Exploratory analysis of one versus two-day intermittent fasting protocols on the gut microbiome and plasma metabolome in adults with overweight/obesity

Alex E Mohr, Paniz Jasbi, Devin A Bowes, Blake Dirks, Corrie M Whisner, Karen M Arciero, Michelle Poe, Haiwei Gu, Eric Gumpricht, Karen L Sweazea, Paul J Arciero, Alex E Mohr, Paniz Jasbi, Devin A Bowes, Blake Dirks, Corrie M Whisner, Karen M Arciero, Michelle Poe, Haiwei Gu, Eric Gumpricht, Karen L Sweazea, Paul J Arciero

Abstract

Nutritional interventions are a promising therapeutic option for addressing obesity and cardiometabolic dysfunction. One such option, intermittent fasting (IF), has emerged as a viable alternative to daily caloric restriction and may beneficially modulate body weight regulation and alter the gut microbiome (GM) and plasma metabolome. This secondary analysis of a larger, registered trial (ClinicalTrials.gov ID: NCT04327141) examined the effect of a four-week intervention comparing one vs. two-consecutive days of IF in combination with protein pacing (IF-P; 4-5 meals/day, >30% protein/day) on the GM, the plasma metabolome, and associated clinical outcomes in overweight and obese adults. Participants (n = 20) were randomly assigned to either a diet consisting of one fasting day (total of 36 h) and six low-calorie P days per week (IF1-P, n = 10) or two fasting days (60 h total) and five low-calorie P days per week (IF2-P, n = 10). The fecal microbiome, clinical outcomes, and plasma metabolome were analyzed at baseline (week 0) and after four weeks. There were no significant time or interaction effects for alpha diversity; however, baseline alpha diversity was negatively correlated with percent body fat change after the four-week intervention (p = 0.030). In addition, beta-diversity for both IF groups was altered significantly by time (p = 0.001), with no significant differences between groups. The IF1-P group had a significant increase in abundance of Ruminococcaceae Incertae Sedis and Eubacterium fissicatena group (q ≤ 0.007), while the IF2-P group had a significant increase in abundance of Ruminococcaceae Incertae Sedis and a decrease in Eubacterium ventriosum group (q ≤ 0.005). The plasma metabolite profile of IF2-P participants displayed significant increases in serine, trimethylamine oxide (TMAO), levulinic acid, 3-aminobutyric acid, citrate, isocitrate, and glucuronic acid (q ≤ 0.049) compared to IF1-P. Fecal short-chain fatty acid concentrations did not differ significantly by time or between groups (p ≥ 0.126). Interestingly, gastrointestinal symptoms were significantly reduced for the IF2-P group but not for the IF1-P group. Our results demonstrate that short-term IF modestly influenced the GM community structure and the plasma metabolome, suggesting these protocols could be viable for certain nutritional intervention strategies.

Keywords: caloric restriction; gastrointestinal symptoms; gut microbiome; intermittent fasting; metabolome; obesity; protein pacing; weight loss.

Conflict of interest statement

This study received funding from Isagenix International, LLC and they were not involved in the study design, collection, analysis, interpretation of data, and the writing of this article or the decision to submit it for publication. Authors AM and EG were employed by Isagenix International, LLC and contributed to the study as noted above. AM was a doctoral candidate at Arizona State University, and this work was in partial fulfillment of his dissertation. The study was conducted at Skidmore College and AM was blinded throughout data collection and analyses. Author PA is a member of the scientific advisory board at Isagenix International, LLC and the International Protein Board. Author CW is on the scientific advisory board for the Wheat Foods Council and the Hass Avocado Board’s Avocado Nutrition Science Advisory. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Mohr, Jasbi, Bowes, Dirks, Whisner, Arciero, Poe, Gu, Gumpricht, Sweazea and Arciero.

Figures

FIGURE 1
FIGURE 1
Overview of study design. After enrollment, a one-week run-in period, and randomization, 20 overweight and obese males and females followed an intermittent fasting, protein pacing-based weight loss diet consisting of one (IF1-P, n = 10) or two (IF2-P, n = 10) fasting days per week for four weeks equated for weekly energy and macronutrient intake. Clinical data and fecal and blood samples were collected at baseline and after week four. “Created with BioRender.com”.
FIGURE 2
FIGURE 2
Variation in gut microbiome diversity metrics at baseline and week four of IF1-P and IF2-P participants. (A) Non-metric multidimensional scaling (NMDS) ordination of Bray-Curtis dissimilarity matrix. The GM of both IF1-P (n = 10) and IF2-P (n = 10) groups shifted significantly from baseline to week four (R2 = 0.042, p < 0.001), but there was no difference between groups by time (R2 = 0.009, p = 0.823). The same participant is connected by a dotted line, starting at baseline and the arrow pointing to the end of the intervention period. (B) The first distances of the Bray-Curtis dissimilarities between IF1-P and IF2-P were not significant (p = 0.579). Boxes denote the interquartile range (IQR) between the first and third quartiles, and the horizontal line defines the median. (C) First principal coordinate (PCo1) values differed by time (p = 0.013), with no differences between groups over time detected (p = 0.473). (D) Shannon diversity index did not change significantly over time for IF1-P and IF2-P groups (p ≥ 0.341). (E) Faith’s PD diversity index did not change significantly over time for IF1-P and IF2-P groups (p = 0.653). Boxes denote the IQR between the first and third quartiles, and the horizontal line defines the median. A dotted line connects the same participant. (F) Correlation of baseline Shannon Diversity with percent body fat loss from the four-week IF intervention. (G) Correlation of baseline Faith’s phylogenetic diversity (PD) with percent body fat loss from the four-week IF intervention. Both groups were combined for the correlation analyses and are displayed in different colors. The gray cloud around the regression line indicates the 95% confidence interval.
FIGURE 3
FIGURE 3
(A) Total bacterial number observed in both IF1-P (n = 10) and IF2-P (n = 10) groups at baseline and week four. No significant effects of time or interaction (p ≥ 0.603) were noted. Total bacterial numbers were calculated as average copies of 16S rRNA gene/g wet feces via qPCR. Group means at each time point are displayed as black bars. (B) Average relative abundance of the most prevalent gut microbiome phyla among study participants for IF1-P (n = 10) and IF2-P (n = 10) at baseline and week four by 16S rRNA sequencing. Phyla with a median relative abundance of less than 1% are collapsed into the category “Other”. (C) Average relative abundance of the most prevalent gut microbiome genera among study participants for IF1-P (n = 10) and IF2-P (n = 10) at baseline and week four by 16S rRNA sequencing. Genera with a median relative abundance of less than 1% are collapsed into the category “Other”.
FIGURE 4
FIGURE 4
(A) Genera were differentially abundant between baseline and week four for IF1-P participants (n = 10). (B) Genera that were differentially abundant between baseline and week four for IF2-P participants (n = 10). Points represent each genera’s log2 fold change (log2FC; effect size). A positive value indicates that a feature increased in abundance at week four (right), and a negative value indicates a decrease in abundance at week four (left). Bars represent 95% confidence intervals derived from the ANCOM-BC model. Note, the genus Incertae Sedis is from the Ruminococcaceae family. Boxplots displaying statistically significant differences in (C) peptidoglycan biosynthesis II and (D) chorismate biosynthesis II from baseline to week four of predicted metabolic pathways for IF1-P participants. Boxplot displaying a statistically significant difference in (E) adenosine nucleotides degradation IV from baseline to week four of the predicted metabolic pathway for IF2-P participants. Pathways are displayed as centered log-ratio (CLR) transformed abundances. Boxes denote the interquartile range (IQR) between the first and third quartiles, and the horizontal line defines the median.
FIGURE 5
FIGURE 5
(A) GLM adjusted for age, sex, BMI, and time, with FDR-correction, and (B) box plots of significant metabolites as indicated by GLM: serine (q = 0.003), TMAO (q = 0.012), levulinic acid (q = 0.017), 3-aminobutyric acid (q = 0.029), citrate (q = 0.033), isocitrate (q = 0.033), and glucuronic acid (q = 0.049). Red lines in box plots denote optimal cutoff values as calculated by the Youden method, black lines indicate median values, and yellow diamonds show group averages.
FIGURE 6
FIGURE 6
(A) Scores plot of OPLS-DA model constructed using seven significant metabolites identified by GLM, showing percent variance accounted for by experimental and orthogonal data. (B) OPLS-DA model overview showing predictive and explanatory capacity (R2X = 0.351, R2Y = 0.237, Q2 = 0.202); y-axis represents proportion of total variance. (C) Permutation test with 100 iterations showing model fit distributions (Perm R2Y p < 0.01, Perm Q2 p < 0.01). (D) ROC analysis of OPLS-DA model for assessing IF-P (AUC = 0.83, 95% CI: 0.70-0.94, sensitivity = 0.8, specificity = 0.8). (E) Box plot of OPLS-DA predictive values; the red line in the box plot denotes the optimal cutoff value, while yellow diamonds show group averages, and black lines illustrate group medians.

References

    1. Virani SS, Alonso A, Benjamin EJ, Bittencourt MS, Callaway CW, Carson AP, et al. Heart disease and stroke statistics-2020 update: a report from the American heart association. Circulation. (2020) 141:e139–596. 10.1161/CIR.0000000000000746
    1. Dietz WH, Baur LA, Hall K, Puhl RM, Taveras EM, Uauy R, et al. Management of obesity: improvement of health-care training and systems for prevention and care. Lancet. (2015) 385:2521–33. 10.1016/S0140-6736(14)61748-7
    1. American College of Cardiology. Expert Panel Report: guidelines (2013) for the management of overweight and obesity in adults. Obes Silver Spring Md. (2014) 22(Suppl 2):S41–410. 10.1002/oby.20660
    1. MacLean PS, Rothman AJ, Nicastro HL, Czajkowski SM, Agurs-Collins T, Rice EL, et al. The accumulating data to optimally predict obesity treatment (ADOPT) core measures project: rationale and approach. Obesity. (2018) 26(Suppl 2):S6–15. 10.1002/oby.22154
    1. Patterson RE, Sears DD. Metabolic effects of intermittent fasting. Annu Rev Nutr. (2017) 37:371–93. 10.1146/annurev-nutr-071816-064634
    1. Mattson MP, Longo VD, Harvie M. Impact of intermittent fasting on health and disease processes. Ageing Res Rev. (2017) 39:46–58. 10.1016/j.arr.2016.10.005
    1. Zajac I, Herreen D, Hunkin H, James-Martin G, Doyen M, Kakoschke N, et al. Modified fasting compared to true fasting improves blood glucose levels and subjective experiences of hunger, food cravings and mental fatigue, but not cognitive function: results of an acute randomised cross-over trial. Nutrients. (2020) 13:65. 10.3390/nu13010065
    1. Arciero PJ, Edmonds R, He F, Ward E, Gumpricht E, Mohr A, et al. Protein-pacing caloric-restriction enhances body composition similarly in obese men and women during weight loss and sustains efficacy during long-term weight maintenance. Nutrients. (2016) 8:476. 10.3390/nu8080476
    1. Arciero PJ, Baur D, Connelly S, Ormsbee MJ. Timed-daily ingestion of whey protein and exercise training reduces visceral adipose tissue mass and improves insulin resistance: the PRISE study. J Appl Physiol. (2014) 117:1–10. 10.1152/japplphysiol.00152.2014
    1. Arciero PJ, Ormsbee MJ, Gentile CL, Nindl BC, Brestoff JR, Ruby M. Increased protein intake and meal frequency reduces abdominal fat during energy balance and energy deficit. Obesity. (2013) 21:1357–66. 10.1002/oby.20296
    1. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. (2013) 505:559–63. 10.1038/nature12820
    1. Asnicar F, Berry SE, Valdes AM, Nguyen LH, Piccinno G, Drew DA, et al. Microbiome connections with host metabolism and habitual diet from 1,098 deeply phenotyped individuals. Nat Med. (2021) 27:321–32. 10.1038/s41591-020-01183-8
    1. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, et al. A core gut microbiome in obese and lean twins. Nature. (2008) 457:480–4. 10.1038/nature07540
    1. Sanna S, van Zuydam NR, Mahajan A, Kurilshikov A, Vila AV, Võsa U, et al. Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nat Genet. (2019) 51:600–5. 10.1038/s41588-019-0350-x
    1. Desai MS, Seekatz AM, Koropatkin NM, Kamada N, Hickey CA, Wolter M, et al. A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell. (2016) 167:1339–53.e21. 10.1016/j.cell.2016.10.043
    1. Bolte LA, Vila AV, Imhann F, Collij V, Gacesa R, Peters V, et al. Long-term dietary patterns are associated with pro-inflammatory and anti-inflammatory features of the gut microbiome. Gut. (2021) 70:1287–98. 10.1136/gutjnl-2020-322670
    1. Walker RL, Vlamakis H, Lee JWJ, Besse LA, Xanthakis V, Vasan RS, et al. Population study of the gut microbiome: associations with diet, lifestyle, and cardiometabolic disease. Genome Med. (2021) 13:188. 10.1186/s13073-021-01007-5
    1. Manor O, Dai CL, Kornilov SA, Smith B, Price ND, Lovejoy JC, et al. Health and disease markers correlate with gut microbiome composition across thousands of people. Nat Commun. (2020) 11:5206. 10.1038/s41467-020-18871-1
    1. Wikoff WR, Anfora AT, Liu J, Schultz PG, Lesley SA, Peters EC, et al. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci. (2009) 106:3698–703. 10.1073/pnas.0812874106
    1. Lamichhane S, Sen P, Dickens AM, Orešiè M, Bertram HC. Gut metabolome meets microbiome: a methodological perspective to understand the relationship between host and microbe. Methods. (2018) 149:3–12. 10.1016/j.ymeth.2018.04.029
    1. Dong TS, Luu K, Lagishetty V, Sedighian F, Woo SL, Dreskin BW, et al. A high protein calorie restriction diet alters the gut microbiome in obesity. Nutrients. (2020) 12:3221. 10.3390/nu12103221
    1. Lassen PB, Belda E, Prifti E, Dao MC, Specque F, Henegar C, et al. Protein supplementation during an energy-restricted diet induces visceral fat loss and gut microbiota amino acid metabolism activation: a randomized trial. Sci Rep-Uk. (2021) 11:15620. 10.1038/s41598-021-94916-9
    1. Ma Y, Sun Y, Sun L, Liu X, Zeng R, Lin X, et al. Effects of gut microbiota and fatty acid metabolism on dyslipidemia following weight-loss diets in women: results from a randomized controlled trial. Clin Nutrition Edinb Scotl. (2021) 40:5511–20. 10.1016/j.clnu.2021.09.021
    1. Mohr AE, Gumpricht E, Sears DD, Sweazea KL. Recent advances and health implications of dietary fasting regimens on the gut microbiome. Am J Physiol Gastrointest Liver Physiol. (2021) 320:G847–63. 10.1152/ajpgi.00475.2020
    1. Cignarella F, Cantoni C, Ghezzi L, Salter A, Dorsett Y, Chen L, et al. Intermittent fasting confers protection in CNS autoimmunity by altering the gut microbiota. Cell Metab. (2018) 27:1222–35.e6. 10.1016/j.cmet.2018.05.006
    1. Gabel K, Marcell J, Cares K, Kalam F, Cienfuegos S, Ezpeleta M, et al. Effect of time restricted feeding on the gut microbiome in adults with obesity: a pilot study. Nutr Heal. (2020) 26:260106020910907. 10.1177/0260106020910907
    1. Ozkul C, Yalinay M, Karakan T. Structural changes in gut microbiome after Ramadan fasting: a pilot study. Benef Microbes. (2020) 11:227–33. 10.3920/BM2019.0039
    1. Zeb F, Wu X, Chen L, Fatima S, Haq IU, Chen A, et al. Effect of time-restricted feeding on metabolic risk and circadian rhythm associated with gut microbiome in healthy males. Br J Nutr. (2020) 123:1216–26. 10.1017/S0007114519003428
    1. Guo Y, Luo S, Ye Y, Yin S, Fan J, Xia M. Intermittent fasting improves cardiometabolic risk factors and alters gut microbiota in metabolic syndrome patients. J Clin Endocrinol Metab. (2020) 106:64–79. 10.1210/clinem/dgaa644
    1. Maifeld A, Bartolomaeus H, Löber U, Avery EG, Steckhan N, Markó L, et al. Fasting alters the gut microbiome reducing blood pressure and body weight in metabolic syndrome patients. Nat Commun. (2021) 12:1970. 10.1038/s41467-021-22097-0
    1. Arciero PJ, Arciero KM, Poe M, Mohr AE, Ives SJ, Arciero A, et al. Intermittent fasting two days versus one day per week, matched for total energy intake and expenditure, increases weight loss in overweight/obese men and women. Nutr J. (2022) 21:36. 10.1186/s12937-022-00790-0
    1. Svedlund J, Sjödin I, Dotevall GGSRS. —clinical rating scale for gastrointestinal symptoms in patients with irritable bowel syndrome and peptic ulcer disease. Digest Dis Sci. (1988) 33:129–34. 10.1007/BF01535722
    1. Kulich KR, Madisch A, Pacini F, Piqué JM, Regula J, Rensburg CJV, et al. Reliability and validity of the Gastrointestinal Symptom Rating Scale (GSRS) and Quality of Life in Reflux and Dyspepsia (QOLRAD) questionnaire in dyspepsia: a six-country study. Health Qual Life Out. (2008) 6:12. 10.1186/1477-7525-6-12
    1. Riegler G, Esposito I. Bristol scale stool form. A still valid help in medical practice and clinical research. Tech Coloproctol. (2001) 5:163–4. 10.1007/s101510100019
    1. Ilhan ZE, Marcus AK, Kang DW, Rittmann BE, Krajmalnik-Brown R. pH-mediated microbial and metabolic interactions in fecal enrichment cultures. Msphere. (2017) 2(3):e00047–17. 10.1128/mSphere.00047-17
    1. Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Lozupone CA, Turnbaugh PJ, et al. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc Natl Acad Sci. (2010) 108(Suppl_1):4516–22. 10.1073/pnas.1000080107
    1. Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Huntley J, Fierer N, et al. Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. Isme J. (2012) 6:1621–4. 10.1038/ismej.2012.8
    1. Methé BA, Nelson KE, Pop M, Creasy HH, Giglio MG, Huttenhower C, et al. A framework for human microbiome research. Nature. (2012) 486:215–21. 10.1038/nature11209
    1. Bolyen E, Rideout JR, Dillon MR, Bokulich NA, Abnet CC, Al-Ghalith GA, et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat Biotechnol. (2019) 37:852–7.
    1. Bokulich NA, Kaehler BD, Rideout JR, Dillon M, Bolyen E, Knight R, et al. Optimizing taxonomic classification of marker-gene amplicon sequences with QIIME 2’s q2-feature-classifier plugin. Microbiome. (2018) 6:90. 10.1186/s40168-018-0470-z
    1. Quast C, Pruesse E, Yilmaz P, Gerken J, Schweer T, Yarza P, et al. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. (2012) 41:D590–6. 10.1093/nar/gks1219
    1. McKnight DT, Huerlimann R, Bower DS, Schwarzkopf L, Alford RA, Zenger KR. Methods for normalizing microbiome data: an ecological perspective. Methods Ecol Evol. (2019) 10:389–400. 10.1111/2041-210X.13115
    1. Douglas GM, Maffei VJ, Zaneveld JR, Yurgel SN, Brown JR, Taylor CM, et al. PICRUSt2 for prediction of metagenome functions. Nat Biotechnol. (2020) 38:685–8. 10.1038/s41587-020-0548-6
    1. Caspi R, Billington R, Ferrer L, Foerster H, Fulcher CA, Keseler IM, et al. The MetaCyc database of metabolic pathways and enzymes and the BioCyc collection of pathway/genome databases. Nucleic Acids Res. (2016) 44:D471–80. 10.1093/nar/gkv1164
    1. Yang YW, Chen MK, Yang BY, Huang XJ, Zhang XR, He LQ, et al. Use of 16S rRNA gene-targeted group-specific primers for real-time PCR analysis of predominant bacteria in mouse feces. Appl Environ Microb. (2015) 81:6749–56. 10.1128/AEM.01906-15
    1. Gregoris TBD, Aldred N, Clare AS, Burgess JG. Improvement of phylum- and class-specific primers for real-time PCR quantification of bacterial taxa. J Microbiol Meth. (2011) 86:351–6. 10.1016/j.mimet.2011.06.010
    1. Bapat A, Schippel N, Shi X, Jasbi P, Gu H, Kala M, et al. Hypoxia promotes erythroid differentiation through the development of progenitors and proerythroblasts. Exp Hematol. (2021) 97:32–46.e35. 10.1016/j.exphem.2021.02.012
    1. Jasbi P, Shi X, Chu P, Elliott N, Hudson H, Jones D, et al. Metabolic profiling of neocortical tissue discriminates alzheimer’s disease from mild cognitive impairment, high pathology controls, and normal controls. J Proteome Res. (2021) 20:4303–17. 10.1021/acs.jproteome.1c00290
    1. Jasbi P, Mohr AE, Shi X, Mahmood T, Zhu Q, Bruening M, et al. Microbiome and metabolome profiles of high screen time in a cohort of healthy college students. Sci Rep. (2022) 12:3452. 10.1038/s41598-022-07381-3
    1. Basile AJ, Jasbi P, Clark W, Shi X, Gu H, Deviche P, et al. A four-week white bread diet does not alter plasma glucose concentrations, metabolic or vascular physiology in mourning doves, Zenaida macroura. Comp Biochem Physiology Part Mol Integr Physiol. (2020) 247:110718. 10.1016/j.cbpa.2020.110718
    1. Johnston CS, Jasbi P, Jin Y, Bauer S, Williams S, Fessler SN, et al. Daily vinegar ingestion improves depression scores and alters the metabolome in healthy adults: a randomized controlled trial. Nutrients. (2021) 13:4020. 10.3390/nu13114020
    1. Gu H, Shi X, Jasbi P, Patterson J. Viruses as therapeutics, methods and protocols. Methods Mol Biol. (2020) 2225:179–97. 10.1007/978-1-0716-1012-1_10
    1. He H, Pan T, Shi X, Yang S, Jasbi P, Jin Y, et al. An integrative cellular metabolomic study reveals downregulated tricarboxylic acid cycle and potential biomarkers induced by tetrabromobisphenol A in human lung A549 cells. Environ Toxicol. (2022) [Online ahead of print]. 10.1002/tox.23657
    1. Mohr AE, Jasbi P, Wyst KBV, van Woerden I, Shi X, Gu H, et al. Association of food insecurity on gut microbiome and metabolome profiles in a diverse college-based sample. Sci Rep. (2022) 12:14358. 10.1038/s41598-022-18515-y
    1. McMurdie PJ, Holmes S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS One. (2013) 8:e61217. 10.1371/journal.pone.0061217
    1. Bokulich NA, Dillon MR, Zhang Y, Rideout JR, Bolyen E, Li H, et al. q2-longitudinal: longitudinal and paired-sample analyses of microbiome data. Msystems. (2018) 3:e00219-18. 10.1128/mSystems.00219-18
    1. Lin H, Peddada SD. Analysis of compositions of microbiomes with bias correction. Nat Commun. (2020) 11:3514. 10.1038/s41467-020-17041-7
    1. Pang Z, Chong J, Zhou G, de Lima Morais DA, Chang L, Barrette M, et al. MetaboAnalyst 5.0: narrowing the gap between raw spectra and functional insights. Nucleic Acids Res. (2021) 49:W388–96. 10.1093/nar/gkab382
    1. Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. (2000) 28:27–30. 10.1093/nar/28.1.27
    1. Trumbo P, Schlicker S, Yates AA, Poos M. Dietary reference intakes for energy, carbohydrate, fiber, fat, fatty acids, cholesterol, protein and amino acids. J Am Diet Assoc. (2002) 102:1621–30. 10.1016/S0002-8223(02)90346-9
    1. Cummings JH, Bingham SA, Heaton KW, Eastwood MA. Fecal weight, colon cancer risk, and dietary intake of nonstarch polysaccharides (dietary fiber). Gastroenterology. (1992) 103:1783–9. 10.1016/0016-5085(92)91435-7
    1. Sowah SA, Milanese A, Schübel R, Wirbel J, Kartal E, Johnson TS, et al. Calorie restriction improves metabolic state independently of gut microbiome composition: a randomized dietary intervention trial. Genome Med. (2022) 14:30. 10.1186/s13073-022-01030-0
    1. Seganfredo FB, Blume CA, Moehlecke M, Giongo A, Casagrande DS, Spolidoro JVN, et al. Weight-loss interventions and gut microbiota changes in overweight and obese patients: a systematic review. Obes Rev. (2017) 18:832–51. 10.1111/obr.12541
    1. Korpela K, Flint HJ, Johnstone AM, Lappi J, Poutanen K, Dewulf E, et al. Gut microbiota signatures predict host and microbiota responses to dietary interventions in obese individuals. PLoS One. (2014) 9:e90702. 10.1371/journal.pone.0090702
    1. Cotillard A, Kennedy SP, Kong LC, Prifti E, Pons N, Chatelier EL, et al. Dietary intervention impact on gut microbial gene richness. Nature. (2013) 500:585–8.
    1. Weirdt RD, de Wiele TV. Micromanagement in the gut: microenvironmental factors govern colon mucosal biofilm structure and functionality. Npj Biofilms Microbiomes. (2015) 1:15026. 10.1038/npjbiofilms.2015.26
    1. Mukherjee A, Lordan C, Ross RP, Cotter PD. Gut microbes from the phylogenetically diverse genus Eubacterium and their various contributions to gut health. Gut Microbes. (2020) 12:1802866. 10.1080/19490976.2020.1802866
    1. Liu Y, Jiang Q, Liu Z, Shen S, Ai J, Zhu Y, et al. Alteration of gut microbiota relates to metabolic disorders in primary aldosteronism patients. Front Endocrinol. (2021) 12:667951. 10.3389/fendo.2021.667951
    1. Muñoz M, Guerrero-Araya E, Cortés-Tapia C, Plaza-Garrido A, Lawley TD, Paredes-Sabja D. Comprehensive genome analyses of Sellimonas intestinalis, a potential biomarker of homeostasis gut recovery. Microb Genom. (2020) 6:mgen000476. 10.1099/mgen.0.000476
    1. Tims S, Derom C, Jonkers DM, Vlietinck R, Saris WH, Kleerebezem M, et al. Microbiota conservation and BMI signatures in adult monozygotic twins. Isme J. (2013) 7:707–17. 10.1038/ismej.2012.146
    1. Kasai C, Sugimoto K, Moritani I, Tanaka J, Oya Y, Inoue H, et al. Comparison of the gut microbiota composition between obese and non-obese individuals in a Japanese population, as analyzed by terminal restriction fragment length polymorphism and next-generation sequencing. BMC Gastroenterol. (2015) 15:100. 10.1186/s12876-015-0330-2
    1. Chen HM, Yu YN, Wang JL, Lin YW, Kong X, Yang CQ, et al. Decreased dietary fiber intake and structural alteration of gut microbiota in patients with advanced colorectal adenoma. Am J Clin Nutr. (2013) 97:1044–52. 10.3945/ajcn.112.046607
    1. Wang T, Cai G, Qiu Y, Fei N, Zhang M, Pang X, et al. Structural segregation of gut microbiota between colorectal cancer patients and healthy volunteers. Isme J. (2012) 6:320–9. 10.1038/ismej.2011.109
    1. Yu J, Feng Q, Wong SH, Zhang D, Liang QY, Qin Y, et al. Metagenomic analysis of faecal microbiome as a tool towards targeted non-invasive biomarkers for colorectal cancer. Gut. (2017) 66:70. 10.1136/gutjnl-2015-309800
    1. Ai D, Pan H, Li X, Gao Y, Liu G, Xia LC. Identifying gut microbiota associated with colorectal cancer using a zero-inflated lognormal model. Front Microbiol. (2019) 10:826. 10.3389/fmicb.2019.00826
    1. Wolf AJ, Underhill DM. Peptidoglycan recognition by the innate immune system. Nat Rev Immunol. (2018) 18:243–54. 10.1038/nri.2017.136
    1. Beli E, Yan Y, Moldovan L, Vieira CP, Gao R, Duan Y, et al. Restructuring of the gut microbiome by intermittent fasting prevents retinopathy and prolongs survival in db/db Mice. Diabetes. (2018) 67:1867–79. 10.2337/db18-0158
    1. Mardinoglu A, Bjornson E, Zhang C, Klevstig M, Söderlund S, Ståhlman M, et al. Personal model-assisted identification of NAD+ and glutathione metabolism as intervention target in NAFLD. Mol Syst Biol. (2017) 13:916. 10.15252/msb.20167422
    1. Ho A, Sinick J, Esko T, Fischer K, Menni C, Zierer J, et al. Circulating glucuronic acid predicts healthspan and longevity in humans and mice. Aging. (2019) 11:7694–706. 10.18632/aging.102281
    1. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. (2013) 19:576–85. 10.1038/nm.3145
    1. Li J, Li Y, Ivey KL, Wang DD, Wilkinson JE, Franke A, et al. Interplay between diet and gut microbiome, and circulating concentrations of trimethylamine N-oxide: findings from a longitudinal cohort of US men. Gut. (2022) 71:724–33. 10.1136/gutjnl-2020-322473
    1. Cho CE, Taesuwan S, Malysheva OV, Bender E, Tulchinsky NF, Yan J, et al. Trimethylamine-N-oxide (TMAO) response to animal source foods varies among healthy young men and is influenced by their gut microbiota composition: a randomized controlled trial. Mol Nutr Food Res. (2017) 61:1600324. 10.1002/mnfr.201600324
    1. Zhu W, Gregory JC, Org E, Buffa JA, Gupta N, Wang Z, et al. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell. (2016) 165:111–24. 10.1016/j.cell.2016.02.011
    1. Bergeron N, Williams PT, Lamendella R, Faghihnia N, Grube A, Li X, et al. Diets high in resistant starch increase plasma levels of trimethylamine-N-oxide, a gut microbiome metabolite associated with CVD risk. Brit J Nutr. (2016) 116:2020–9. 10.1017/S0007114516004165
    1. Mohr AE, Ramos C, Tavarez K, Arciero PJ. Lower postprandial thermogenic response to an unprocessed whole food meal compared to an iso-energetic/macronutrient meal replacement in young women: a single-blind randomized cross-over trial. Nutrients. (2020) 12:2469. 10.3390/nu12082469
    1. Mohr AE, Minicucci O, Long D, Miller VJ, Keller A, Sheridan C, et al. Resistant starch combined with whey protein increases postprandial metabolism and lowers glucose and insulin responses in healthy adult men. Foods. (2021) 10:537. 10.3390/foods10030537
    1. Shin SY, Fauman EB, Petersen AK, Krumsiek J, Santos R, Huang J, et al. An atlas of genetic influences on human blood metabolites. Nat Genet. (2014) 46:543–50. 10.1038/ng.2982
    1. Suhre K, Shin SY, Petersen AK, Mohney RP, Meredith D, Altmaier E, et al. Human metabolic individuality in biomedical and pharmaceutical research. Nature. (2011) 477:54–60. 10.1038/nature10354
    1. Visconti A, Roy CIL, Rosa F, Rossi N, Martin TC, Mohney RP, et al. Interplay between the human gut microbiome and host metabolism. Nat Commun. (2019) 10:4505. 10.1038/s41467-019-12476-z
    1. Shah SH, Crosslin DR, Haynes CS, Nelson S, Turer CB, Stevens RD, et al. Branched-chain amino acid levels are associated with improvement in insulin resistance with weight loss. Diabetologia. (2012) 55:321–30. 10.1007/s00125-011-2356-5
    1. Zheng Y, Ceglarek U, Huang T, Li L, Rood J, Ryan DH, et al. Weight-loss diets and 2-y changes in circulating amino acids in 2 randomized intervention trials1–3. Am J Clin Nutr. (2016) 103:505–11. 10.3945/ajcn.115.117689
    1. Strasser B, Berger K, Fuchs D. Effects of a caloric restriction weight loss diet on tryptophan metabolism and inflammatory biomarkers in overweight adults. Eur J Nutr. (2015) 54:101–7. 10.1007/s00394-014-0690-3
    1. Bruss MD, Khambatta CF, Ruby MA, Aggarwal I, Hellerstein MK. Calorie restriction increases fatty acid synthesis and whole body fat oxidation rates. Am J Physiol Endocrinol Metab. (2010) 298:E108–16. 10.1152/ajpendo.00524.2009
    1. Okita N, Tsuchiya T, Fukushima M, Itakura K, Yuguchi K, Narita T, et al. Chronological analysis of caloric restriction-induced alteration of fatty acid biosynthesis in white adipose tissue of rats. Exp Gerontol. (2015) 63:59–66. 10.1016/j.exger.2015.01.043
    1. Ford JH. Saturated fatty acid metabolism is key link between cell division, cancer, and senescence in cellular and whole organism aging. Age. (2010) 32:231–7. 10.1007/s11357-009-9128-x
    1. Almario CV, Ballal ML, Chey WD, Nordstrom C, Khanna D, Spiegel BMR. Burden of gastrointestinal symptoms in the United States: results of a nationally representative survey of over 71,000 Americans. Am J Gastroenterol. (2018) 113:1701–10. 10.1038/s41395-018-0256-8
    1. Aziz I, Palsson OS, Törnblom H, Sperber AD, Whitehead WE, Simrén M. The prevalence and impact of overlapping Rome IV-diagnosed functional gastrointestinal disorders on somatization, quality of life, and healthcare utilization: a cross-sectional general population study in three countries. Am J Gastroenterol. (2017) 113:86–96. 10.1038/ajg.2017.421
    1. Stocks NP, Gonzalez-Chica D, Hay P. Impact of gastrointestinal conditions, restrictive diets and mental health on health-related quality of life: cross-sectional population-based study in Australia. Bmj Open. (2019) 9:e026035. 10.1136/bmjopen-2018-026035
    1. Ward FW, Coates ME. Gastrointestinal pH measurement in rats: influence of the microbial flora, diet and fasting. Lab Anim. (1987) 21:216–22. 10.1258/002367787781268693
    1. Sowah SA, Riedl L, Damms-Machado A, Johnson TS, Schübel R, Graf M, et al. Effects of weight-loss interventions on short-chain fatty acid concentrations in blood and feces of adults: a systematic review. Adv Nutr. (2019) 10:673–84. 10.1093/advances/nmy125
    1. Kohmoto T, Tsuji K, Kaneko T, Shiota M, Fukui F, Takaku H, et al. Metabolism of 13C-isomaltooligosaccharides in healthy men. Biosci Biotechnol Biochem. (1992) 56:937–40. 10.1271/bbb.56.937
    1. Høverstad T, Fausa O, Bjørneklett A, Bøhmer T. Short-chain fatty acids in the normal human feces. Scand J Gastroenterol. (2018) 19:375–81. 10.1080/00365521.1984.12005738
    1. Cummings JH, Pomare EW, Branch WJ, Naylor CP, Macfarlane GT. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut. (1987) 28:1221. 10.1136/gut.28.10.1221
    1. Costea PI, Hildebrand F, Arumugam M, Bäckhed F, Blaser MJ, Bushman FD, et al. Enterotypes in the landscape of gut microbial community composition. Nat Microbiol. (2018) 3:8–16. 10.1038/s41564-017-0072-8
    1. Wilmanski T, Rappaport N, Earls JC, Magis AT, Manor O, Lovejoy J, et al. Blood metabolome predicts gut microbiome α-diversity in humans. Nat Biotechnol. (2019) 37:1217–28. 10.1038/s41587-019-0233-9

Source: PubMed

3
Suscribir