIA-2023-0016

    CrossRef   Google Scholar

    [132]

    Li M, Chen X, Deng J, Ouyang D, Wang D, et al. 2020. Effect of thermal processing on free and bound phenolic compounds and antioxidant activities of hawthorn. Food Chemistry 332:127429

    doi: 10.1016/j.foodchem.2020.127429

    CrossRef   Google Scholar

    [133]

    Chen Y, Wang J, Zou L, Cao H, Ni X, et al. 2023. Dietary proanthocyanidins on gastrointestinal health and the interactions with gut microbiota. Critical Reviews in Food Science and Nutrition 63(23):6285−308

    doi: 10.1080/10408398.2022.2030296

    CrossRef   Google Scholar

    [134]

    Ângela Aragão M M, Pires L, Santos-Buelga C, Barros L, Calhelha RC. 2024. Revitalising riboflavin: Unveiling its timeless significance in human physiology and health. Foods 13(14):2255

    doi: 10.3390/foods13142255

    CrossRef   Google Scholar

    [135]

    Nosratabadi S, Ashtary-Larky D, Hosseini F, Namkhah Z, Mohammadi S, et al. 2023. The effects of vitamin C supplementation on glycemic control in patients with type 2 diabetes: A systematic review and meta-analysis. Diabetes & Metabolic Syndrome 17(8):102824

    doi: 10.1016/j.dsx.2023.102824

    CrossRef   Google Scholar

    [136]

    Ranjitha Gracy TK, Sharanyakanth PS, Radhakrishnan M. 2020. Non-thermal technologies: Solution for hazardous pesticides reduction in fruits and vegetables. Critical Reviews in Food Science and Nutrition 62(7):1782−99

    doi: 10.1080/10408398.2020.1847029

    CrossRef   Google Scholar

    [137]

    Barbhuiya RI, Singha P, Singh SK. 2021. A comprehensive review on impact of non-thermal processing on the structural changes of food components. Food Research International 149:110647

    doi: 10.1016/j.foodres.2021.110647

    CrossRef   Google Scholar

    [138]

    Stübler AS, Lesmes U, Juadjur A, Heinz V, Rauh C, et al. 2020. Impact of pilot-scale processing (thermal, PEF, HPP) on the stability and bioaccessibility of polyphenols and proteins in mixed protein- and polyphenol-rich juice systems. Innovative Food Science & Emerging Technologies 64:102426

    doi: 10.1016/j.ifset.2020.102426

    CrossRef   Google Scholar

    [139]

    Yildiz S, Pokhrel PR, Unluturk S, Barbosa-Cánovas GV. 2021. Shelf life extension of strawberry juice by equivalent ultrasound, high pressure, and pulsed electric fields processes. Food Research International 140:110040

    doi: 10.1016/j.foodres.2020.110040

    CrossRef   Google Scholar

    [140]

    Noguera NH, Lima DC, Filho EGA, Fonteles TV, Rodrigues S. 2021. Influence of different non-thermal processing on guava, orange, and tangerine juices and the food matrix effects. Food and Bioprocess Technology 14:1662−72

    doi: 10.1007/s11947-021-02663-6

    CrossRef   Google Scholar

    [141]

    Mehta D, Sharma N, Bansal V, Sangwan RS, Yadav SK. 2022. Impact of ultrasonication, ultraviolet and atmospheric cold plasma processing on quality parameters of tomato-based beverage in comparison with thermal processing. Innovative Food Science & Emerging Technologies 52:343−49

    doi: 10.1016/j.ifset.2019.01.015

    CrossRef   Google Scholar

    [142]

    Armas Díaz Y, Ferreiro Cotorruelo MS, Battino MA. 2023. The role of dietary polyphenols in the control of chronic noncommunicable diseases. Food Safety and Health 1(1):13−21

    doi: 10.1002/fsh3.12013

    CrossRef   Google Scholar

    [143]

    Tan C, Wang M, Kong Y, Wan M, Deng H, et al. 2022. Anti-inflammatory and intestinal microbiota modulation properties of high hydrostatic pressure treated cyanidin-3-glucoside and blueberry pectin complexes on dextran sodium sulfate-induced ulcerative colitis mice. Food & Function 13(8):4384−98

    doi: 10.1039/d1fo03376j

    CrossRef   Google Scholar

    [144]

    Rodríguez Ó, Gomes WF, Rodrigues S, Fernandes FAN. 2017. Effect of indirect cold plasma treatment on cashew apple juice (Anacardium occidentale L.). LWT 84:457−63

    doi: 10.1016/j.lwt.2017.06.010

    CrossRef   Google Scholar

    [145]

    Zhang B, Tan C, Zou F, Sun Y, Shang N, et al. 2022. Impacts of cold plasma technology on sensory, nutritional and safety quality of food: A review. Foods 11(18):2818

    doi: 10.3390/foods11182818

    CrossRef   Google Scholar

    [146]

    Yan J, Xue Q, Chen W, Wang K, Peng D, et al. 2022. Probiotic-fermented rice buckwheat alleviates high-fat diet-induced hyperlipidemia in mice by suppressing lipid accumulation and modulating gut microbiota. Food Research International 155:111125

    doi: 10.1016/j.foodres.2022.111125

    CrossRef   Google Scholar

    [147]

    Chai Z, Yan Y, Zan S, Meng X, Zhang F. 2022. Probiotic-fermented blueberry pomace alleviates obesity and hyperlipidemia in high-fat diet C57BL/6J mice. Food Research International 157:111396

    doi: 10.1016/j.foodres.2022.111396

    CrossRef   Google Scholar

    [148]

    Yan Y, Zhang F, Chai Z, Liu M, Battino M, et al. 2019. Mixed fermentation of blueberry pomace with L. rhamnosus GG and L. plantarum-1: Enhance the active ingredient, antioxidant activity and health-promoting benefits. Food and Chemical Toxicology 131:110541

    doi: 10.1016/j.fct.2019.05.049

    CrossRef   Google Scholar

    [149]

    Zhu Z, Huang A, Chen M, Wang J, Li Z, et al. 2023. Impacts of selenium enrichment on nutritive value and obesity prevention of Cordyceps militaris: A nutritional, secondary metabolite, and network pharmacological analysis. Food Chemistry: X 19:100788

    doi: 10.1016/j.fochx.2023.100788

    CrossRef   Google Scholar

  • Cite this article

    Huang A, Wu Q, Thanuphol P, da Cruz LL, Xie Z, et al. 2024. Gut microbiota-associated metabolites in metabolic diseases and their impact from food processing. Food Innovation and Advances 3(4): 438−448 doi: 10.48130/fia-0024-0038
    Huang A, Wu Q, Thanuphol P, da Cruz LL, Xie Z, et al. 2024. Gut microbiota-associated metabolites in metabolic diseases and their impact from food processing. Food Innovation and Advances 3(4): 438−448 doi: 10.48130/fia-0024-0038

Figures(3)

Article Metrics

Article views(211) PDF downloads(144)

REVIEW   Open Access    

Gut microbiota-associated metabolites in metabolic diseases and their impact from food processing

Food Innovation and Advances  3 2024, 3(4): 438−448  |  Cite this article

Abstract: Gut microbiota-associated metabolites can be synthesized endogenously or derived from dietary nutrients and host compounds. Among them, alkaloids, terpenes, and flavones originating from edible and medicinal foods have attracted remarkable interest recently and play crucial roles in metabolic diseases. The efficacy of these metabolites is susceptible to dietary intervention, especially after food processing. Therefore, this review comprehensively summarizes the different sources of common gut microbial metabolites, including microbial self-synthesis, biodegradation of exogenous substances (mainly dietary nutrients), and participation in host metabolism. In addition, the latest studies on novel metabolites such as alkaloids, terpenoids, and flavonoids are discussed, and their action mechanisms on metabolic diseases are elaborated. How food processing impacts dietary nutrients and their metabolites is carefully examined, as well as their effects on disease modification. These insights could contribute to a deeper understanding of the mechanisms by which diet efficacy helps prevent metabolic diseases, particularly through gut microbial metabolites.

    • The microbial community in the gut significantly impacts various physiological processes, including food metabolism, vitamin synthesis, gut mucosal barrier integrity, immunomodulation, and pathogen defense[1]. Disruptions in the gut microbiota are associated with various metabolic diseases, such as obesity, diabetes, anxiety, and depression[2,3]. Microbial metabolites are synthesized de novo by bacteria or formed through bioconversion of foreign substances (mainly dietary nutrients) or host-derived compounds[4]. Emerging evidence highlights the involvement of gut microbiota in host physiology and metabolic pathways, largely through the production of diverse metabolites[5]. Consequently, these gut microbiota-associated metabolites are pivotal in unraveling the mechanisms that govern host health.

      A significant challenge in studying the health effects of microbes is differentiating metabolites produced by the microbiota and those generated in human tissues or directly derived from dietary sources. Metagenomic analysis, which catalogs the genes present in the microbial environment, aids in providing mechanistic insights into the metabolic changes linked to disease[6]. Metabolomic analysis explores which compounds may mediate the relationship between microbial activity and host disease, offering the possibility of interpreting various metabolite sources[7]. The gut microbiota can convert certain compounds into active metabolites with improved bioavailability and therapeutic properties than their original forms[810]. Representative compounds transformed by the gut microbiota include alkaloids, flavonoids, and terpenoids[11,12]. These naturally occurring active ingredients are widely present in various foods and, even at low levels, can significantly contribute to anti-inflammatory, antioxidant, and lipid-lowering effects, which can help improve metabolic diseases[13]. Further clarification of these novel metabolite types and their action mechanisms is crucial for understanding how the gut microbiota mediates dietary bioactivity.

      Dietary nutrients, frequently altered during food processing, are a primary factor influencing gut microbial-related metabolites. Throughout processing, dietary nutrients undergo various changes, affecting microbial metabolism in different ways[14]. This review summarizes three distinct sources of microbial metabolites and describes the regulatory roles of representative microbial metabolites from each category in host disease states. Moreover, it details the roles of three novel bacterial metabolites—alkaloids, terpenoids, and flavonoids—in specific aspects of human health, as well as their interactions with the gut microbiota. The aim is to clarify the mechanisms involved and enhance understanding of how specific bacterial species and metabolites influence human health. Additionally, how food processing influences the nutritional makeup of the diet and its potential effects on disease management are highlighted, which is essential for assessing the relationship between diet and health.

    • Metabolites involved in gut microbiota are crucial mediators of their efficacies[15]. These metabolites can broadly be classified into three categories: (i) those synthesized de novo by gut microbes; (ii) those produced by gut microbes directly from dietary components; and (iii) those produced by the host and subsequently modified by gut microbes[13]. Some of these metabolites may have multiple sources, and here representative examples of these metabolites in most cases are summarized and discussed (Fig. 1).

      Figure 1. 

      Types of microbial metabolites derived from various sources (created with BioRender.com).

    • Research indicates that the gut microbiota can synthesize vitamin K and the majority of water-soluble B vitamins like biotin, riboflavin, thiamine, folates, pantothenic acid, and nicotinic acid[16]. For instance, cobalamin (vitamin B12) is exclusively produced by microbes, specifically anaerobic bacteria[17]. Genomic analysis combined with in vivo and in vitro metabolic experiments confirmed that Cetobacterium somerae CS2105-BJ is capable of de novo vitamin B12 synthesis. It contains essential genes (hemL, cbiT/cobD, and cobC) that play a role in various stages of B12 biosynthesis, which are responsible for the synthesis of uroporphyrinogen III, adenosylmethylcobalamin, and lower ligand, respectively[18,19]. In situations where dietary vitamin B2 was depleted in a mouse model, the gut microbiota provided short-term compensation[20]. Certain vitamins, like B vitamins, which serve as precursors to essential metabolic cofactors, can be synthesized by specific gut bacteria known as prototrophic bacteria. However, they must also be obtained from other bacterial species (auxotrophic bacteria) and from the host’s diet[21]. This highlights that microbial synthesis of specific metabolites may depend on environmental conditions and the availability of exogenous sources, necessitating multi-bacterial cooperation. Additionally, amino acids and fatty acids can be synthesized de novo by gut microbes. Research has demonstrated ruminal bacteria such as Selenomonas ruminantium, Streptococcus bovis, and Prevotella bryantii engage in amino acid synthesis under physiological conditions, particularly enriching glutamic acid and aspartate in the presence of peptides[22]. Bacteroidetes are known symbionts capable of sphingolipid production. For example, Bacteroides thetaiotaomicron can initiate biological sphingolipid synthesis via serine palmitoyl transferase[23].

    • Microbial dietary metabolites include short-chain fatty acids (SCFAs), trimethylamine oxide (TMAO), and indole and its derivatives. SCFAs, primarily derived from dietary fiber fermentation, are a major focus. For example, resistant starch type 3 from potatoes promotes the production of propionic and acetic acids by Bifidobacterium, Ruminococcus, Bacteroides, and Coprococcus[24]. Pectin, rich in rhamnogalacturonan-I, significantly promotes the proliferation of Bifidobacterium, Faecalibaculum, and Lactobacilli in C57BL/6J mice, and it is metabolized to SCFAs[25]. Dietary inulin supports SCFA-producing Ruminococcaceae and Lachnospiraceae, increasing fecal SCFA concentrations[26]. Specifically, Bifidobacterium produce lactate and acetate, Bacteroides generate propionate and acetate, and Butyrivibrio and Fusobacterium produce butyrate as their primary metabolites[27]. Other bacteria that produce SCFAs include Anaerostipes, Anaerotruncus, Bacteroides, Coprococcus, Clostridium, Dialister, Eubacterium, Faecalibacterium, Lactobacillus paracasei, Odoribacter, Parabacteroides, and Ruminococcus[2832].

      Tryptophan metabolites, particularly indoles, such as 3-indolepropionic acid, indoleacetic acid, indole acrylate, indoleacrylic acid, indole-3-carbaldehyde, and tryptamine, form another extensively studied group of dietary metabolites[33]. For instance, 3-indolepropionic acid is biosynthesized from tryptophan by Clostridium sporogenes[34]. Commensal Peptostreptococcus metabolizes tryptophan into indoleacrylic acid[35], while Bifidobacterium longum CCFM1029 produces indole-3-carbaldehyde[36]. Putrescine, derived from arginine through a hybrid pathway involving Escherichia coli and Enterococcus faecalis, is another notable metabolite. Bifidobacterium spp. that produce acidic compounds accelerate putrescine production[37].

      Choline metabolites are classic nitrogenous dietary compounds. The synthesis of TMAO increases following the consumption of a diet high in phosphatidylcholine and L-carnitine[38]. Gut microbiota, like Citrobacter freundii, are capable of transforming into trimethylamine (TMA), which is then oxidized to TMAO[9]. Research has indicated that trigonelline can reduce TMAO levels by inhibiting the activity of C. freundii and flavin-containing monooxygenase 3 in mice. In vitro experiments also showed that the addition of fenuelline to choline-rich medium could significantly inhibit the production of C. freundii and reduce TMA and MAO production[9,39]. In another study involving obese hypertensive individuals, Lactobacillus plantarum was found to decarboxylate ornithine to produce putrescine[40]. Spermidine production is facilitated by some gut microbiota, such as E. coli and Bacteroides, utilizing carboxyspermidine decarboxylase and spermidine synthase[41,42]. Additionally, inosine, as a purine nucleotide metabolite, may be produced by Lactobacillus during the fermentation of barley leaves[43]. In general, there are a wide variety of microbial metabolites from dietary sources, and the efficacy of many substances, including SCFAs and indole derivatives, have been extensively studied with their relevant mechanism identified. Whether new dietary metabolites with good efficacy beyond these substances can be identified needs further exploration.

    • The liver serves as the primary organ to regulate cholesterol homeostasis, controlling both cholesterol intake and de novo synthesis. Endogenous cholesterol synthesis is mainly regulated by the SCAP-SREBP-HMGCR pathway[44,45]. In addition, a microbial enzyme called ismA has been identified previously that can convert cholesterol into coprostanol, a lipid that is excreted from the body rather than being absorbed[46]. Yao et al.[47]identified a gene encoding the sulfotransferase (BtSULT, BT0416), which is widely found in Bacteroides and mediates the sulfation of cholesterol.

      Bile acids (BAs), which are synthesized in the liver from cholesterol under the action of cholesterol- 7α-hydroxylase (CYP7A1), and are conjugated with taurine or glycine, undergo modifications by gut microbiota to produce secondary BA metabolites[43,48,49]. Pruss et al.[50] used stable isotope tracers along with bacterial and host genes to uncover a common metabolic pathway of hippuric acid production by host microorganisms. They confirmed that Clostridium sporogenes reduced phenylalanine into phenylpropionic acid[50-51]. The third type of metabolite, which is metabolized by the host and subsequently modified by microorganisms, has not been detailedly explained in most studies. This complexity likely arises from the intricate interactions between the host and gut microbiota, which makes it challenging to distinctly delineate their respective roles in metabolite modification.

    • Metabolic diseases, such as diabetes, atherosclerosis, hypertension, and obesity, are often linked with dysregulation of gut microbial metabolites[9]. Natural products-derived metabolites, including alkaloids, terpenoids, and flavonoids, undergo substantial modification by gut microbiota, affecting their pharmacological effects and roles in metabolic diseases[52]. These compounds typically exhibit low absorption rates in intestinum tenue and are further broken down by microbiota in the colon into microbial metabolites, which have been confirmed to play a significant role in regulating metabolic diseases[52]. The following section will specifically explore the interaction between these diet-derived metabolites and gut microbiota, as well as their functions in metabolic diseases (Fig. 2).

      Figure 2. 

      Natural product-derived gut microbial metabolites in metabolic diseases. Different colors refer to different classes of metabolites, with red representing alkaloids, green representing terpenoids, and yellow representing flavonoids. The blue boxes indicate key sites on the pathways, and the brown boxes indicate key sites shared by the three metabolites (created with BioRender.com).

    • Gut microbiota are essential for the absorption of alkaloids, such as Staphylococcus aureus, Enterococcus faecalis, and Enterococcus faecium, and they possess nitrate reductase enzymes that metabolize alkaloids[53,54]. Previous studies have indicated that the cytochrome CYP450 family, including CYP2D6, CYP1A2, CYP51, and CYP7A1, plays a significant role in the metabolic transformation of alkaloids[5557], including O-demethylation and hydroxylation[55 ]. Alkaloids can influence the body’s ecological physiology by regulating gut microbiota and their metabolic products. Supplementation with alkaloids increases the abundance of Bacteroides, Parabacteroides[58], Ruminococcus[59], Blautia[60], Faecalibaculum, Allobaculum[61], Clostridiales, and Lactobacillus[62], which are shown to produce SCFAs, TMAO, and BAs[61,63,64].

      The primary causes of obesity are abnormalities in energy balance and weight regulation, often accompanied by enlarged fat cells and changes in insulin function, which can further lead to the development of diabetes. Peroxisome proliferator activated receptor alpha (PPARα) and peroxlsome proliferator-activated receptor-gamma coactivator-1 alpha (PGC1α) are associated with the proliferation of adipose tissue and adipocyte hypertrophy following dietary intake, while targets such as liver X receptor (LXR), farnesoid X receptor (FXR), and CYP7A1 play crucial roles in regulating lipid, glucose, and energy metabolism, making them key targets for the treatment of metabolic diseases such as obesity and diabetes[6567]. Alkaloids can effectively act on the above targets and play a key role in disease improvement. For instance, palmatine treatment can regulate BA metabolism, reduce cholesterol accumulation, and alleviate obesity by enhancing the expression of PPARα and CYP7A1 while suppressing the expression of FXR[68]. Berberine can inhibit hepatic lipogenesis by up-regulating the expression of intestinal FXR and fibroblast growth factor 15, as well as activating the AMP-activated protein kinase (AMPK)-dependent Raf-1 pathway, thereby affecting the expression of the low-density lipoprotein receptor (LDLR)[62,69]. Notably, LDLR is a transmembrane glycoprotein primarily responsible for clearing lipoproteins, and an effective therapeutic target for treating hypercholesterolemia and related cardiovascular diseases[69]. In diabetic mice, supplementing with ruthenine improved the disease by activating the PPARα/PGC1α signaling pathway and upregulating β-oxidation-related genes in the liver[70]. In addition, studies have shown that Streptococcus faecalis can convert berberine in Rhizoma coptidis into oxyberberine, a potent hypoglycemic metabolite[54]. This metabolite contributes to anti-diabetes by upregulating the mRNA expression of the pancreatic nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway and activating the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway[71].

      Alkaloids exhibit anti-inflammatory properties by inhibiting pathways such as TRIF-dependent NF-κB signaling. For example, oxyberberine modulates the toll-like receptor 4 (TLR4)-myeloid differentiation factor 88 (MyD88)-NF-κB pathway[54] and the TLR4/PI3K/NF-κB pathways, reducing inflammatory responses[71]. For another, berberine can exert its effects by inhibiting the phosphorylation of insulin receptor substrate-1 (IRS-1) and increasing the phosphorylation of AKT in adipose tissue and cultured adipocytes[72]. These studies highlight the significant role of alkaloids in human health by interacting with gut microbiota and their metabolic products, thereby contributing to the maintenance of homeostasis. Despite extensive research on the structure and physiological activity of alkaloids metabolites, investigations into their in vivo roles and specific mechanisms in regulating human health are still limited.

    • Terpenoids have demonstrated efficacy in ameliorating metabolic disorders by regulating gut microbiota. Studies indicate that terpenoids such as fucoxanthin affect the Firmicutes/Bacteroidetes ratio and influence the abundance of key species, which are pivotal in anti-obesity activities[73]. Additionally, terpenoids have been shown to enhance populations of Lactobacillus, Bifidobacterium, Clostridium sensu stricto 1, Turicibacter, uncultured Erysipelotrichaceae, Faecalibaculum, and Ruminococcus, while reducing Lachnospiraceae NK4A136, uncultured Lachnospiraceae, and taxa such as Prevotellaceae UCG-003 and Oscillospira[7476]. In another context, the administration of ginsenoside extract enriched gut E. faecalis significantly[77]. Zhang et al.[78] confirmed that a guava extract abundant in 1-3 glucose-based residues of triterpenoid glycosides has significantly regulated Elusimicrobium, Lachnospiraceae UCG-004, acetic acid, butyric acid, and 1β-hydroxycholic acid in T2DM rats. Gentiopicroside has been shown to directly bind FGFR1, promoting FGF21 signaling, and subsequently activating PI3K/AKT and AMPK pathways to regulate glucose and lipid metabolism[79]. Reports indicate that gentiopicroside can be metabolized by intestinal bacteria into potential pharmacologically active metabolites such as erythrocentaurin, gentianine, and gentianal[80]. Consumption of erythrocentaurin can inhibit α-amylase activity, improving postprandial hyperglycemia, while supplementation with gentiopicroside may reduce inflammation by inhibiting TLR4/NLRP3-mediated pyroptosis[80,81].

      The anti-inflammatory mechanisms of terpenoids involve several key factors. Glycyrrhetic acid is a triterpenoid saponin extracted from licorice. It can be hydrolyzed by β-D glucuronidase of Eubacterium sp. strain GLH and then converted to Glycyrrhetic acid 3-O-mono-D-glucuronide, enhancing anti-inflammatory activity[82]. Han et al.[75]illustrated that lactucin curtails p38, extracellular signal-regulated kinase (ERK), and AKT phosphorylation within lipopolysaccharides (LPS)-stimulated RAW264.7 cells, consequently diminishing the mRNA and protein levels of inducible nitric oxide synthase and cyclooxygenase-2, and attenuating interleukin-6 (IL-6) production. Jolkinolide B mitigates LPS-induced degradation of NF-κB inhibitor alpha (IκBα) and phosphorylation of p65 and MAPK, subsequently reducing histological alterations and inflammatory markers[83]. Limonin, a triterpenoid extracted from citrus, mitigates intestinal inflammation by downregulating p-STAT3/miR-214 levels, demonstrating anti-inflammatory and apoptotic effects[84]. Fucoxanthin, derived from marine algae, activates Nrf2/Antioxidant response element (ARE) through PI3K/Akt pathways, enhancing antioxidant synthesis[8587]. Fucoxanthin also activates AMPK, reducing fatty acid synthesis via the sirtuin 1 (SIRT1)/AMPK pathway[88]and improving mitochondrial function while reducing oxidative stress in endothelial cells through the AMPK-Akt-cAMP response element-binding protein (CREB)-PGC1α pathway[89].

      Phytol, a plastid terpenoid synthesized via the methylerythritol phosphate pathway in plastids, is released when chlorophyll molecules are partially digested in animals[90]. It is absorbed in the small intestine and converted to phytanic acid in the liver[91]. Phytanic acid activates PPARγ and the retinoid-X-receptor (RXR), regulating lipid metabolism in various cell types[92]. The activation of PPARγ in adipose tissues improves insulin resistance, while PPARα activation in the liver lowers circulating lipid levels[93,94]. Terpenoids derived from Resina commiphora modulate lipid metabolism by augmenting PPARα and carnitine palmitoyl transferase 1 (CPT1) expression[95]. Auraptene, a novel PPARα and PPARγ ligand found in citrus fruits exerts anti-atherosclerotic and anti-diabetic effects by balancing anti-inflammatory/pro-inflammatory cytokine levels in adipocytes[96]. These functions of dietary terpenoids are important for the control of metabolic diseases through dietary intervention.

    • Flavonoids can be classified into several categories: flavones (including apigenin and luteolin), flavonols (including quercetin and curcumin), flavanones (including naringenin and hesperidin), flavanols (including catechin and epicatechin), isoflavones (including daidzein and genistein), and anthocyanins (including pelargonidin and cyanidin). They are primarily derived from fruits, vegetables, legumes, onions, seeds, plant roots and stems, tea, wine, olive oil, tomato sauce, and various dietary supplements[97,98]. After supplementing the diet with flavonoid-rich foods, such as soybean, bilberry, citrus fruit, and green tea, the enzymes produced by gut bacteria undergo various reactions including deglycosylation, demethylation, and oxidation, which generate biologically active metabolites with health benefits such as genistein, quercetin, anthocyanin, and catechin[99]. Santangelo et al.[100] summarized multiple studies showing that quercetin inhibits some intestinal bacteria such as Streptococcus, Lactobacillus, Bifidobacterium, and Bacteroides, and was converted into small molecules like phenylacetic acid, phenylpropanoic acid, dihydroxyphenylacetic acid, hydroxybenzoic acid, dihydroxybenzoic acid, and propionic acid, absorbed and utilized by the body[101]. The catabolism of cyanidin-3-O-glucoside in the gastrointestinal tract yields bioactive metabolites like protocatechuic acid, phloroglucinaldehyde, vanillic acid, and ferulic acid, supporting the integrity of the mucosal barrier and improving the health of the microbiota[102,103]. Escherichia fergusonii and E. coli metabolize curcumin into dihydrocurcumin, tetrahydrocurcumin, and ferulic acid[104]. In particular, Bifidobacterium animalis subsp. lactis AD011 efficiently converted 85% of quercetin 3-glucoside and isorhamnetin 3-glucoside into quercetin and isorhamnetin, respectively, within just 2 h without degrading the flavonoid backbone[105].

      Flavonoids exert significant effects on metabolic diseases through interactions with key targets such as PPAR, AMPK, Nrf2, and NF-κB. For instance, they can bind to non-phosphorylated signal transducer and activator of transcription 3 (STAT3) in visceral adipose tissue, reducing its phosphorylation and transcriptional activity. This action leads to a decreased expression of the STAT3 target gene CD36, which typically influences adipogenesis and anti-visceral obesity by suppressing PPAR-γ expression[106]. Daidzein influences cardiac energy metabolism by modulating Sirtuin 3 (SIRT3), thereby improving lipid, glucose and ketone body metabolism disorders, and mitigating mitochondrial dysfunction, which is crucial for meeting cardiac ATP demands both in vivo and in vitro[107]. Daidzein influences the LXR signaling pathway, activating RXR, PPARα, and AMPKα, which enhances energy metabolism[108,109]. In skeletal muscle, daidzein inhibits the Glut4/AMPK/Forkhead box O (FoxO) pathway and reduces the expression of atrogin1 and muscle ring finger protein1, thereby inhibiting the protein degradation of the skeletal muscle[110].

      Insulin induces GLUT-4 transport from the vesicle to the plasma membrane, thereby promoting glucose uptake in cells, through the PI3K/Akt pathway. Disruption of these pathways may lead to insulin resistance, which can subsequently result in diabetes[111]. Fraction D, rich in flavonoids isolated from Enicostema littorale, enhances cell glucose uptake by upregulating IRS-1/PI3K/Akt pathway components[112]. Both cyanidin-3-O-β-glucoside and pinocembrin inhibit inflammatory pathways like NF-κB and TLR4/MD2, providing therapeutic benefits for liver and intestinal inflammation[113,114]. Cyanidin-3-O-β-glucoside treatment stimulates SIRT1 activity, inhibits NF-κB acetylation, and subsequently inhibits the activation of inflammasomes and the release of pro-inflammatory cytokines in liver cell lines[113]. Quercetin in diet improves experimental colitis by enhancing the anti-inflammatory and antibacterial capacities of macrophages through the Nrf2/Heme Oxygenase 1 (HO-1) pathway[115]. Additionally, the inclusion of Eucommia ulmoides flavones leads to increased levels of phosphorylated Akt, IκBα, and IKKα/β, while simultaneously reducing the expression of Bax and Caspase-3 proteins in LPS-treated cells[116]. Other flavonoid metabolites, including protocatechuic acid, phloroglucinol, and vanillic acid, can downregulate the MAPK pathway by inhibiting the phosphorylation of ERK, c-Jun N-terminal kinase (JNK), and p38[117,118]. Overall, flavonoid metabolites play active roles in modulating various pathways, including NF-κB, MAPK, ERK, JNK, and p38, contributing to their anti-inflammatory and metabolic regulatory effects in different physiological conditions.

    • Food processing, including thermal processing and non-thermal processing, plays a crucial role in our daily diet[119]. These processes can alter the nutritional composition of food by introducing new compounds or converting existing ones[120] (Fig. 3). Considering that utilizing dietary interventions to improve metabolic diseases has become an effective means[121], its efficacy is highly correlated with gut microbiota metabolites as described previously. Therefore, comprehending the effects of food processing technology is crucial for assessing the nutritional value of processed foods and selecting appropriate processed foods based on differential health needs.

      Figure 3. 

      Impact of food processing on dietary nutrients and their health benefits via gut microbiota and related metabolites. The blue arrows represent the impact of thermal processing, while the purple arrows represent the impact of non-thermal processing (created with BioRender.com).

    • Heat treatments, such as boiling, baking, stewing, steaming, frying, and roasting, are widely used in both domestic and industrial food processing[122]. Previous research has indicated that the carbohydrate levels in grains, vegetables, and fruits typically rise following heat treatment[123, 124]. For example, heat-treated highland barley has been found to have a high dietary fiber content[124]. Similarly, it is found that boiling increases the content of fiber by 8.32% in garden cress seeds[125]. The increase in dietary fiber after thermal processing yields some beneficial outcomes, such as boosting the prevalence of beneficial gut bacteria including Bifidobacteria and Bacteroides, while suppressing harmful bacteria like Helicobacter and Enterococcus[126]. It also enhances the levels of SCFAs. These typical gut bacteria and metabolites are strongly associated with lipid metabolism and can effectively improve metabolic diseases[127,128]. The variability of these nutrients highlights that different processing methods may impact the efficacy of food intervention in varying ways, possibly due to changes in active components and their related metabolites[129,130].

      Nevertheless, it is crucial to recognize that most nutrients, including proteins, phenols, vitamins, and minerals, are lost after thermal processing[123]. During heat treatment, protein structures can undergo modifications due to direct oxidation or complex reactions with other components of the food, leading to changes in processing characteristics and a reduction in nutritional value, with potential negative health implications[131]. For example, heat treatment of hawthorn has been shown to cause the degradation of soluble phenols and proanthocyanidins[132], which are vital in the gastrointestinal tract due to their antioxidant, anti-inflammatory, and antibacterial properties[133]. These compounds also promote the growth of probiotics like Bifidobacterium and Lactobacillus, offering potential health benefits. Additionally, steaming fresh tea significantly reduces the levels of vitamins (B2, B3, and C) and minerals[123]. Vitamin B2 has been reported to prevent cancer, hyperglycemia, hypertension, diabetes, oxidative stress, and other health conditions, while vitamin C is known for its anti-inflammatory, antioxidant, and hypoglycemic effects[134,135]. The loss of these nutrients significantly reduces the health benefits of processed foods. Taken together, while heat treatment can increase dietary fiber, it can also have varying degrees of negative effects on the original nutrients and health benefits of the diet.

    • Non-thermal processing technologies, such as cold plasma treatment, irradiation, high-pressure processing, ultrasound, pulsed light technology, pulsed electric field treatment, and microbial fermentation, are widely used in food processing to circumvent the negative effects of heat[136]. These methods are designed to eliminate the need for high temperatures, thereby preserving the nutritional integrity of food[137]. For example, high-pressure electric field treatment has been demonstrated to elevate the anthocyanin content in strawberries, thereby improving its bioavailability[138,139]. In addition, ultrasound and ozone treatments have been found to increase the levels of phenols and vitamin C, and the antioxidant capacity of guava juice[140]. Mehta et al. found that cold plasma treatment for 10 min retained up to 95% of vitamin C [141]. These polyphenols and vitamins are crucial for human health[142], highlighting the potential of non-thermal processing to optimize the nutritional retention and even enhance the nutritional value of food products. As demonstrated by Tan et al.[143], high hydrostatic pressure treatment at 400 MPa for 15 min altered the monosaccharide composition of Cyanidin 3-glucoside and blueberry pectin complexes, decreasing mannose, fructose, glucose, xylose, and arabinose while increasing galactose, which improved colitis in mice through reducing intestinal oxidative stress, enhancing anti-inflammatory factors, and inhibiting the NF-κB signaling pathway mediated by the gut microbiota. Similar benefits have been observed with plasma treatment of orange juice and apple juice[144]. In terms of proteins, cold plasma treatment can oxidize hairtail muscle proteins, creating a more intensive protein network, which improves the texture and taste of the hairtail[145].

      Microbial fermentation in food processing can lead to the formation of specific nutritional components due to the involvement of microorganisms. For instance, probiotic fermentation significantly increases γ-aminobutyric acid, rutin, total polyphenols, and total flavonoids in rice buckwheat[146]. Fermented rice buckwheat has been shown to reverse high-fat diet-induced intestinal dysbiosis, suppress hepatic cholesterol synthesis and lipogenesis, and stimulate lipolysis by rebalancing the Firmicutes/Bacteroidetes ratio, enhancing SCFA-producing bacteria like Bacteroidetes, Lactobacillus, and Blautia, and increasing total SCFA content[147]. Furthermore, blueberry residue fermented by Lactobacillus rhamnosus GG and Lactobacillus plantarum-1 significantly suppressed inflammation-related cytokines (tumor necrosis factor-α, IL-1β, IL-6), upregulated PPAR-α, and downregulated sterol regulatory element-binding protein-1 and fatty acid synthase, thereby enhancing lipid metabolism[147]. Additionally, Yan et al.[148]observed a rise in lactic acid and a decrease in citric acid during the fermentation process. Recently, new non-thermal technologies, such as selenium processing, have emerged. For example, our previous study found that selenium processing markedly increased total carbohydrate and soluble dietary fiber content while decreasing protein and insoluble dietary fiber content in Cordyceps militaris[149]. It also enhanced the biosynthesis of secondary metabolites, such as terpenoids and alkaloids. The application of these new technologies not only preserves food quality and enhances flavor but also allows for adjusting the nutritional composition of food to meet enhanced functional requirements.

    • Gut microbial-associated metabolites are systematically categorized into three groups: de novo synthesis, microbial dietary metabolism, and host metabolic contributions. The beneficial role and potential mechanism of novel gut microbial metabolites derived from natural products in chronic diseases are becoming clear. Furthermore, gut microbiota and their associated metabolites provide valuable insights into how food processing influences nutritional value and health benefits. However, the intricate diversity and complexity of these metabolites make the precise molecular mechanisms underlying their effects still poorly comprehended. To address this, future studies should aim to: (i) accurately elucidate the entire production process of metabolites; (ii) clarify the spatial and temporal efficacy targets of metabolites; and (iii) comprehensively understand the regulatory patterns of different types of metabolites in the body. Achieving these goals in future research will pave the way for deeper exploration of gut microbial metabolites and their health functions, providing more insightful comprehension and innovation in this field.

      • This work was supported by the Guangdong Basic and Applied Basic Research Foundation (2023A1515010744), National Natural Science Foundation of China (32202014), GDAS' Project of Science and Technology Development (2022GDASZH-2022010101), and State Key Laboratory of Applied Microbiology Southern China (SKLAM002-2022).

      • The authors confirm their contribution to the paper as follows: writing - original draft: Huang A; visualization: Huang A, Chen M; writing - review & editing: Ding Y, Zhu Z, Thanuphol P, da Cruz LL, Xie Z; supervision: Ding Y, Zhang F, Zhu Z; resources: Wu Q, Ding Y, Zhu Z; project administration: Ding Y, Zhu Z; funding acquisition: Wu Q, Ding Y, Zhu Z. All authors reviewed the results and approved the final version of the manuscript.

      • All data generated or analyzed during this study are included in this published article.

      • The authors declare that they have no conflict of interest.

      • Copyright: © 2024 by the author(s). Published by Maximum Academic Press on behalf of China Agricultural University, Zhejiang University and Shenyang Agricultural University. This article is an open access article distributed under Creative Commons Attribution License (CC BY 4.0), visit https://creativecommons.org/licenses/by/4.0/.
    Figure (3)  References (149)
  • About this article
    Cite this article
    Huang A, Wu Q, Thanuphol P, da Cruz LL, Xie Z, et al. 2024. Gut microbiota-associated metabolites in metabolic diseases and their impact from food processing. Food Innovation and Advances 3(4): 438−448 doi: 10.48130/fia-0024-0038
    Huang A, Wu Q, Thanuphol P, da Cruz LL, Xie Z, et al. 2024. Gut microbiota-associated metabolites in metabolic diseases and their impact from food processing. Food Innovation and Advances 3(4): 438−448 doi: 10.48130/fia-0024-0038

Catalog

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return