Short Chain Fatty Acids Prevent High-fat-diet-induced Obesity in Mice by Regulating G Protein-coupled Receptors and Gut Microbiota

Yuanyuan Lu, Chaonan Fan, Ping Li, Yanfei Lu, Xuelian Chang, Kemin Qi, Yuanyuan Lu, Chaonan Fan, Ping Li, Yanfei Lu, Xuelian Chang, Kemin Qi

Abstract

Elucidating the mechanisms by which short chain fatty acids (SCFA) reduce body weight may assist in the development of an effective weight control strategy. Dietary supplementation of acetate, propionate, butyrate or their admixture was shown to significantly inhibit the body weight gain induced by high-fat diet feeding. Supplementation of SCFAs caused significant changes in the expressions of G-protein coupled receptor 43 (GPR43) and GPR41 characterized by increases in the adipose tissue and reductions in the colon. Additionally, they influenced the bacterial community structure in feces, with a reduction in the proportion of Firmicutes and an increase in the proportion of Bacteroidetes. The effects of dietary SCFAs on the GPR expression and gut microbiota composition may further result in body weight reduction by enhancing triglyceride hydrolysis and FFA oxidation in the adipose tissue, promoting beige adipogenesis and mitochondrial biogenesis, and inhibiting chronic inflammation.

Figures

Figure 1. SCFAs protect against the high-fat…
Figure 1. SCFAs protect against the high-fat diet induced increases in body weight.
Three- to four-week-old C57BL/6 J male mice were fed a HFD diet and four SCFA-containing HFD diets—HFD-A, HFD-P, HFD-B and HFD-SCFA—with a low-fat diet as the control (C). (A) Body weight changes during the sixteen weeks of feeding (n = 15). (B and C) Food and energy intake per mouse at 2, 8 and 12 weeks of feeding (n = 15). Data are shown as the mean ± SD. *Compared to the lean control (C) group, P < 0.05; #compared to the HFD group, P < 0.05.
Figure 2. SCFAs regulate mRNA expressions of…
Figure 2. SCFAs regulate mRNA expressions of GRP43, GPR41, gut hormone peptides and adipokines in DIO mice.
Three- to four-week-old C57BL/6 J male mice were fed different diets for 16 weeks, the mice were sacrificed and the epididymal fat and intestine were dissected in order to measure mRNA expressions of the genes investigated. (A–E) mRNA expressions in the adipose tissue; (F–I) mRNA expressions in the colon; (J–N) correlation between GPR43 and related genes. n = 8–10 in each group. *Compared to the lean control (C) group, P < 0.05; #compared to the HFD group, P < 0.05.
Figure 3. SCFAs regulate lipid metabolism.
Figure 3. SCFAs regulate lipid metabolism.
Three- to four-week-old C57BL/6 J male mice were fed different diets for 16 weeks, the mice were sacrificed and the epididymal fat, liver, intestine and muscle were dissected for measuring mRNA expressions of related genes. Blood samples were harvested by heart puncture and plasma was separated. mRNA expressions of cpt1 and cpt2 in the adipose tissue (A–C), liver (D–F) and muscle (G–I); mRNA expressions of LPL, Fiaf and HSL in the adipose tissue (J–L); plasma FFA concentration (M); ACC mRNA in the adipose tissue (N) and liver (O); correlation between fat GRP43 and cpts (P–R). n = 8–10 in each group. *Compared to the lean control (C) group, P < 0.05; #compared to the HFD group, P < 0.05.
Figure 4. SCFAs regulate beige adipocyte differentiation…
Figure 4. SCFAs regulate beige adipocyte differentiation and mitochondrial biogenesis.
Three- to four-week-old C57BL/6 J male mice were fed different diets for 16 weeks, the mice were sacrificed and the epididymal fat was dissected for measuring mRNA expressions of related genes. (A–F) Mitochondrial biogenesis-related genes (PGC-1a, NRF-1, Tfam, β-F1-ATPase, COX IV and cyt-c); G: mtDNA quantity; (H–J) beige adipocyte markers (Tbx1, Tmem26, CD137). (K–M) correlation between fat GRP43 and related genes. n = 8–10 in each group. *Compared to the lean control (C) group, P < 0.05; #compared to the HFD group, P < 0.05.
Figure 5. SCFAs alter gut microbiota in…
Figure 5. SCFAs alter gut microbiota in DIO mice.
(A–E) 454 pyrosequencing analysis of the fecal microbiota composition from mice fed different diets at the levels of the phylum, class, order, family and genus. (H–K) Bacterial communities based on weighted UniFrac distance. (F and G) Correlation between the ratio of Firmicutes to Bacteroidetes and the GPR43 expression levels in the adipose tissue and colon. n = 15 in each group.

References

    1. Jura M. & Kozak L. P. Obesity and related consequences to ageing. Age 38, 23 (2016).
    1. Dietz W. H. et al. Management of obesity: improvement of health-care training and systems for prevention and care. Lancet 385, 2521–2533 (2015).
    1. Kaczmarczyk M. M., Miller M. J. & Freund G. G. The health benefits of dietary fiber: beyond the usual suspects of type 2 diabetes mellitus, cardiovascular disease and colon cancer. Metabolism 61, 1058–1066 (2012).
    1. Edwards C. A., Xie C. & Garcia A. L. Dietary fibre and health in children and adolescents. Proc Nutr Soc 74, 292–302 (2015).
    1. Liu X., Wu Y., Li F. & Zhang D. Dietary fiber intake reduces risk of inflammatory bowel disease: result from a meta-analysis. Nutr Res 35, 753–758 (2015).
    1. Keenan M. J. et al. Role of resistant starch in improving gut health, adiposity, and insulin resistance. Adv Nutr 6, 198–205 (2015).
    1. Anastasovska J. et al. Fermentable carbohydrate alters hypothalamic neuronal activity and protects against the obesogenic environment. Obesity (Silver Spring) 20, 1016–1023 (2012).
    1. Wanders A. J. et al. Effects of dietary fibre on subjective appetite, energy intake and body weight: a systematic review of randomized controlled trials. Obes Rev 12, 724–739 (2011).
    1. Tan J. et al. The role of short-chain fatty acids in health and disease. Adv Immunol 121, 91–119 (2014).
    1. Canfora E. E., Jocken J. W.& Blaak E. E. Short-chain fatty acids in control of body weight and insulin sensitivity. Nat Rev Endocrinol 11, 577–591 (2015).
    1. Binder H. J. Role of colonic short-chain fatty acid transport in diarrhea. Annu Rev Physiol 72, 297–313 (2010).
    1. Walker A. W. et al. pH and peptide supply can radically alter bacterial populations and short-chain fatty acid ratios within microbial communities from the human colon. Appl Environ Microbiol 71, 3692–3700 (2005).
    1. Byrne C. S., Chambers E. S., Morrison D. J. & Frost G. The role of short chain fatty acids in appetite regulation and energy homeostasis. Int J Obes (Lond) 39, 1331–1338 (2015).
    1. Chambers E. S., Morrison D. J. & Frost G. Control of appetite and energy intake by SCFA: what are the potential underlying mechanisms? Proc Nutr Soc 74, 328–336 (2015).
    1. Hara T. et al. Role of free fatty acid receptors in the regulation of energy metabolism. Biochim Biophys Acta 1841, 1292–1300 (2014).
    1. Ang Z. & Ding J. L. GPR41 and GPR43 in Obesity and Inflammation - Protective or Causative? Front Immunol 7, 28 (2016).
    1. Kimura I. et al. The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat Commun 4, 1829 (2013).
    1. Bjursell M. et al. Improved glucose control and reduced body fat mass in free fatty acid receptor 2-deficient mice fed a high-fat diet. Am J Physiol Endocrinol Metab 300, E211–E220 (2011).
    1. Bäckhed F. et al. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci USA 101, 15718–15723 (2004).
    1. Ley R. E. et al. Obesity alters gut microbial ecology. Proc Natl Acad Sci USA 102, 11070–11075 (2005).
    1. Million M., Lagier J. C., Yahav D. & Paul M. Gut bacterial microbiota and obesity. Clin Microbiol Infect 19, 305–313 (2013).
    1. Turnbaugh P. J. et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 444, 1027–1031 (2006).
    1. Schwiertz A. et al. Microbiota and SCFA in lean and overweight healthy subjects. Obesity (Silver Spring) 18, 190–195 (2010).
    1. Lin H. V. et al. Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLoS One 7, e35240 (2012).
    1. Chambers E. S. et al. Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut 64, 1744–1754 (2015).
    1. den Besten G. et al. Short-Chain Fatty Acids Protect Against High-Fat Diet-Induced Obesity via a PPARγ-Dependent Switch From Lipogenesis to Fat Oxidation. Diabetes 64, 2398–2408 (2015).
    1. Rahat-Rozenbloom S., Fernandes J., Gloor G. B. & Wolever T. M. Evidence for greater production of colonic short-chain fatty acids in overweight than lean humans. Int J Obes (Lond) 38, 1525–1531 (2014).
    1. Murphy E. F. et al. Composition and energy harvesting capacity of the gut microbiota: relationship to diet, obesity and time in mouse models. Gut 59, 1635–1642 (2010).
    1. Psichas A. et al. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. Int J Obes (Lond) 39, 424–429 (2015).
    1. Zaibi M. S. et al. Roles of GPR41 and GPR43 in leptin secretory responses of murine adipocytes to short chain fatty acids. FEBS Lett 584, 2381–2386 (2010).
    1. Hong Y. H. et al. Acetate and propionate short chain fatty acids stimulate adipogenesis via GPCR43. Endocrinology. 146, 5092–5099 (2005).
    1. Frost G. et al. Effect of short chain fatty acids on the expression of free fatty acid receptor 2 (Ffar2), Ffar3 and early-stage adipogenesis. Nutr Diabetes. 4, e128 (2014).
    1. Shen W. et al. Epigenetic modification of the leptin promoter in diet-induced obese mice and the effects of n-3 polyunsaturated fatty acids. Sci Rep. 4, 5282 (2014).
    1. Weyer C. et al. Hypoadiponectinemia in obesity and type 2 diabetes: close association with insulin resistance and hyperinsulinemia. J Clin Endocrinol Metab. 86, 1930–1935 (2001).
    1. Levy J. R., Davenport B., Clore J. N. & Stevens W. Lipid metabolism and resistin gene expression in insulin-resistant Fischer 344 rats. Am J Physiol Endocrinol Metab. 282, E626–E633 (2002).
    1. Krishnan J. et al. Dietary obesity-associated Hif1α activation in adipocytes restricts fatty acid oxidation and energy expenditure via suppression of the Sirt2-NAD+ system. Genes Dev 26, 259–270 (2012).
    1. Tedesco L. et al. Cannabinoid type 1 receptor blockade promotes mitochondrial biogenesis through endothelial nitric oxide synthase expression in white adipocytes. Diabetes. 57, 2028–2036 (2008).
    1. Wu J. et al. Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human. Cell. 150, 366–376 (2012).
    1. Lee Y. H., Petkova A. P., Mottillo E. P. & Granneman J. G. In vivo identification of bipotential adipocyte progenitors recruited by b3-adrenoceptor activation and high-fat feeding. Cell Metab. 15, 480–491 (2012).
    1. Krajmalnik-Brown R., Ilhan Z. E., Kang D. W. & DiBaise J. K. Effects of gut microbes on nutrient absorption and energy regulation. Nutr Clin Pract 27, 201–214 (2012).
    1. Ley R. E., Turnbaugh P. J., Klein S. & Gordon J. I. Microbial ecology: human gut microbes associated with obesity. Nature 444, 1022–1023 (2006).
    1. Nadal I. et al. Shifts in clostridia, bacteroides and immunoglobulin-coating fecal bacteria associated with weight loss in obese adolescents. Int J Obes (Lond) 33, 758–767 (2009).
    1. Mai V., McCrary Q. M., Sinha R. & Glei M. Associations between dietary habits and body mass index with gut microbiota composition and fecal water genotoxicity: an observational study in African American and Caucasian American volunteers. Nutr J 8, 49 (2009).
    1. Duncan S. H. et al. Human colonic microbiota associated with diet, obesity and weight loss. Int J Obes (Lond) 32, 1720–1724 (2008).
    1. Zhang H. et al. Human gut microbiota in obesity and after gastric bypass. Proc Natl Acad Sci USA 106, 2365–2370 (2009).
    1. Bäckhed F., Manchester J. K., Semenkovich C. F. & Gordon J. I. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci USA 104, 979–984 (2007).
    1. Raybould H. E. Gut microbiota, epithelial function and derangements in obesity. J Physiol 590, 441–446 (2012).
    1. Geurts L. et al. Gut microbiota controls adipose tissue expansion, gut barrier and glucose metabolism: novel insights into molecular targets and interventions using prebiotics. Benef Microbes 5, 3–17 (2014).
    1. Cani P. D. et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 56, 1761–1772 (2007).
    1. Samuel B. S. et al. Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proc Natl Acad Sci USA 105, 16767–16772 (2008).
    1. Karlsson C. L. et al. The microbiota of the gut in preschool children with normal and excessive body weight. Obesity (Silver Spring) 20, 2257–2261 (2012).
    1. Sawin E. A. et al. Glycomacropeptide is a prebiotic that reduces Desulfovibrio bacteria, increases cecal short-chain fatty acids, and is anti-inflammatory in mice. Am J Physiol Gastrointest Liver Physiol 309, G590–G 601 (2015).
    1. Garcia-Mazcorro J. F., Ivanov I., Mills D. A. & Noratto G. Influence of whole-wheat consumption on fecal microbial community structure of obese diabetic mice. PeerJ 4, e1702 (2016).
    1. Daniel H. et al. High-fat diet alters gut microbiota physiology in mice. ISME J 8, 295–308 (2014).
    1. Patrone V. et al. Postoperative Changes in Fecal Bacterial Communities and Fermentation Products in Obese Patients Undergoing Bilio-Intestinal Bypass. Front Microbiol 7, 200 (2016).
    1. Kameyama K. & Itoh K. Intestinal colonization by a Lachnospiraceae bacterium contributes to the development of diabetes in obese mice. Microbes Environ 29, 427–430 (2014).
    1. Cani P. D. et al. Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia. Diabetologia 50, 2374–2383 (2007).
    1. Million M. et al. Obesity-associated gut microbiota is enriched in Lactobacillus reuteri and depleted in Bifidobacterium animalis and Methanobrevibacter smithii. Int. J. Obes 36, 817–825 (2012).
    1. Million M. et al. Correlation between body mass index and gut concentrations of Lactobacillus reuteri, Bifidobacterium animalis, Methanobrevibacter smithii and Escherichia coli. Int. J. Obes. (Lond.) 37, 1460–1466 (2013).
    1. Trevellin E. et al. Exercise training induces mitochondrial biogenesis and glucose uptake in subcutaneous adipose tissue through eNOS-dependent mechanisms. Diabetes. 63, 2800–2811 (2014).
    1. D’Antona G. et al. Branched-chain amino acid supplementation promotes survival and supports cardiac and skeletal muscle mitochondrial biogenesis in middle-aged mice. Cell metabolism 12, 362–372 (2010).
    1. Tangerman A. & Nagengast F. M. A gas chromatographic analysis of fecal short-chain fatty acids, using the direct injection method. Anal Biochem. 236, 1–8 (1996).
    1. Caporaso J. G. et al. Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. ISME J 6, 1621–1624 (2012).
    1. Xie Z. et al. An anaerobic dynamic membrane bioreactor (AnDMBR) for landfill leachate treatment: performance and microbial community identification. Bioresour Technol 161, 29–39 (2014).
    1. Yasir M. et al. Comparison of the gut microbiota of people in France and Saudi Arabia. Nutr Diabetes 5, e153 (2015).
    1. Lu X. M., Lu P. Z. & Zhang H. Bacterial communities in manures of piglets and adult pigs bred with different feeds revealed by 16S rDNA 454 pyrosequencing. Appl Microbiol Biotechnol 98, 2657–2665 (2014).
    1. Noval Rivas M. et al. A microbiota signature associated with experimental food allergy promotes allergic sensitization and anaphylaxis. J Allergy Clin Immunol 131, 201–212 (2013).
    1. Srinivasan S. et al. Bacterial communities in women with bacterial vaginosis: high resolution phylogenetic analyses reveal relationships of microbiota to clinical criteria. PLoS One 7, e37818 (2012).

Source: PubMed

3
Subscribe