Exposure to a Healthy Gut Microbiome Protects Against Reproductive and Metabolic Dysregulation in a PCOS Mouse Model

Pedro J Torres, Bryan S Ho, Pablo Arroyo, Lillian Sau, Annie Chen, Scott T Kelley, Varykina G Thackray, Pedro J Torres, Bryan S Ho, Pablo Arroyo, Lillian Sau, Annie Chen, Scott T Kelley, Varykina G Thackray

Abstract

Polycystic ovary syndrome (PCOS) is a common endocrine disorder affecting ∼10% to 15% of reproductive-aged women worldwide. Diagnosis requires two of the following: hyperandrogenism, oligo-ovulation or anovulation, and polycystic ovaries. In addition to reproductive dysfunction, many women with PCOS display metabolic abnormalities associated with hyperandrogenism. Recent studies have reported that the gut microbiome is altered in women with PCOS and rodent models of the disorder. However, it is unknown whether the gut microbiome plays a causal role in the development and pathology of PCOS. Given its potential role, we hypothesized that exposure to a healthy gut microbiome would protect against development of PCOS. A cohousing study was performed using a letrozole-induced PCOS mouse model that recapitulates many reproductive and metabolic characteristics of PCOS. Because mice are coprophagic, cohousing results in repeated, noninvasive inoculation of gut microbes in cohoused mice via the fecal-oral route. In contrast to letrozole-treated mice housed together, letrozole mice cohoused with placebo mice showed significant improvement in both reproductive and metabolic PCOS phenotypes. Using 16S rRNA gene sequencing, we also observed that the overall composition of the gut microbiome and the relative abundance of Coprobacillus and Lactobacillus differed in letrozole-treated mice cohoused with placebo mice compared with letrozole mice housed together. These results suggest that dysbiosis of the gut microbiome may play a causal role in PCOS and that modulation of the gut microbiome may be a potential treatment option for PCOS.

Copyright © 2019 Endocrine Society.

Figures

Figure 1.
Figure 1.
Cohousing letrozole mice with placebo mice protected against development of the PCOS metabolic phenotype. Design of cohousing study with pubertal female mice housed two per cage in three different housing arrangements that resulted in four groups of mice (n = 8 per group): P, LET, Pch, and LETch (A). Letrozole treatment resulted in metabolic dysfunction compared with placebo including (B–F) increased weight, abdominal adiposity, FBG, and insulin levels and insulin resistance. (B–F) Compared with LET mice, LETch mice showed a decrease in body weight, a decrease in abdominal adiposity, a decrease in FBG and insulin levels, and restored insulin sensitivity. Graph error bars represent SEM. Different letters or an asterisk symbol were used to indicate significant differences in a one-way ANOVA or repeated-measures two-way ANOVA followed by post hoc comparisons with the Tukey-Kramer honestly significant difference test (P < 0.05).
Figure 2.
Figure 2.
Letrozole mice cohoused with placebo mice did not become hyperandrogenemic or acyclic. The cohousing study included four groups of mice (n = 8 per group): P, LET, Pch, and LETch. Letrozole treatment resulted in increased (A) testosterone and (B) LH levels compared with placebo. LETch mice displayed a decrease in (A) testosterone and (B) LH and (C) a restoration of estrous cyclicity compared LET mice stuck in diestrus. Stages of the estrous cycle are indicated as diestrus (D), metestrus (M), estrus (E), and proestrus (P). Graphs illustrating the estrous cycle stages of representative mice from the four groups are shown. Graph error bars represent SEM. Different letters were used to indicate significant differences in a one-way ANOVA followed by post hoc comparisons with the Tukey-Kramer honestly significant difference test (P < 0.05).
Figure 3.
Figure 3.
Cohousing letrozole mice with placebo mice improved the ovarian phenotype. The cohousing study included four groups of mice (n = 8 per group): P, LET, Pch, and LETch. (A) Letrozole treatment resulted in a lack of corpora lutea, cystlike follicles, and hemorrhagic cysts in the ovaries compared with placebo mice. (A) Unlike LET mice, LETch mice lacked polycystic ovaries, and their ovaries contained corpora lutea (CL) which is evidence of ovulation. Scale bars represent 250 µm. (B–E) Letrozole treatment also resulted in increased ovarian weight and increased mRNA expression of several ovarian genes important in ovarian follicular development and steroidogenesis. (B) Ovarian weight was lower in LETch mice compared with LET mice. Fshr and Cyp19 mRNA levels were similar between LET and LETch mice, whereas Cyp17 was lower in LETch mice compared with LET mice. Graph error bars represent standard error of the mean. Different letters were used to indicate significant differences in a one-way ANOVA followed by post hoc comparisons with the Tukey-Kramer honestly significant difference test (P < 0.05).
Figure 4.
Figure 4.
Gut microbiome was similar in all cohousing treatment groups before treatment. The cohousing study included four groups of mice: P, LET, Pch, and LETch (n = 8 per group with the exception of n = 7 for P time 4 and n = 6 for Pch and LETch time 5). No significant differences in (A) gut microbial richness (α-diversity, Faith PD) or (B) community composition (β-diversity, weighted UniFrac) were observed among cohousing treatment groups before treatment (week 0; n = 8 per group). One-way ANOVA was used to compare α-diversity among the groups, and analysis of similarity (ANOSIM) test was used to compare β-diversity among the groups.
Figure 5.
Figure 5.
Cohousing letrozole mice with placebo mice did not restore α-diversity of the gut microbiome. The cohousing study included four groups of mice: (A) P, (B) LET, (C) Pch, and (D) LETch (n = 8 per group with the exception of n = 7 for P time 4 and n = 6 for Pch and LETch time 5). α-Diversity as approximated by Faith PD ranked estimate was graphed over time for the four groups. Results of linear regression model (LM) and P value are in the box insets, and the gray shaded area indicates the 95% CI for the line of best fit. P values for the linear mixed effects model (LME) were obtained by the likelihood ratio test of the full model, with the effect in question (time) against the model without the effect in question, and are in the box insets.
Figure 6.
Figure 6.
Cohousing letrozole mice with placebo mice influenced the overall composition of the gut bacterial community over time. The cohousing study included four groups of mice: P, LET, Pch, and LETch (n = 8 per group with the exception of n = 7 for P time 4 and n = 6 for Pch and LETch time 5). (A) Unconstrained PCoA of weighted UniFrac distances demonstrated changes in the microbial composition (β-diversity) among samples collected after treatment. Permutational ANOVA of the weighted UniFrac distances indicated that cohousing had a strong influence on the gut microbial community (P = 0.001). (B) Constrained CAP of weighted UniFrac distances further illustrated the relationship between β-diversity and posttreatment with a significant effect of constraining the data based on the cohousing treatment group (P = 0.001). (C–H) Samples from the different groups were then compared at each time point. Permutational ANOVA of the weighted UniFrac distances was done for each time point.
Figure 7.
Figure 7.
Specific bacterial genera were associated with improvement of the PCOS phenotype during cohousing. The cohousing study included four groups of mice: P, LET, Pch, and LETch (n = 8 per group with the exception of n = 7 for P time 4 and n = 6 for Pch and LETch time 5). Results from the DESeq2 differential abundance analysis were expressed as log2 fold change for (A) the comparison of P and LET mice and (B) the comparison of LETch and LET mice. Positive log2 fold changes represent bacterial genera increased in (A) P mice relative to LET mice or (B) LETch relative to LET mice, whereas negative changes represent bacterial general increased in (A) LET relative to P mice or (B) LET relative to LETch mice. *P < 0.05; **P < 0.01; ***P < 0.001.

References

    1. Fauser BC, Tarlatzis BC, Rebar RW, Legro RS, Balen AH, Lobo R, Carmina E, Chang J, Yildiz BO, Laven JS, Boivin J, Petraglia F, Wijeyeratne CN, Norman RJ, Dunaif A, Franks S, Wild RA, Dumesic D, Barnhart K. Consensus on women’s health aspects of polycystic ovary syndrome (PCOS): the Amsterdam ESHRE/ASRM-Sponsored 3rd PCOS Consensus Workshop Group. Fertil Steril. 2012;97:28–38 e25.
    1. Azziz R, Carmina E, Chen Z, Dunaif A, Laven JSE, Legro RS, Lizneva D, Natterson-Horowtiz B, Teede HJ, Yildiz BO. Polycystic ovary syndrome. Nat Rev Dis Primers. 2016;2:16057.
    1. Dumesic DA, Oberfield SE, Stener-Victorin E, Marshall JC, Laven JS, Legro RS. Scientific statement on the diagnostic criteria, epidemiology, pathophysiology, and molecular genetics of polycystic ovary syndrome. Endocr Rev. 2015;36(5):487–525.
    1. Churchill SJ, Wang ET, Pisarska MD. Metabolic consequences of polycystic ovary syndrome. Minerva Ginecol. 2015;67(6):545–555.
    1. Goodman NF, Cobin RH, Futterweit W, Glueck JS, Legro RS, Carmina E;. American Association of Clinical Endocrinologists (AACE); American College of Endocrinology (ACE); Androgen Excess and PCOS Society. American Association of Clinical Endocrinologists, American College of Endocrinology, and Androgen Excess and PCOS Society disease state clinical review: guide to the best practices in the evaluation and treatment of polycystic ovary syndrome—part 2. Endocr Pract. 2015;21(12):1415–1426.
    1. Barber TM, Wass JAH, McCarthy MI, Franks S. Metabolic characteristics of women with polycystic ovaries and oligo-amenorrhoea but normal androgen levels: implications for the management of polycystic ovary syndrome. Clin Endocrinol (Oxf). 2007;66(4):513–517.
    1. Moghetti P, Tosi F, Bonin C, Di Sarra D, Fiers T, Kaufman J-M, Giagulli VA, Signori C, Zambotti F, Dall’Alda M, Spiazzi G, Zanolin ME, Bonora E. Divergences in insulin resistance between the different phenotypes of the polycystic ovary syndrome. J Clin Endocrinol Metab. 2013;98(4):E628–E637.
    1. Legro RS, Driscoll D, Strauss JF, Fox J, Dunaif A. Evidence for a genetic basis for hyperandrogenemia in polycystic ovary syndrome. Proc Natl Acad Sci USA. 1998;95(25):14956–14960.
    1. Vink JM, Sadrzadeh S, Lambalk CB, Boomsma DI. Heritability of polycystic ovary syndrome in a Dutch twin-family study. J Clin Endocrinol Metab. 2006;91(6):2100–2104.
    1. Abbott DH, Bacha F. Ontogeny of polycystic ovary syndrome and insulin resistance in utero and early childhood. Fertil Steril. 2013;100(1):2–11.
    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. Walker AW, Lawley TD. Therapeutic modulation of intestinal dysbiosis. Pharmacol Res. 2013;69(1):75–86.
    1. Bäumler AJ, Sperandio V. Interactions between the microbiota and pathogenic bacteria in the gut. Nature. 2016;535(7610):85–93.
    1. Gensollen T, Iyer SS, Kasper DL, Blumberg RS. How colonization by microbiota in early life shapes the immune system. Science. 2016;352(6285):539–544.
    1. Natividad JM, Verdu EF. Modulation of intestinal barrier by intestinal microbiota: pathological and therapeutic implications. Pharmacol Res. 2013;69(1):42–51.
    1. den Besten G, van Eunen K, Groen AK, Venema K, Reijngoud DJ, Bakker BM. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res. 2013;54(9):2325–2340.
    1. Ridlon JM, Kang DJ, Hylemon PB, Bajaj JS. Bile acids and the gut microbiome. Curr Opin Gastroenterol. 2014;30(3):332–338.
    1. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444(7122):1022–1023.
    1. Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, Liang S, Zhang W, Guan Y, Shen D, Peng Y, Zhang D, Jie Z, Wu W, Qin Y, Xue W, Li J, Han L, Lu D, Wu P, Dai Y, Sun X, Li Z, Tang A, Zhong S, Li X, Chen W, Xu R, Wang M, Feng Q, Gong M, Yu J, Zhang Y, Zhang M, Hansen T, Sanchez G, Raes J, Falony G, Okuda S, Almeida M, LeChatelier E, Renault P, Pons N, Batto JM, Zhang Z, Chen H, Yang R, Zheng W, Li S, Yang H, Wang J, Ehrlich SD, Nielsen R, Pedersen O, Kristiansen K, Wang J. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490(7418):55–60.
    1. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, Sogin ML, Jones WJ, Roe BA, Affourtit JP, Egholm M, Henrissat B, Heath AC, Knight R, Gordon JI. A core gut microbiome in obese and lean twins. Nature. 2009;457(7228):480–484.
    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. Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, Griffin NW, Lombard V, Henrissat B, Bain JR, Muehlbauer MJ, Ilkayeva O, Semenkovich CF, Funai K, Hayashi DK, Lyle BJ, Martini MC, Ursell LK, Clemente JC, Van Treuren W, Walters WA, Knight R, Newgard CB, Heath AC, Gordon JI. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013;341(6150):1241214.
    1. Liu R, Zhang C, Shi Y, Zhang F, Li L, Wang X, Ling Y, Fu H, Dong W, Shen J, Reeves A, Greenberg AS, Zhao L, Peng Y, Ding X. Dysbiosis of gut microbiota associated with clinical parameters in polycystic ovary syndrome. Front Microbiol. 2017;8:324.
    1. Lindheim L, Bashir M, Münzker J, Trummer C, Zachhuber V, Leber B, Horvath A, Pieber TR, Gorkiewicz G, Stadlbauer V, Obermayer-Pietsch B. Alterations in gut microbiome composition and barrier function are associated with reproductive and metabolic defects in women with polycystic ovary syndrome (PCOS): a pilot study. PLoS One. 2017;12(1):e0168390.
    1. Torres PJ, Siakowska M, Banaszewska B, Pawelczyk L, Duleba AJ, Kelley ST, Thackray VG. Gut microbial diversity in women with polycystic ovary syndrome correlates with hyperandrogenism. J Clin Endocrinol Metab. 2018;103(4):1502–1511.
    1. Insenser M, Murri M, Del Campo R, Martínez-García MA, Fernández-Durán E, Escobar-Morreale HF. Gut microbiota and the polycystic ovary syndrome: influence of sex, sex hormones, and obesity. J Clin Endocrinol Metab. 2018;103(7):2552–2562.
    1. Kelley ST, Skarra DV, Rivera AJ, Thackray VG. The gut microbiome is altered in a letrozole-induced mouse model of polycystic ovary syndrome. PLoS One. 2016;11(1):e0146509.
    1. Guo Y, Qi Y, Yang X, Zhao L, Wen S, Liu Y, Tang L. Association between polycystic ovary syndrome and gut microbiota. PLoS One. 2016;11(4):e0153196.
    1. Moreno-Indias I, Sánchez-Alcoholado L, Sánchez-Garrido MA, Martín-Núñez GM, Pérez-Jiménez F, Tena-Sempere M, Tinahones FJ, Queipo-Ortuño MI. Neonatal androgen exposure causes persistent gut microbiota dysbiosis related to metabolic disease in adult female rats. Endocrinology. 2016;157(12):4888–4898.
    1. Sherman SB, Sarsour N, Salehi M, Schroering A, Mell B, Joe B, Hill JW. Prenatal androgen exposure causes hypertension and gut microbiota dysbiosis. Gut Microbes. 2018;9(5):400–421.
    1. Kauffman AS, Thackray VG, Ryan GE, Tolson KP, Glidewell-Kenney CA, Semaan SJ, Poling MC, Iwata N, Breen KM, Duleba AJ, Stener-Victorin E, Shimasaki S, Webster NJ, Mellon PL. A novel letrozole model recapitulates both the reproductive and metabolic phenotypes of polycystic ovary syndrome in female mice. Biol Reprod. 2015;93(3):69.
    1. Skarra DV, Hernández-Carretero A, Rivera AJ, Anvar AR, Thackray VG. Hyperandrogenemia induced by letrozole treatment of pubertal female mice results in hyperinsulinemia prior to weight gain and insulin resistance. Endocrinology. 2017;158(9):2988–3003.
    1. Maier L, Pruteanu M, Kuhn M, Zeller G, Telzerow A, Anderson EE, Brochado AR, Fernandez KC, Dose H, Mori H, Patil KR, Bork P, Typas A. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature. 2018;555(7698):623–628.
    1. RRID:, .
    1. RRID:, .
    1. RRID:, .
    1. RRID:, .
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta C(T)) method. Methods. 2001;25(4):402–408.
    1. Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Huntley J, Fierer N, Owens SM, Betley J, Fraser L, Bauer M, Gormley N, Gilbert JA, Smith G, Knight R. 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. Bokulich NA, Kaehler BD, Rideout JR, Dillon M, Bolyen E, Knight R, Huttley GA, Gregory Caporaso J. Optimizing taxonomic classification of marker-gene amplicon sequences with QIIME 2’s q2-feature-classifier plugin. Microbiome. 2018;6(1):90.
    1. Katoh K, Standley DM. MAFFT multiple sequence alignment software version 7: improvements in performance and usability. Mol Biol Evol. 2013;30(4):772–780.
    1. Price MN, Dehal PS, Arkin AP. FastTree: computing large minimum evolution trees with profiles instead of a distance matrix. Mol Biol Evol. 2009;26(7):1641–1650.
    1. Faith DP. Conservation evaluation and phylogenetic diversity. Biol Conserv. 1992;61(1):1–10.
    1. Lozupone C, Knight R. UniFrac: a new phylogenetic method for comparing microbial communities. Appl Environ Microbiol. 2005;71(12):8228–8235.
    1. Lozupone C, Lladser ME, Knights D, Stombaugh J, Knight R. UniFrac: an effective distance metric for microbial community comparison. ISME J. 2011;5(2):169–172.
    1. McMurdie PJ, Holmes S. Phyloseq: a bioconductor package for handling and analysis of high-throughput phylogenetic sequence data. Pac Symp Biocomput. 2012:235–246.
    1. Anderson MJ, Willis TJ. Canonical analysis of principal coordinates: a useful method of constrained ordination for ecology. Ecology. 2003;84(2):511–525.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):550.
    1. Kumar A, Magoffin D, Munir I, Azziz R. Effect of insulin and testosterone on androgen production and transcription of SULT2A1 in the NCI-H295R adrenocortical cell line. Fertil Steril. 2009;92(2):793–797.
    1. Zhang G, Veldhuis JD. Insulin drives transcriptional activity of the CYP17 gene in primary cultures of swine theca cells. Biol Reprod. 2004;70(6):1600–1605.
    1. Hou YP, He QQ, Ouyang HM, Peng HS, Wang Q, Li J, Lv XF, Zheng YN, Li SC, Liu HL, Yin AH. Human gut microbiota associated with obesity in Chinese children and adolescents. BioMed Res Int. 2017;2017:7585989.
    1. Tap J, Mondot S, Levenez F, Pelletier E, Caron C, Furet JP, Ugarte E, Muñoz-Tamayo R, Paslier DL, Nalin R, Dore J, Leclerc M. Towards the human intestinal microbiota phylogenetic core. Environ Microbiol. 2009;11(10):2574–2584.
    1. Armougom F, Henry M, Vialettes B, Raccah D, Raoult D. Monitoring bacterial community of human gut microbiota reveals an increase in Lactobacillus in obese patients and Methanogens in anorexic patients. PLoS One. 2009;4(9):e7125.
    1. Štšepetova J, Sepp E, Kolk H, Lõivukene K, Songisepp E, Mikelsaar M. Diversity and metabolic impact of intestinal Lactobacillus species in healthy adults and the elderly. Br J Nutr. 2011;105(8):1235–1244.
    1. Million M, Angelakis E, Paul M, Armougom F, Leibovici L, Raoult D. Comparative meta-analysis of the effect of Lactobacillus species on weight gain in humans and animals. Microb Pathog. 2012;53(2):100–108.
    1. Thackray VG. Sex, microbes, and polycystic ovary syndrome. Trends Endocrinol Metab. 2019;30(1):54–65.
    1. Plottel CS, Blaser MJ. Microbiome and malignancy. Cell Host Microbe. 2011;10(4):324–335.
    1. Teede H, Deeks A, Moran L. Polycystic ovary syndrome: a complex condition with psychological, reproductive and metabolic manifestations that impacts on health across the lifespan. BMC Med. 2010;8(1):41.
    1. Pellock SJ, Redinbo MR. Glucuronides in the gut: sugar-driven symbioses between microbe and host. J Biol Chem. 2017;292(21):8569–8576.

Source: PubMed

3
Abonnieren