Sex differences in lipid metabolism are affected by presence of the gut microbiota

Annemarie Baars, Annemarie Oosting, Mirjam Lohuis, Martijn Koehorst, Sahar El Aidy, Floor Hugenholtz, Hauke Smidt, Mona Mischke, Mark V Boekschoten, Henkjan J Verkade, Johan Garssen, Eline M van der Beek, Jan Knol, Paul de Vos, Jeroen van Bergenhenegouwen, Floris Fransen, Annemarie Baars, Annemarie Oosting, Mirjam Lohuis, Martijn Koehorst, Sahar El Aidy, Floor Hugenholtz, Hauke Smidt, Mona Mischke, Mark V Boekschoten, Henkjan J Verkade, Johan Garssen, Eline M van der Beek, Jan Knol, Paul de Vos, Jeroen van Bergenhenegouwen, Floris Fransen

Abstract

Physiological processes are differentially regulated between men and women. Sex and gut microbiota have each been demonstrated to regulate host metabolism, but it is unclear whether both factors are interdependent. Here, we determined to what extent sex-specific differences in lipid metabolism are modulated via the gut microbiota. While male and female Conv mice showed predominantly differential expression in gene sets related to lipid metabolism, GF mice showed differences in gene sets linked to gut health and inflammatory responses. This suggests that presence of the gut microbiota is important in sex-specific regulation of lipid metabolism. Further, we explored the role of bile acids as mediators in the cross-talk between the microbiome and host lipid metabolism. Females showed higher total and primary serum bile acids levels, independent of presence of microbiota. However, in presence of microbiota we observed higher secondary serum bile acid levels in females compared to males. Analysis of microbiota composition displayed sex-specific differences in Conv mice. Therefore, our data suggests that bile acids possibly play a role in the crosstalk between the microbiome and sex-specific regulation of lipid metabolism. In conclusion, our data shows that presence of the gut microbiota contributes to sex differences in lipid metabolism.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Sex-specific effects in overall ileal gene expression and related biological functions. (A) PCA was performed on IQR-based top 1,000 most variable genes in microarray analysis of male and female GF and Conv mice. (B) Significantly up- and down-regulated genes of males compared to females shown for GF and Conv mice. (C,D) Significantly up- and down-regulated genes of male compared to female (p ≤ 0.01) mice were compared between GF and Conv mice and are presented as Venn diagrams (based on intensity-based moderate t-statistics (IBMT) regularised t-test p ≤ 0.01). (E,F) IPA analysis on genes that were significantly differentially expressed between sexes in GF and Conv mice (p ≤ 0.01). The top 25 regulated biological functions in GF and Conv mice based on male-female differences are shown. Hatched bars = biological functions that occur in the top 25 of both GF and Conv mice. Filled bars = biological functions that are specific for either GF or Conv mice.
Figure 2
Figure 2
Sex-specific transcriptional effects in lipid metabolism sub-functions. (A,B) Top 18 sex-specific significantly regulated (p ≤ 0.01) sub-functions of lipid-metabolism in GF and Conv mice based on IPA. Hatched bars = sub-functions that are regulated in both GF and Conv mice. Filled bars = sub-functions that are specific for either Conv or GF mice. (C) Heat maps based on genes from 40 sub-functions in lipid metabolism for male versus female differences in Conv mice based on IPA. Gene expression is shown on a color scale for GF and Conv mice with blue indicating lower levels than average expression of respective group of mice; red shows higher values than average expression of respective group of mice. (D) Heat maps of target genes of ileum bile acid metabolism (based on KEGG database, genome net). Gene expression is shown on a color scale for Conv and GF mice. Blue shows lower levels than average expression of respective group of mice; red shows higher values than average expression of respective group of mice.
Figure 3
Figure 3
Serum bile acid concentrations. (A) Serum total bile acid concentrations. (B) Hydrophobicity index. (C) Serum primary bile acid concentrations. (D) Serum secondary bile acid concentrations. Data is presented as median and the 25–75th percentile of n ≤ 5 (GF), n ≤ 8–10 (Conv). P-values p < 0.05 were considered statistically significant and depicted as *p < 0.05; **p < 0.01 and ***p < 0.001.
Figure 4
Figure 4
Serum primary bile acid concentrations. (A) TCA. (B) TCDCA. (C) CA. (D) TAMCA. (E) TBMCA. (F) BMCA. Data is presented as median and the 25–75th percentile of n ≤ 5 (GF), n ≤ 8–10 (Conv). P-values p < 0.05 were considered statistically significant and depicted as *p < 0.05; **p < 0.01 and ***p < 0.001.
Figure 5
Figure 5
Serum secondary bile acid concentrations. (A) DCA. (B) OMCA. (C) UDCA. (D) HDCA. (E) THDCA. Data is presented as median and the 25th and 75th percentile of n ≤ 5 (GF), n ≤ 8–10 (Conv). P-values p < 0.05 were considered statistically significant and depicted as *<0.05; **<0.01 and ***p < 0.001.
Figure 6
Figure 6
Faecal microbiota characteristics. (A) Firmicutes and Bacteroides of Conv mice. (B) Firmicutes and Bacteroides index of Conv mice. (C,D) Microbiota composition of all significant differences of the genera between males and females (p ≤ 0.05). Data is presented as the relative abundance and SEM. Bacterial BSH populations are depicted in italics and bacteria expressing 7α/7ß-dehydroxylase are represented in bold (based on filtered uniprot database and Ridlon 2006).

References

    1. Murphy, M. O. & Loria, A. S. Sex-Specific Effects of Stress on Metabolic and Cardiovascular Disease: Are women at a higher risk?American journal of physiology. Regulatory, integrative and comparative physiology, ajpregu.00185.02016, 10.1152/ajpregu.00185.2016 (2017).
    1. Mauvais-Jarvis F. Sex differences in metabolic homeostasis, diabetes, and obesity. Biology of sex differences. 2015;6:14. doi: 10.1186/s13293-015-0033-y.
    1. Roeters van Lennep JE, Westerveld HT, Erkelens DW, van der Wall EE. Risk factors for coronary heart disease: implications of gender. Cardiovascular research. 2002;53:538–549. doi: 10.1016/S0008-6363(01)00388-1.
    1. Sugiyama MG, Agellon LB. Sex differences in lipid metabolism and metabolic disease risk. Biochemistry and cell biology. 2012;90:124–141. doi: 10.1139/o11-067.
    1. Mittendorfer B. Sexual dimorphism in human lipid metabolism. The Journal of nutrition. 2005;135:681–686. doi: 10.1093/jn/135.4.681.
    1. Mischke M, et al. Maternal Western-style high fat diet induces sex-specific physiological and molecular changes in two-week-old mouse offspring. PloS one. 2013;8:e78623. doi: 10.1371/journal.pone.0078623.
    1. Steegenga WT, et al. Sexually dimorphic characteristics of the small intestine and colon of prepubescent C57BL/6 mice. Biology of sex differences. 2014;5:11. doi: 10.1186/s13293-014-0011-9.
    1. Wijchers PJ, Festenstein RJ. Epigenetic regulation of autosomal gene expression by sex chromosomes. Trends in genetics. 2011;27:132–140. doi: 10.1016/j.tig.2011.01.004.
    1. Ling G, Sugathan A, Mazor T, Fraenkel E, Waxman DJ. Unbiased, genome-wide in vivo mapping of transcriptional regulatory elements reveals sex differences in chromatin structure associated with sex-specific liver gene expression. Molecular and cellular biology. 2010;30:5531–5544. doi: 10.1128/MCB.00601-10.
    1. Sugathan A, Waxman DJ. Genome-wide analysis of chromatin states reveals distinct mechanisms of sex-dependent gene regulation in male and female mouse liver. Molecular and cellular biology. 2013;33:3594–3610. doi: 10.1128/MCB.00280-13.
    1. Waxman DJ, O’connor C. Growth hormone regulation of sex-dependent liver gene expression. Molecular endocrinology. 2006;20:2613–2629. doi: 10.1210/me.2006-0007.
    1. Leuenberger N, Pradervand S, Wahli W. Sumoylated PPARα mediates sex-specific gene repression and protects the liver from estrogen-induced toxicity in mice. The Journal of clinical investigation. 2009;119:3138. doi: 10.1172/JCI39019.
    1. Wang X, Magkos F, Mittendorfer B. Sex differences in lipid and lipoprotein metabolism: it’s not just about sex hormones. The Journal of Clinical Endocrinology & Metabolism. 2011;96:885–893. doi: 10.1210/jc.2010-2061.
    1. Sommer F, Backhed F. The gut microbiota–masters of host development and physiology. Nature reviews. Microbiology. 2013;11:227–238. doi: 10.1038/nrmicro2974.
    1. Velagapudi VR, et al. The gut microbiota modulates host energy and lipid metabolism in mice. Journal of lipid research. 2010;51:1101–1112. doi: 10.1194/jlr.M002774.
    1. Wahlström A, Sayin SI, Marschall H-U, Bäckhed F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell metabolism. 2016;24:41–50. doi: 10.1016/j.cmet.2016.05.005.
    1. Tremaroli V, Backhed F. Functional interactions between the gut microbiota and host metabolism. Nature. 2012;489:242–249. doi: 10.1038/nature11552.
    1. Baars A, Oosting A, Knol J, Garssen J, van Bergenhenegouwen J. The Gut Microbiota as a Therapeutic Target in IBD and Metabolic Disease: A Role for the Bile Acid Receptors FXR and TGR5. Microorganisms. 2015;3:641–666. doi: 10.3390/microorganisms3040641.
    1. Inagaki T, et al. Fibroblast growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis. Cell metabolism. 2005;2:217–225. doi: 10.1016/j.cmet.2005.09.001.
    1. Kim I, et al. Differential regulation of bile acid homeostasis by the farnesoid X receptor in liver and intestine. Journal of lipid research. 2007;48:2664–2672. doi: 10.1194/jlr.M700330-JLR200.
    1. Sayin SI, et al. Gut microbiota regulates bile acid metabolism by reducing the levels of tauro-beta-muricholic acid, a naturally occurring FXR antagonist. Cell metabolism. 2013;17:225–235. doi: 10.1016/j.cmet.2013.01.003.
    1. Out C, et al. Gut microbiota inhibit Asbt-dependent intestinal bile acid reabsorption via Gata4. J Hepatol. 2015;63:697–704. doi: 10.1016/j.jhep.2015.04.030.
    1. Ridlon JM, Kang D-J, Hylemon PB. Bile salt biotransformations by human intestinal bacteria. Journal of lipid research. 2006;47:241–259. doi: 10.1194/jlr.R500013-JLR200.
    1. Joyce, S. A., Shanahan, F., Hill, C. & Gahan, C. G. Bacterial bile salt hydrolase in host metabolism: potential for influencing astrointestinal microbe-host crosstalk. Gut microbes, 00–00 (2014).
    1. Joyce SA, et al. Regulation of host weight gain and lipid metabolism by bacterial bile acid modification in the gut. Proceedings of the National Academy of Sciences. 2014;111:7421–7426. doi: 10.1073/pnas.1323599111.
    1. Mueller S, et al. Differences in Fecal Microbiota in Different European Study Populations in Relation to Age, Gender, and Country: a Cross-Sectional Study. Applied and Environmental Microbiology. 2006;72:1027–1033. doi: 10.1128/AEM.72.2.1027-1033.2006.
    1. Haro C, et al. Intestinal microbiota is influenced by gender and body mass index. PloS one. 2016;11:e0154090. doi: 10.1371/journal.pone.0154090.
    1. Wang, J.-j. et al. Sex differences in colonization of gut microbiota from a man with short-term vegetarian and inulin-supplemented diet in germ-free mice. Scientific Reports6 (2016).
    1. Fu, Z. D., Csanaky, I. L. & Klaassen, C. D. Gender-divergent profile of bile acid homeostasis during aging of mice (2012).
    1. Turley SD, Schwarz M, Spady DK, Dietschy JM. Gender-related differences in bile acid and sterol metabolism in outbred CD-1 mice fed low- and high-cholesterol diets. Hepatology. 1998;28:1088–1094. doi: 10.1002/hep.510280425.
    1. Selwyn FP, Csanaky IL, Zhang Y, Klaassen CD. Importance of large intestine in regulating bile acids and glucagon-like peptide-1 in germ-free mice. Drug Metabolism and Disposition. 2015;43:1544–1556. doi: 10.1124/dmd.115.065276.
    1. Larsson, E. et al. Analysis of gut microbial regulation of host gene expression along the length of the gut and regulation of gut microbial ecology through MyD88. Gut, gutjnl-2011-301104 (2011).
    1. Sheng L, et al. Gender Differences in Bile Acids and Microbiota in Relationship with Gender Dissimilarity in Steatosis Induced by Diet and FXR Inactivation. Scientific Reports. 2017;7:1748. doi: 10.1038/s41598-017-01576-9.
    1. Kasahara K, et al. Commensal bacteria at the crossroad between cholesterol homeostasis and chronic inflammation in atherosclerosis. Journal of Lipid Research. 2017;58:519–528. doi: 10.1194/jlr.M072165.
    1. Org, E. et al. Sex differences and hormonal effects on gut microbiota composition in mice. Gut Microbes, 0, 10.1080/19490976.2016.1203502 (2016).
    1. Swann JR, et al. Systemic gut microbial modulation of bile acid metabolism in host tissue compartments. Proceedings of the National Academy of Sciences. 2011;108:4523–4530. doi: 10.1073/pnas.1006734107.
    1. Hirokane H, Nakahara M, Tachibana S, Shimizu M, Sato R. Bile acid reduces the secretion of very low density lipoprotein by repressing microsomal triglyceride transfer protein gene expression mediated by hepatocyte nuclear factor-4. Journal of Biological Chemistry. 2004;279:45685–45692. doi: 10.1074/jbc.M404255200.
    1. Kast HR, et al. Farnesoid X-activated receptor induces apolipoprotein C-II transcription: a molecular mechanism linking plasma triglyceride levels to bile acids. Molecular Endocrinology. 2001;15:1720–1728. doi: 10.1210/mend.15.10.0712.
    1. Watanabe M, et al. Bile acids lower triglyceride levels via a pathway involving FXR, SHP, and SREBP-1c. The Journal of clinical investigation. 2004;113:1408–1418. doi: 10.1172/JCI21025.
    1. Murphy E, et al. Composition and energy harvesting capacity of the gut microbiota: relationship to diet, obesity and time in mouse models. Gut. 2010;59:1635–1642. doi: 10.1136/gut.2010.215665.
    1. Dai, M. et al. Evolving gene/transcript definitions significantly alter the interpretation of GeneChip data. Nucleic Acids Research33 (2005).
    1. Bolstad B, Collin F, Simpson K, Irizarry R, Speed T. Experimental design and low-level analysis of microarray data. International review of neurobiology. 2004;60:25–58. doi: 10.1016/S0074-7742(04)60002-X.
    1. Storey JD. A direct approach to false discovery rates. Journal of the Royal Statistical Society: Series B (Statistical Methodology) 2002;64:479–498. doi: 10.1111/1467-9868.00346.
    1. Berry D, Mahfoudh KB, Wagner M, Loy A. Barcoded primers used in multiplex amplicon pyrosequencing bias amplification. Applied and environmental microbiology. 2011;77:7846–7849. doi: 10.1128/AEM.05220-11.
    1. Ramiro-Garcia, J. et al. NG-Tax, a highly accurate and validated pipeline for analysis of 16S rRNA amplicons from complex biomes. F1000Research5 (2016).
    1. Caporaso JG, et al. QIIME allows analysis of high-throughput community sequencing data. Nature methods. 2010;7:335–336. doi: 10.1038/nmeth.f.303.
    1. Thissen D, Steinberg L, Kuang D. Quick and easy implementation of the Benjamini-Hochberg procedure for controlling the false positive rate in multiple comparisons. Journal of Educational and Behavioral Statistics. 2002;27:77–83. doi: 10.3102/10769986027001077.
    1. Fransen F, et al. The Impact of Gut Microbiota on Gender-Specific Differences inImmunity. Frontiers in immunology. 2017;8:754. doi: 10.3389/fimmu.2017.00754.

Source: PubMed

3
購読する