Calorie restriction improves metabolic state independently of gut microbiome composition: a randomized dietary intervention trial

Solomon A Sowah, Alessio Milanese, Ruth Schübel, Jakob Wirbel, Ece Kartal, Theron S Johnson, Frank Hirche, Mirja Grafetstätter, Tobias Nonnenmacher, Romy Kirsten, Marina López-Nogueroles, Agustín Lahoz, Kathrin V Schwarz, Jürgen G Okun, Cornelia M Ulrich, Johanna Nattenmüller, Arnold von Eckardstein, Daniel Müller, Gabriele I Stangl, Rudolf Kaaks, Tilman Kühn, Georg Zeller, Solomon A Sowah, Alessio Milanese, Ruth Schübel, Jakob Wirbel, Ece Kartal, Theron S Johnson, Frank Hirche, Mirja Grafetstätter, Tobias Nonnenmacher, Romy Kirsten, Marina López-Nogueroles, Agustín Lahoz, Kathrin V Schwarz, Jürgen G Okun, Cornelia M Ulrich, Johanna Nattenmüller, Arnold von Eckardstein, Daniel Müller, Gabriele I Stangl, Rudolf Kaaks, Tilman Kühn, Georg Zeller

Abstract

Background: The gut microbiota has been suggested to play a significant role in the development of overweight and obesity. However, the effects of calorie restriction on gut microbiota of overweight and obese adults, especially over longer durations, are largely unexplored.

Methods: Here, we longitudinally analyzed the effects of intermittent calorie restriction (ICR) operationalized as the 5:2 diet versus continuous calorie restriction (CCR) on fecal microbiota of 147 overweight or obese adults in a 50-week parallel-arm randomized controlled trial, the HELENA Trial. The primary outcome of the trial was the differential effects of ICR versus CCR on gene expression in subcutaneous adipose tissue. Changes in the gut microbiome, which are the focus of this publication, were defined as exploratory endpoint of the trial. The trial comprised a 12-week intervention period, a 12-week maintenance period, and a final follow-up period of 26 weeks.

Results: Both diets resulted in ~5% weight loss. However, except for Lactobacillales being enriched after ICR, post-intervention microbiome composition did not significantly differ between groups. Overall weight loss was associated with significant metabolic improvements, but not with changes in the gut microbiome. Nonetheless, the abundance of the Dorea genus at baseline was moderately predictive of subsequent weight loss (AUROC of 0.74 for distinguishing the highest versus lowest weight loss quartiles). Despite the lack of consistent intervention effects on microbiome composition, significant study group-independent co-variation between gut bacterial families and metabolic biomarkers, anthropometric measures, and dietary composition was detectable. Our analysis in particular revealed associations between insulin sensitivity (HOMA-IR) and Akkermansiaceae, Christensenellaceae, and Tanerellaceae. It also suggests the possibility of a beneficial modulation of the latter two intestinal taxa by a diet high in vegetables and fiber, and low in processed meat.

Conclusions: Overall, our results suggest that the gut microbiome remains stable and highly individual-specific under dietary calorie restriction.

Trial registration: The trial, including the present microbiome component, was prospectively registered at ClinicalTrials.gov NCT02449148 on May 20, 2015.

Keywords: Gut microbiome; Intermittent calorie restriction; Obesity; Overweight; Weight loss.

Conflict of interest statement

The authors declare that they have no competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Modified CONSORT diagram showing the flow of participants in the HELENA Trial. ICR – intermittent calorie restriction; CCR – continuous calorie restriction; CTR – control group; CONSORT – consolidated standards of reporting trials
Fig. 2
Fig. 2
Overview of the study and intervention effects on the gut microbiome composition. a Graphical overview of the HELENA Trial design showing details of all data collected over the course of the trial. In total, 150 overweight/obese adults were randomly assigned to the ICR (5 days eucaloric diet, 2 days of ~25% energy requirement), CCR (daily reduction of ~20% of caloric intake) or CTR group, 147 of them provided stool samples at baseline. Anthropometric measurements were performed for all participants. Blood samples were collected at all study timepoints for the assessment of plasma/serum concentrations of routine biomarkers. Dietary information was collected at baseline, week 12, and week 50. The amount of weight loss was significantly higher among ICR and CCR participants compared to CTR participants at all study timepoints, as previously published by Schübel et al. [22]. Differences in weight change across intervention groups at each timepoint was assessed using linear mixed models adjusted for age and sex, with participant identifiers as random effects in the model. A significant difference, i.e., p < 0.05 is indicated by (*). ICR – intermittent calorie restriction; CCR – continuous calorie restriction; CTR – controls. b The gut microbiota was predominantly composed of bacteria belonging to Clostridia followed by Bacteroidia as shown by stacked bar charts of taxonomic classes across timepoints and subdivided by intervention group. c Non-metric multidimensional scaling (NMDS) plots of Bray-Curtis dissimilarity distances based on ASV relative abundances between samples after 12 weeks of the intervention. Each point represents the microbial community of a sample, and the colour indicates the intervention group to which it belongs. NMDS plots do not show clustering by intervention groups and PERMANOVA does not indicate a statistically significant difference between groups. d Intra-individual variability (same subject compared across time points) was significantly lower compared with inter-individual variability (different subjects, same group and different subjects, different groups). Technical replicates in this study showed a very low variation in Bray-Curtis dissimilarity distances based on ASV relative abundances, attesting to a high technical reproducibility. Statistical significance as indicated above boxplots was assessed by Wilcoxon signed-rank test. e There was no significant intervention effect on overall gut microbiota alpha diversity (Shannon Index) across all intervention timepoints. All boxplots show the interquartile ranges (IQRs) as boxes, with the median as a black horizontal line and the whiskers extending up to the most extreme points within 1.5-fold IQR. Additional file 3: Source data 2
Fig. 3
Fig. 3
Changes in Lactobacillales abundance and processed meat intake, and associations between changes in Lactobacillales abundance and processed meat intake. a The relative abundance of the order Lactobacillales increased in the ICR group after the 12-week intervention but returned to levels similar to baseline at the end of follow-up as shown by boxplots (defined as in Fig. 3); statistical significance was assessed by Wilcoxon signed-rank test. b Participants in the ICR group significantly reduced their intake of processed meat during the 12-week intervention period, although intakes increased again at follow-up. In contrast, there was no significant change in the consumption of processed meat in either the CCR or CTR groups as shown by boxplots. c The changes in the abundance of Lactobacillales after 12 weeks were significantly associated with the changes in processed meat intake (Spearman correlation, q = 0.02). Scatter plot showing Spearman correlation between changes in the relative abundance of Lactobacillales and changes in processed meat intake. d The genus Streptococcus, which belongs to the order Lactobacillales, also significantly co-varied with the changes in processed meat intake after the 12-week intervention (q = 0.02). e Abundance changes in Lactobacillales and its member genus Streptococcus were significantly associated with changes in processed meat intake after 12 weeks. Bar length indicates q value (FDR-corrected p-value) and effect size is color-coded. Additional file 4: Source data
Fig. 4
Fig. 4
Association of weight loss with anthropometric, clinical markers, metabolic biomarkers and the gut microbiome. a Weight loss was significantly associated with decreases in body fat compartments (e.g., VAT and SAT), and routine biomarkers (e.g., cholesterol and LDL), but not with changes in gut microbial and small-molecule metabolites, with the exception of choline. Only variables significantly associated with weight loss are labelled (red dots). Measured metabolites (unlabelled due to a lack of association with weight loss) aside routine biomarkers such as those of glucose metabolism, e.g., insulin, comprised bile acids, SCFAs, TMAO, and its precursors, amino acids, and acylcarnitines, but also inflammatory cytokines (Additional file 5: Source data 4). Variables shown by dark red spots were significantly associated with weight loss also at follow-up, i.e., week 50. b The changes in weight after 12 weeks were not significantly associated with any individual bacterial taxon across all taxonomic levels, after FDR correction (all taxonomic levels included in the plot) c There was no differential association of weight loss with gut microbial alpha diversity across all timepoints. Boxplots show changes in alpha diversity over time according to quartiles of weight loss (Q1—quartile 1, Q2—quartile 2, Q3—quartile 3, and Q4—quartile 4, see Fig. 2 for definition of boxplots). Noticeably, metabolic improvements based on the changes in anthropometric and body composition parameters as well as blood concentrations of some biomarkers were greater among participants in Q4 who achieved the highest amount of weight loss after 12 weeks (See Additional file 2: Table S8). As also indicated by the volcano plot in a, there was a significant linear association between weight loss and changes in d visceral adipose tissue, e liver fat, f leptin, g gamma-glutamyl transferase (GGT), h choline, and i cholesterol after the 12-week intervention period. j Baseline abundances, rather than intervention group or weight loss accounted for the largest variation in post-intervention relative abundance of bacteria (here shown at phylum level). Bar plot showing the variations in phyla relative abundance (see colour key) after 12 weeks of intervention accounted for by age at recruitment, sex, baseline phyla abundance, and weight loss (%). Models were generated separately for weight loss and intervention, while partial R-squares of age, sex, and baseline abundances are from the model including weight loss. Additional file 5: Source data 4
Fig. 5
Fig. 5
Association of baseline gut microbiota composition with weight loss. A higher abundance of the genus Dorea at baseline may be associated with a difficulty in losing excess weight. a Volcano plot showing the difference in the relative abundance of Dorea between participants in the highest quartile of weight loss and those in the lowest at baseline. Baseline Dorea abundances were significantly higher among participants in the lowest quartile of weight loss. b Baseline Dorea abundances rather than Prevotella-to-Bacteroides ratio or gut microbiota richness was predictive of post-intervention weight loss. AUROC showing how the aforementioned parameters were predictive of weight loss. c The relative abundance of Dorea at baseline was positively associated with weight loss, i.e., a higher abundance corresponds to a lower amount of weight loss. A scatter plot showing the association between the relative abundance of Dorea at baseline and weight loss after 12 weeks. Additional file 6: Source data 5
Fig. 6
Fig. 6
Association of the microbiome with anthropometric measurements, metabolic biomarkers, and dietary intake across timepoints. Several core families within the gut microbiome significantly co-vary with some of the clinical biomarkers, body composition measures, and dietary intake assessed in the trial. a Heatmap showing the strength of association calculated irrespective of weight loss quartile or outcome. Phylum affiliation for each family is indicated as a colour strip on the left. Significance for each association after FDR correction is indicated by asterisks (q < 0.1 (*), q < 0.05 (**), and q < 0.01 (***)). Asterisks colored yellow were no longer significant after adjustment for weight loss in the LME models. b Many of the associations shown in a were consistent, even after adjusting for weight loss in the LME models. The yellow points represent the associations for which significance was lost after adjustment for weight loss. c–i Scatterplots for selected significant associations based on the LME models in a. Log-transformed relative abundances of gut bacterial families were corrected for participant-specific offsets using the regression intercepts. The regression slopes for each weight loss quartile has been shown for associations that were significantly influenced by the degree of weight loss (refer to legend) (raw LME model plots without individual-specific abundance correction are shown in Figure S7). j Correlation network between bacterial families and anthropometric measurements, clinical markers and food and energy intake based on the data from a. The network includes bacterial families with at least one significant association to any of the aforementioned parameters and other families that show significant correlation across LME model coefficients (rows of the heatmap in a) with any of those families (Spearman correlation q < 0.05, edge thickness proportional to rho). Edges between families and the parameters are included if the absolute effect size (estimated by the LME) exceeded 0.075 (significant associations are indicated by stronger edges). Additional file 7: Source data 6.

References

    1. Calle EE, Rodriguez C, Walker-Thurmond K, Thun MJ. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med. 2003;348(17):1625–1638.
    1. Pi-Sunyer FX. The obesity epidemic: pathophysiology and consequences of obesity. Obes Res. 2002;10 Suppl 2:97s–104s.
    1. Smith SR, Lovejoy JC, Greenway F, Ryan D, de Jonge L, de la Bretonne J, et al. Contributions of total body fat, abdominal subcutaneous adipose tissue compartments, and visceral adipose tissue to the metabolic complications of obesity. Metabolism. 2001;50(4):425–435.
    1. Shoelson SE, Herrero L, Naaz A. Obesity, inflammation, and insulin resistance. Gastroenterology. 2007;132(6):2169–2180.
    1. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444(7122):1027–1031.
    1. Chambers ES, Preston T, Frost G, Morrison DJ. Role of gut microbiota-generated short-chain fatty acids in metabolic and cardiovascular health. Curr Nutr Rep. 2018;7(4):198–206.
    1. Yoshimoto S, Loo TM, Atarashi K, Kanda H, Sato S, Oyadomari S, et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature. 2013;499(7456):97–101.
    1. Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472(7341):57–63.
    1. Liu R, Hong J, Xu X, Feng Q, Zhang D, Gu Y, et al. Gut microbiome and serum metabolome alterations in obesity and after weight-loss intervention. Nat Med. 2017;23(7):859–868.
    1. Cotillard A, Kennedy SP, Kong LC, Prifti E, Pons N, Le Chatelier E, et al. Dietary intervention impact on gut microbial gene richness. Nature. 2013;500(7464):585–588.
    1. Frost F, Storck LJ, Kacprowski T, Gärtner S, Rühlemann M, Bang C, et al. A structured weight loss program increases gut microbiota phylogenetic diversity and reduces levels of Collinsella in obese type 2 diabetics: A pilot study. Plos One. 2019;14(7):e0219489.
    1. Heinsen FA, Fangmann D, Müller N, Schulte DM, Rühlemann MC, Türk K, et al. Beneficial effects of a dietary weight loss intervention on human gut microbiome diversity and metabolism are not sustained during weight maintenance. Obes Facts. 2016;9(6):379–391.
    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(8):832–851.
    1. Leeming ER, Johnson AJ, Spector TD, Le Roy CI. Effect of diet on the gut microbiota: rethinking intervention duration. Nutrients. 2019;11(12):2862.
    1. Gerber GK. The dynamic microbiome. FEBS Lett. 2014;588(22):4131–4139.
    1. David LA, Materna AC, Friedman J, Campos-Baptista MI, Blackburn MC, Perrotta A, et al. Host lifestyle affects human microbiota on daily timescales. Genome Biolo. 2014;15(7):R89.
    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(6):1222–35.e6.
    1. Liu Z, Dai X, Zhang H, Shi R, Hui Y, Jin X, et al. Gut microbiota mediates intermittent-fasting alleviation of diabetes-induced cognitive impairment. Nat Commun. 2020;11(1):855.
    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(9):1867–1879.
    1. Li G, Xie C, Lu S, Nichols RG, Tian Y, Li L, et al. Intermittent fasting promotes white adipose browning and decreases obesity by shaping the gut microbiota. Cell Metab. 2017;26(5):801.
    1. Muñoz-Hernández L, Márquez-López Z, Mehta R, Aguilar-Salinas CA. Intermittent fasting as part of the management for T2DM: from animal models to human clinical studies. Curr Diab Rep. 2020;20(4):13.
    1. Schübel R, Nattenmuller J, Sookthai D, Nonnenmacher T, Graf ME, Riedl L, et al. Effects of intermittent and continuous calorie restriction on body weight and metabolism over 50 wk: a randomized controlled trial. Am J Clin Nutr. 2018;108(5):933–945.
    1. Schubel R, Graf ME, Nattenmuller J, Nabers D, Sookthai D, Gruner LF, et al. The effects of intermittent calorie restriction on metabolic health: rationale and study design of the HELENA Trial. Contemp Clin Trials. 2016;51:28–33.
    1. Oberritter H, Schäbethal K, von Ruesten A, Boeing H. The DGE Nutrition Circle – presentation and basis of the food-related recommendations from the German Nutrition Society (DGE) Ernährungs Umschau. 2013;2:24–29.
    1. World Medical Association World Medical Association Declaration of Helsinki: ethical principles for medical research involving human subjects. JAMA. 2013;310(20):2191–2194.
    1. Chan JC, Kioh DY, Yap GC, Lee BW, Chan EC. A novel LCMSMS method for quantitative measurement of short-chain fatty acids in human stool derivatized with (12) C- and (13) C-labelled aniline. J Pharm Biomed Anal. 2017;138:43–53.
    1. Zeng M, Cao H. Fast quantification of short chain fatty acids and ketone bodies by liquid chromatography-tandem mass spectrometry after facile derivatization coupled with liquid-liquid extraction. J Chromatog B Analyt Technol Biomed Life Sci. 2018;1083:137–145.
    1. Sowah SA, Hirche F, Milanese A, Johnson TS, Grafetstatter M, Schubel R, et al. Changes in plasma short-chain fatty acid levels after dietary weight loss among overweight and obese adults over 50 weeks. Nutrients. 2020;12(2):452.
    1. Lindner M, Abdoh G, Fang-Hoffmann J, Shabeck N, Al-Sayrafi M, Al-Janahi M, et al. Implementation of extended neonatal screening and a metabolic unit in the State of Qatar: developing and optimizing strategies in cooperation with the Neonatal Screening Center in Heidelberg. J Inherit Metab Dis. 2007;30(4):522–529.
    1. Garcia-Canaveras JC, Donato MT, Castell JV, Lahoz A. Targeted profiling of circulating and hepatic bile acids in human, mouse, and rat using a UPLC-MRM-MS-validated method. J Lipid Res. 2012;53(10):2231–2241.
    1. Mueller DM, Allenspach M, Othman A, Saely CH, Muendlein A, Vonbank A, et al. Plasma levels of trimethylamine-N-oxide are confounded by impaired kidney function and poor metabolic control. Atherosclerosis. 2015;243(2):638–644.
    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 USA. 2011;108 Suppl 1(Suppl 1):4516–4522.
    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(8):1621–1624.
    1. Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJ, Holmes SP. DADA2: High-resolution sample inference from Illumina amplicon data. Nat Methods. 2016;13(7):581–583.
    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. 2013;41(Database issue):D590–D596.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Series B (Methodological) 1995;57(1):289–300.
    1. Jari O, Guillaume B, Friendly M, Kindt R, Legendre P, McGlinn D, et al. Vegan: Community Ecology Package. R package version 2.5-6. 2019.
    1. Kunzetsova A, Brockhoff P, Christensen R. lmerTest package: tests in linear mixed effect models. J Stat Softw. 2017;82:1–26.
    1. Human Microbiome Project Consortium Structure, function and diversity of the healthy human microbiome. Nature. 2012;486(7402):207–214.
    1. Grembi JA, Nguyen LH, Haggerty TD, Gardner CD, Holmes SP, Parsonnet J. Gut microbiota plasticity is correlated with sustained weight loss on a low-carb or low-fat dietary intervention. Sci Rep. 2020;10(1):1405.
    1. R Core Team . R: a language and environment for statistical computing. Vienna: R Foundation for Statistical Computing; 2019.
    1. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, et al. Diversity of the human intestinal microbial flora. Science. 2005;308(5728):1635–1638.
    1. Caporaso JG, Lauber CL, Costello EK, Berg-Lyons D, Gonzalez A, Stombaugh J, et al. Moving pictures of the human microbiome. Genome Biol. 2011;12(5):R50.
    1. Hjorth MF, Christensen L, Larsen TM, Roager HM, Krych L, Kot W, et al. Pretreatment Prevotella-to-Bacteroides ratio and salivary amylase gene copy number as prognostic markers for dietary weight loss. Am J Clin Nutr. 2020;111(5):1079–1086.
    1. Hjorth MF, Blaedel T, Bendtsen LQ, Lorenzen JK, Holm JB, Kiilerich P, et al. Prevotella-to-Bacteroides ratio predicts body weight and fat loss success on 24-week diets varying in macronutrient composition and dietary fiber: results from a post-hoc analysis. Int J Obes. 2019;43(1):149-57.
    1. Biesiekierski JR, Jalanka J, Staudacher HM. Can gut microbiota composition predict response to dietary treatments? Nutrients. 2019;11(5):1134.
    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. 2014;505(7484):559–563.
    1. Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334(6052):105–108.
    1. Walker AW, Ince J, Duncan SH, Webster LM, Holtrop G, Ze X, et al. Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J. 2011;5(2):220–230.
    1. Chittim CL, Martínez Del Campo A, Balskus EP. Gut bacterial phospholipase Ds support disease-associated metabolism by generating choline. Nat Microbiol. 2019;4(1):155–163.
    1. Heianza Y, Sun D, Li X, DiDonato JA, Bray GA, Sacks FM, et al. Gut microbiota metabolites, amino acid metabolites and improvements in insulin sensitivity and glucose metabolism: the POUNDS Lost trial. Gut. 2019;68(2):263–270.
    1. Lent-Schochet D, McLaughlin M, Ramakrishnan N, Jialal I. Exploratory metabolomics of metabolic syndrome: a status report. World J Diabetes. 2019;10(1):23–36.
    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(1):8–16.
    1. Roager HM, Vogt JK, Kristensen M, Hansen LBS, Ibrugger S, Maerkedahl RB, et al. Whole grain-rich diet reduces body weight and systemic low-grade inflammation without inducing major changes of the gut microbiome: a randomised cross-over trial. Gut. 2019;68(1):83-93.
    1. Fragiadakis GK, Wastyk HC, Robinson JL, Sonnenburg ED, Sonnenburg JL, Gardner CD. Long-term dietary intervention reveals resilience of the gut microbiota despite changes in diet and weight. Am J Clin Nutr. 2020;111(6):1127–1136.
    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(6):e90702.
    1. Ley RE, Backhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI. Obesity alters gut microbial ecology. Proc Natl Acad Sci USA. 2005;102(31):11070–11075.
    1. von Schwartzenberg RJ, Bisanz JE, Lyalina S, Spanogiannopoulos P, Ang QY, Cai J, et al. Caloric restriction disrupts the microbiota and colonization resistance. Nature. 2021;595(7866):272–277.
    1. Kuipers F, Bloks VW, Groen AK. Beyond intestinal soap--bile acids in metabolic control. Nat Rev Endocrinol. 2014;10(8):488–498.
    1. Nadal I, Santacruz A, Marcos A, Warnberg J, Garagorri JM, Moreno LA, et al. Shifts in clostridia, bacteroides and immunoglobulin-coating fecal bacteria associated with weight loss in obese adolescents. Int J Obes (2005). 2009;33(7):758–767.
    1. Damms-Machado A, Mitra S, Schollenberger AE, Kramer KM, Meile T, Konigsrainer A, et al. Effects of surgical and dietary weight loss therapy for obesity on gut microbiota composition and nutrient absorption. BioMed Res Int. 2015;2015:806248.
    1. Duncan SH, Belenguer A, Holtrop G, Johnstone AM, Flint HJ, Lobley GE. Reduced dietary intake of carbohydrates by obese subjects results in decreased concentrations of butyrate and butyrate-producing bacteria in feces. Appl Environ Microbiol. 2007;73(4):1073–1078.
    1. Daas MC, de Roos NM. Intermittent fasting contributes to aligned circadian rhythms through interactions with the gut microbiome. Benef Microbes. 2021;12(2):147-161.
    1. Remely M, Tesar I, Hippe B, Gnauer S, Rust P, Haslberger AG. Gut microbiota composition correlates with changes in body fat content due to weight loss. Benef Microbes. 2015;6(4):431–439.
    1. Fouladi F, Brooks AE, Fodor AA, Carroll IM, Bulik-Sullivan EC, Tsilimigras MCB, et al. The role of the gut microbiota in sustained weight loss following Roux-en-Y gastric bypass surgery. Obes Surg. 2019;29(4):1259–1267.
    1. Schutkowski A, König B, Kluge H, Hirche F, Henze A, Schwerdtle T, et al. Metabolic footprint and intestinal microbial changes in response to dietary proteins in a pig model. J Nutr Biochem. 2019;67:149–160.
    1. Zhu Y, Lin X, Zhao F, Shi X, Li H, Li Y, et al. Meat, dairy and plant proteins alter bacterial composition of rat gut bacteria. Sci Rep. 2015;5(1):15220.
    1. Zhu Y, Lin X, Li H, Li Y, Shi X, Zhao F, et al. Intake of meat proteins substantially increased the relative abundance of Genus Lactobacillus in rat feces. Plos one. 2016;11(4):e0152678.
    1. Allaf M, Elghazaly H, Mohamed OG, Fareen MFK, Zaman S, Salmasi AM, et al. Intermittent fasting for the prevention of cardiovascular disease. Cochrane Database Syst Rev. 2021;1:Cd013496.
    1. Schwingshackl L, Zähringer J, Nitschke K, Torbahn G, Lohner S, Kühn T, et al. Impact of intermittent energy restriction on anthropometric outcomes and intermediate disease markers in patients with overweight and obesity: systematic review and meta-analyses. Crit Rev Food Sci Nutr. 2021;61(8):1293–1304.
    1. Vitale R, Kim Y. The effects of intermittent fasting on glycemic control and body composition in adults with obesity and type 2 diabetes: a systematic review. Metab Syndr RelatDisord. 2020;18(10):450–461.
    1. Schneeberger M, Everard A, Gómez-Valadés AG, Matamoros S, Ramírez S, Delzenne NM, et al. Akkermansia muciniphila inversely correlates with the onset of inflammation, altered adipose tissue metabolism and metabolic disorders during obesity in mice. Sci Rep. 2015;5:16643.
    1. Zhang X, Shen D, Fang Z, Jie Z, Qiu X, Zhang C, et al. Human gut microbiota changes reveal the progression of glucose intolerance. Plos One. 2013;8(8):e71108.
    1. Depommier C, Everard A, Druart C, Plovier H, Van Hul M, Vieira-Silva S, et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study. nat Med. 2019;25(7):1096–1103.
    1. Waters JL, Ley RE. The human gut bacteria Christensenellaceae are widespread, heritable, and associated with health. BMC Biol. 2019;17(1):83.
    1. Tavella T, Rampelli S, Guidarelli G, Bazzocchi A, Gasperini C, Pujos-Guillot E, et al. Elevated gut microbiome abundance of Christensenellaceae, Porphyromonadaceae and Rikenellaceae is associated with reduced visceral adipose tissue and healthier metabolic profile in Italian elderly. Gut Microbes. 2021;13(1):1–19.
    1. Goodrich JK, Waters JL, Poole AC, Sutter JL, Koren O, Blekhman R, et al. Human genetics shape the gut microbiome. Cell. 2014;159(4):789–799.
    1. Jiang W, Wu N, Wang X, Chi Y, Zhang Y, Qiu X, et al. Dysbiosis gut microbiota associated with inflammation and impaired mucosal immune function in intestine of humans with non-alcoholic fatty liver disease. Sci Rep. 2015;5:8096.
    1. Caussy C, Tripathi A, Humphrey G, Bassirian S, Singh S, Faulkner C, et al. A gut microbiome signature for cirrhosis due to nonalcoholic fatty liver disease. Nat Commun. 2019;10(1):1406.
    1. Anhê FF, Jensen BAH, Varin TV, Servant F, Van Blerk S, Richard D, et al. Type 2 diabetes influences bacterial tissue compartmentalisation in human obesity. Nat Metab. 2020;2(3):233–242.
    1. Wang J-H, Shin NR, Lim S-K, Im U, Song E-J, Nam Y-D, et al. Diet control more intensively disturbs gut microbiota than genetic background in wild type and ob/ob mice. Front Microbiol. 2019;10:1292.
    1. Gomez-Arango LF, Barrett HL, Wilkinson SA, Callaway LK, McIntyre HD, Morrison M, et al. Low dietary fiber intake increases Collinsella abundance in the gut microbiota of overweight and obese pregnant women. Gut Microbes. 2018;9(3):189–201.
    1. Ghosh TS, Rampelli S, Jeffery IB, Santoro A, Neto M, Capri M, et al. Mediterranean diet intervention alters the gut microbiome in older people reducing frailty and improving health status: the NU-AGE 1-year dietary intervention across five European countries. Gut. 2020;69(7):1218–1228.
    1. Sowah SA, Milanese A, Schübel R, Wirbel J, Kartal E, Johnson TS, et al. Dataset - Calorie restriction improves metabolic state independently of gut microbiome composition Figshare. 2020.
    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: 16S rRNA amplicon sequencing data; ENA Accession Number PRJEB40697, European Nucleotide Archive (ENA) database. 2020.

Source: PubMed

3
Sottoscrivi