The effects of daily fasting hours on shaping gut microbiota in mice

Linghao Li, Yuxin Su, Fanglin Li, Yueying Wang, Zhongren Ma, Zhuo Li, Junhong Su, Linghao Li, Yuxin Su, Fanglin Li, Yueying Wang, Zhongren Ma, Zhuo Li, Junhong Su

Abstract

Background: It has recently been reported that intermittent fasting shapes the gut microbiota to benefit health, but this effect may be influenced to the exact fasting protocols. The purpose of this study was to assess the effects of different daily fasting hours on shaping the gut microbiota in mice. Healthy C57BL/6 J male mice were subjected to 12, 16 or 20 h fasting per day for 1 month, and then fed ad libitum for an extended month. Gut microbiota was analyzed by 16S rRNA gene-based sequencing and food intake was recorded as well.

Results: We found that cumulative food intake was not changed in the group with 12 h daily fasting, but significantly decreased in the 16 and 20 h fasting groups. The composition of gut microbiota was altered by all these types of intermittent fasting. At genus level, 16 h fasting led to increased level of Akkermansia and decreased level of Alistipes, but these effects disappeared after the cessation of fasting. No taxonomic differences were identified in the other two groups.

Conclusions: These data indicated that intermittent fasting shapes gut microbiota in healthy mice, and the length of daily fasting interval may influence the outcome of intermittent fasting.

Keywords: Daily fasting hours; Food intake; Gut microbiota; Mouse model.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Food intake during intermittent fasting. a Experimental design; b The amount of total food intake during fasting; c The amount of food intake per day at each time point during the two-month study period; d The amount of total food intake 1 month after the cessation of intermittent fasting. *p < 0.05, **p < 0.01, ***p < 0.001 by one-way ANOVA, followed by Tukey’s post hoc for multiple comparisons. e Pearson correlation between cumulative food intake and daily fasting hours during fasting. f Pearson correlation between cumulative food intake and daily fasting hours 1 month after the cessation of fasting
Fig. 2
Fig. 2
Changes in body weight during intermittent fasting. a Body weight at the end of daily fasting; b Body weight before the start of daily fasting; *p < 0.05, **p < 0.01, ***p < 0.001 by one-way ANOVA, followed by Tukey’s post hoc for multiple comparisons
Fig. 3
Fig. 3
Analysis of gut bacterial communities by 16S rRNA analysis from fed and fasted mice. a Principal Co-ordinates Analysis (PCoA) on Bray-Curtis dissimilarities of bacterial communities from four different fasting regimens at two time points. Each point corresponds to a community from a single mouse. Colors indicate community identity. Ellipses show the 95% confidence intervals. Coloured arrows indicate community shift from day 30 to day 60. Intra-group differences were indicated by using ANOSIM test. ***p ≤ 0.001. b The Figure shows the percentage of each community contributed by the indicated phyla. Time point and daily fasting durations are indicated below the Figure. Taxa that discriminated between fasted and control mice during fasting (c) or 1 month after the cessation of fasting (d). Taxa with a log LDA (linear discriminant analysis) score above 4.00 as determined by using LEfSe. Data shown are the log10 linear discriminant analysis (LDA) scores following LEfSe analyses and the hierarch of discriminating taxa visualized as cladograms for taxonomic comparisons between fasted and control mice

References

    1. Hooper LV, Macpherson AJ. Immune adaptations that maintain homeostasis with the intestinal microbiota. Nat Rev Immunol. 2010;10(3):159.
    1. Lynch SV, Pedersen O. The human intestinal microbiome in health and disease. N Engl J Med. 2016;375(24):2369–2379.
    1. Clemente JC, Ursell LK, Parfrey LW, Knight R. The impact of the gut microbiota on human health: an integrative view. Cell. 2012;148(6):1258–1270.
    1. Mattson MP, Wan R. Beneficial effects of intermittent fasting and caloric restriction on the cardiovascular and cerebrovascular systems. J Nutr Biochem. 2005;16(3):129–137.
    1. Halagappa VKM, Guo Z, Pearson M, Matsuoka Y, Cutler RG, LaFerla FM, et al. Intermittent fasting and caloric restriction ameliorate age-related behavioral deficits in the triple-transgenic mouse model of Alzheimer’s disease. Neurobiol Dis. 2007;26(1):212–220.
    1. Mattson MP, Longo VD, Harvie M. Impact of intermittent fasting on health and disease processes. Ageing Res Rev. 2017;39:46–58.
    1. Salti I, Bénard E, Detournay B, Bianchi-Biscay M, Le Brigand C, Voinet C, et al. A population-based study of diabetes and its characteristics during the fasting month of Ramadan in 13 countries: results of the epidemiology of diabetes and Ramadan 1422/2001 (EPIDIAR) study. Diabetes Care. 2004;27(10):2306–2311.
    1. Ibrahim IA, Hassan EA, Alkhan AM, Hussein MA, Alhabashi AF, Ali TZ, et al. Ramadan fasting in kidney transplant recipients: a single-centre retrospective study. J Transplant. 2018;2018:4890978.
    1. Lessan N, Saadane I, Alkaf B, Hambly C, Buckley AJ, Finer N, et al. The effects of Ramadan fasting on activity and energy expenditure. Am J Clin Nutr. 2018;107(1):54–61.
    1. Hassanein M, Al-Arouj M, Hamdy O, Bebakar WMW, Jabbar A, Al-Madani A, et al. Diabetes and Ramadan: practical guidelines. Diabetes Res Clin Pract. 2017;126:303–316.
    1. Gorkiewicz G, Moschen A. Gut microbiome: a new player in gastrointestinal disease. Virchows Arch. 2018;472(1):159–172.
    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):1222–1235.e1226.
    1. Janssen S, Depoortere I. Nutrient sensing in the gut: new roads to therapeutics? Trends Endocrinol Metab. 2013;24(2):92–100.
    1. Mitchell SJ, Bernier M, Mattison JA, Aon MA, Kaiser TA, Anson RM, et al. Daily fasting improves health and survival in male mice independent of diet composition and calories. Cell Metab. 2019;29(1):221–228.
    1. Le Roy T, Llopis M, Lepage P, Bruneau A, Rabot S, Bevilacqua C, et al. Intestinal microbiota determines development of non-alcoholic fatty liver disease in mice. Gut. 2013;62(12):1787–1794.
    1. Hildebrandt MA, Hoffmann C, Sherrill-Mix SA, Keilbaugh SA, Hamady M, Chen YY, et al. High-fat diet determines the composition of the murine gut microbiome independently of obesity. Gastroenterology. 2009;137(5):1716–1724.e1711-1712.
    1. Longo VD, Mattson MP. Fasting: molecular mechanisms and clinical applications. Cell Metab. 2014;19(2):181–192.
    1. Malinowski B, Zalewska K, Wesierska A, Sokolowska MM, Socha M, Liczner G, et al. Intermittent fasting in cardiovascular disorders-an overview. Nutrients. 2019;11(3):673.
    1. Emberson JR, Whincup PH, Walker M, Thomas M, Alberti KG. Biochemical measures in a population-based study: effect of fasting duration and time of day. Ann Clin Biochem. 2002;39(Pt 5):493–501.
    1. Lee KH, Ko BH, Paik JY, Jung KH, Choe YS, Choi Y, et al. Effects of anesthetic agents and fasting duration on 18F-FDG biodistribution and insulin levels in tumor-bearing mice. J Nucl Med. 2005;46(9):1531–1536.
    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(4):672–685.e674.
    1. Kang Y, Cai Y, Zhang X, Kong X, Su J. Altered gut microbiota in RA: implications for treatment. Z Rheumatol. 2017;76(5):451–457.
    1. Carding S, Verbeke K, Vipond DT, Corfe BM, Owen LJ. Dysbiosis of the gut microbiota in disease. Microb Ecol Health Dis. 2015;26:26191.
    1. Martin B, Mattson MP, Maudsley S. Caloric restriction and intermittent fasting: two potential diets for successful brain aging. Ageing Res Rev. 2006;5(3):332–353.
    1. Patterson RE, Sears DD. Metabolic effects of intermittent fasting. Annu Rev Nutr. 2017;37:371–393.
    1. Wilkinson MJ, Manoogian ENC, Zadourian A, Lo H, Fakhouri S, Shoghi A, et al. Ten-hour time-restricted eating reduces weight, blood pressure, and Atherogenic lipids in patients with metabolic syndrome. Cell Metab. 2020;31(1):92–104.
    1. Stekovic S, Hofer SJ, Tripolt N, Aon MA, Royer P, Pein L, et al. Alternate day fasting improves physiological and molecular markers of aging in healthy, non-obese humans. Cell Metab. 2019;30(3):462–46+.
    1. Anhê FF, Roy D, Pilon G, Dudonné S, Matamoros S, Varin TV, et al. A polyphenol-rich cranberry extract protects from diet-induced obesity, insulin resistance and intestinal inflammation in association with increased Akkermansia spp. population in the gut microbiota of mice. Gut. 2015;64(6):872–883.
    1. Saulnier DM, Riehle K, Mistretta TA, Diaz MA, Mandal D, Raza S, et al. Gastrointestinal microbiome signatures of pediatric patients with irritable bowel syndrome. Gastroenterology. 2011;141(5):1782–1791.
    1. Naseribafrouei A, Hestad K, Avershina E, Sekelja M, Linløkken A, Wilson R, et al. Correlation between the human fecal microbiota and depression. Neurogastroenterol Motil. 2014;26(8):1155–1162.
    1. Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet–induced obesity and diabetes in mice. Diabetes. 2008;57(6):1470–1481.
    1. Kim SJ, Kim SE, Kim AR, Kang S, Park MY, Sung MK. Dietary fat intake and age modulate the composition of the gut microbiota and colonic inflammation in C57BL/6J mice. BMC Microbiol. 2019;19(1):193.
    1. Su J, Li L, Wang Y, Ma X-X, Ma Z, Peppelenbosch MP, et al. Seasonal analysis of the gut microbiota in adult and young captive Asian black bears in northeast China. Anim Biol. 2020;70(1):109–119.
    1. Berg J, Brandt KK, Al-Soud WA, Holm PE, Hansen LH, Sorensen SJ, et al. Selection for Cu-tolerant bacterial communities with altered composition, but unaltered richness, via long-term Cu exposure. Appl Environ Microbiol. 2012;78(20):7438–7446.
    1. Bokulich NA, Subramanian S, Faith JJ, Gevers D, Gordon JI, Knight R, et al. Quality-filtering vastly improves diversity estimates from Illumina amplicon sequencing. Nat Methods. 2013;10(1):57–59.
    1. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, et al. QIIME allows analysis of high-throughput community sequencing data. Nat Methods. 2010;7(5):335.
    1. Haas BJ, Gevers D, Earl AM, Feldgarden M, Ward DV, Giannoukos G, et al. Chimeric 16S rRNA sequence formation and detection in Sanger and 454-pyrosequenced PCR amplicons. Genome Res. 2011;21(3):494–504.
    1. Edgar RC. UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat Methods. 2013;10(10):996–998.
    1. Wang Q, Garrity GM, Tiedje JM, Cole JR. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl Environ Microbiol. 2007;73(16):5261–5267.
    1. Oksanen J, Blanchet FG, Kindt R, Legendre P, O’hara RB, Simpson GL, et al. Vegan: community ecology package. R package version 1.17–4. 2010.

Source: PubMed

3
S'abonner