Skip to main content

Effects of gut microbiota on omega-3-mediated ovary and metabolic benefits in polycystic ovary syndrome mice

Abstract

Background

Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disorder that frequently exhibits low-grade inflammation, pro-oxidant activity, and gut dysbiosis. PCOS has become one of the leading causes of female infertility worldwide. Recently, omega-3 polyunsaturated fatty acids (PUFAs) have been proven to benefit metabolic disorders in PCOS patients. However, its roles in the regulation of metabolic and endocrinal balances in PCOS pathophysiology are not clear. In the present study, we aimed to explore how omega-3 PUFAs alleviate ovarian dysfunction and insulin resistance in mice with dehydroepiandrosterone (DHEA)-induced PCOS by modulating the gut microbiota.

Methods

We induced PCOS in female mice by injecting them with DHEA and then treated them with omega-3 PUFAs. 16S ribosomal DNA (rDNA) amplicon sequencing, fecal microbiota transplantation (FMT) and antibiotic treatment were used to evaluate the role of microbiota in the regulation of ovarian functions and insulin resistance (IR) by omega-3 PUFAs. To further investigate the mechanism of gut microbiota on omega-3-mediated ovarian and metabolic protective effects, inflammatory and oxidative stress markers in ovaries and thermogenic markers in subcutaneous and brown adipose tissues were investigated.

Results

We found that oral supplementation with omega-3 PUFAs ameliorates the PCOS phenotype. 16S rDNA analysis revealed that omega-3 PUFA treatment increased the abundance of beneficial bacteria in the gut, thereby alleviating DHEA-induced gut dysbiosis. Antibiotic treatment and FMT experiments further demonstrated that the mechanisms underlying omega-3 benefits likely involve direct effects on the ovary to inhibit inflammatory cytokines such as IL-1β, TNF-α and IL-18. In addition, the gut microbiota played a key role in the improvement of adipose tissue morphology and function by decreasing multilocular cells and thermogenic markers such as Ucp1, Pgc1a, Cited and Cox8b within the subcutaneous adipose tissues.

Conclusion

These findings indicate that omega-3 PUFAs ameliorate androgen-induced gut microbiota dysbiosis. The gut microbiota plays a key role in the regulation of omega-3-mediated IR protective effects in polycystic ovary syndrome mice. Moreover, omega-3 PUFA-regulated improvements in the ovarian dysfunction associated with PCOS likely involve direct effects on the ovary to inhibit inflammation. Our findings suggest that omega-3 supplementation may be a promising therapeutic approach for the treatment of PCOS by modulating gut microbiota and alleviating ovarian dysfunction and insulin resistance.

Introduction

Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disorder that affects 6–10% of women of reproductive age [1, 2]. PCOS is characterized by clinical features including androgen excess irregular menses, ovulatory dysfunction and polycystic ovarian morphology [3]. Additionally, PCOS is often accompanied by metabolic disorders such as insulin resistance (IR), hyperandrogenemia, hyperinsulinemia and obesity [4, 5]. Moreover, subsequent morbidities, such as type 2 diabetes mellitus (T2DM), cardiovascular disease, endometrial cancer and psychiatric conditions, exert much influence on individuals with PCOS [6,7,8]. Infertility and metabolic syndrome are major concerns of women with PCOS [9]. The proposed mechanisms underlying PCOS vary from genetic, lifestyle, and environmental triggers, but the pathogenesis of PCOS is poorly understood [10,11,12]. Meanwhile, evidence has shown that hyperandrogenemia, inflammation, and oxidative damage play a key role in the pathophysiology of PCOS [13]. PCOS females have significantly increased levels of proinflammatory factors, such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL- 18 and monocyte chemotactic protein (MCP-1) [14]. To date, there is no approved pharmacologic therapy for PCOS because the pathogenesis of PCOS is poorly understood and there is no consensus on the underlying mechanisms, and most drugs, such as oral contraceptives, antiandrogens, insulin sensitizers, and aromatase inhibitors, are used to treat the symptoms of PCOS in an off-label manner [15]. Lifestyle modifications and weight loss remain significant therapies for PCOS. Recently, nutrition interventions such as low-glycemic index diets and Mediterranean diets have become recommended treatments for women with PCOS [16,17,18,19].

Nutritional intervention for women with PCOS has attracted attention. In recent years, studies have shown that dietary fat is considered a biological regulator affecting metabolism, and omega-3 PUFAs may be the most effective metabolic fatty acid regulator [20,21,22,23]. Animal studies have shown that omega-3 PUFAs can prevent IR by reducing inflammation and depositing fat in insulin-sensitive tissues [24]. In addition, omega-3 PUFAs can positively affect fertility, as they play an essential role in steroidogenesis [25]. However, the role and mechanism of omega-3 fatty acid supplementation in PCOS is still uncertain. Therefore, we designed a DHEA-induced mouse model to investigate the effects of supplementation with omega-3 PUFAs on PCOS mice.

Increasing evidence shows that alterations in the gut microbiome have specific impacts on every stage of female reproduction, including follicle and oocyte maturation in the ovary, fertilization and embryo migration [26,27,28]. Recent studies have shown that PCOS is inextricably linked to the gut microbiota, and individuals with PCOS have gut microbiota communities different from those of healthy controls [29,30,31]. It has been proposed that IR, metabolic abnormalities, and sex hormone concentrations in women with PCOS may affect the diversity and composition of the gut microbiota. Moreover, gut microbiota dysbiosis could contribute to IR and obesity in patients with PCOS because of the stimulation of inflammatory activities and disruption of energy balance [32, 33]. The restoration of gut microbiota dysbiosis through dietary probiotic inulin supplementation or α-linolenic acid-rich flaxseed oil contributes to the amelioration of PCOS [34]. Accumulating evidence suggests a correlation between omega-3 PUFAs and the gut microbiota. Omega-3 PUFAs protect against intestinal barrier and permeability dysfunctions by increasing healthy bacterial composition and anti-inflammatory compounds such as short-chain fatty acids (SCFAs) while inhibiting the production of pro-inflammatory mediators such as lipopolysaccharide (LPS) [35]. Omega-3 PUFAs can increase the abundance of lactic acid-producing bacteria and decrease the Firmicutes-to-Bacteroidetes (F/B) ratio in high-fat diet-fed mice [36]. However, the effect of omega-3 PUFAs on the gut microbiota in PCOS models has not been investigated. Considering the regulatory effect of omega-3 PUFAs on the gut microbiota, associations among omega-3 PUFAs, the gut microbiota and PCOS deserve investigation. Future studies could investigate whether omega-3 PUFAs directly modulate gut microbial composition and function and whether this modulation has therapeutic effects in PCOS models.

In the present study, we aimed to explore how omega-3 PUFAs alleviate ovarian dysfunction and IR in mice with DHEA-induced PCOS by modulating the gut microbiota. We investigated the regulation of omega-3 PUFAs on gut microbial composition in DHEA-induced PCOS mouse models. We report that omega-3 PUFAs attenuate IR in PCOS in a gut microbiota-dependent manner. Moreover, omega-3 PUFA-regulated improvements in the ovarian dysfunction associated with PCOS likely involve direct effects on the ovary to inhibit inflammation. We show that omega-3 PUFAs represent potential prebiotic agents for the treatment of PCOS.

Methods

Animals and treatments

Female (3 weeks old) C57BL/6 mice were purchased from Jiangsu ALF Biotechnology Company and housed in specific pathogen-free (SPF) conditions with free access to regular rodent chow and water. The SPF animal facility, including germ-free isolators at Jinling Hospital, was maintained in a 12-hour light and dark cycle at 23 ± 2 °C and 50 ± 10% humidity. Use a microisolator (CBC filter bonneted) and individually ventilated cages (PIV/IVC). Sterilize or disinfect food, water, cages and anything that will come in contact with the mice. The cage and water were changed twice a week. All animal protocols were approved by the Animal Care and Use Committee of Jinling Hospital. All of the experiments were carried out in accordance with the approved guidelines by Jinling Hospital. They were randomly divided into different groups and housed in individually ventilated cages (3–5 mice in each cage).

The DHEA-induced PCOS mouse model was established based on a previous report [37]. Mice in the experimental groups were subcutaneously injected with DHEA dissolved in sesame oil (D4000; Sigma–Aldrich; 60 mg/kg body weight per day for 21 days, n = 6 each).

For the omega-3-treated mice, after 3 weeks of DHEA treatment, the mice received another 8 weeks of treatment by gavage with omega-3 PUFAs (2 g/kg every 2 days) for the omega-3 group (n = 6 each), while PCOS mice were treated with an equal volume of corn oil at 2 g/kg for comparison. The mice were anesthetized with ether and killed by cervical dislocation. Cervical dislocation after anesthesia is a humane method of euthanasia according to the Guide for the Care and Use of Laboratory Animals (Eighth Edition) made by the Institutional Animal Care & Use Committee.

Serum was collected to measure sex hormone levels at diestrus stages. The ovaries were processed for hematoxylin and eosin staining. The remaining ovaries were quickly frozen and stored at − 80 °C. For H&E staining, ovaries were fixed in 4% paraformaldehyde (PFA) for 24 h and embedded in paraffin. Section (5 μm) were prepared and dewaxed prior to straining. The sera were collected for hormone level analysis; ovarian tissues were collected for H&E staining and real-time quantitative PCR analysis.

Vaginal smears and estrous cycle determination

Vaginal smears were taken daily at 9:00 a.m. from the 75th to the 84th day after the first day of treatment. The stage of the estrous cycle was determined by microscopic analysis of the predominant cell type in vaginal smears following H&E staining. Proestrus consists of round, nucleated epithelial cells; estrus consists of cornified squamous epithelial cells; metestrus consists of epithelial cells and leukocytes; and diestrus consists of nucleated epithelial cells and a predominance of leukocytes. The estrous cycle was determined by classic criteria [38].

Serum analysis

Blood samples were collected in 1.5 mL EP tubes, blood clotting was performed for 1 h at 4 °C, and the tubes were centrifuged at 500 × g/4°C for 10 min to harvest serum. Serum samples and conditional cell culture media were stored at -80 °C for subsequent serum determinations. The levels of follicle-stimulating hormone (FSH) (CEA830Mu; Cloud-Clone), luteinizing hormone (LH) (CEA441Mu; Cloud-Clone), estradiol 2 (E2) (ml001962; mlbio) and testosterone (T) (ml001954; mlbio) were determined by enzyme-linked immunosorbent assay kits for mice.

RNA extraction and real-time quantitative PCR (qPCR)

Total RNA was extracted using a Total RNA Purification Kit (B0004-plus; EZBioscience), and cDNA was reverse-transcribed from 1 µg total RNA using HiScript III RT SuperMix (R312-01; Vazyme) according to the manufacturer’s protocol. RT–qPCR was performed in triplicate in 96-well plates using SYBR qPCR Master Mix (Q111-02; Vazyme) with gene-specific forward and reverse primers. cDNA was amplified for 40 cycles using a Roche LC480 Real-time PCR system. β-actin was used as a reference gene. The qPCR primers used in this study can be found in Table 1.

Table 1 Primer sequences for RT-qPCR (from 5’ to 3’ terminal)

16S rDNA amplicon sequencing and bioinformatics analysis

Microbial DNA was extracted using HiPure StoolDNA Kits (Magen, Guangzhou, China) according to the manufacturer’s protocols. High-throughput sequencing was used to research the V3 − V4 region of 16S rDNA with a sequencing depth of 60,000 in the control, PCOS and omega-3 groups (n = 3 each). The 16S rDNA V3-V4 region of the ribosomal RNA gene was amplified by PCR (94 °C for 2 min, followed by 30 cycles at 98 °C for 10 s, 62 °C for 30 s, and 68 °C for 30 s and a final extension at 68 °C for 5 min) using primers 341 F: CCTACGGGNGGCWGCAG; 806R: GGACTACHVGGGTATCTAAT. PCRs were performed in triplicate in a 50 µL mixture containing 5 µL of 10. KOD buffer, 5 µL of 2 mM dNTPs, 3 µL of 25 mM MgSO4, 1.5 µL of each primer (10 µM), 1 µL of KOD polymerase, and 100 ng of template DNA. Noisy sequences of raw tags were filtered by the QIIME (version 1.9.1) pipeline under specific filtering conditions to obtain high-quality clean tags. Clean tags were searched against the reference database (version r20110519) to perform reference-based chimera checking using the UCHIME algorithm. All chimeric tags were removed, and finally, the obtained effective tags were used for further analysis. The effective tags were clustered into operational taxonomic units (OTUs) of ≥ 97% similarity using the UPARSE (version 9.2.64) pipeline. Taxonomic classification was conducted by BLAST (version 2.6.0), searching the representative OTU sequences against the NCBI 16S ribosomal RNA Database (Bacteria and archaea) (http://www.ncbi.nlm.nih.gov) (version 202,101) using the best hit with strict criteria (E value < e-5, query coverage ≥ 60% and the following identity thresholds: a hit with sequence identity ≥ 92% was considered to belong to the same species; with sequence identity ≥ 88% as indicators of belonging to the same genus; with sequence identity ≥ 85% as indicators of the same family; with sequence identity ≥ 80% as indicators of the same order; the classes were inferred when sequence identity ≥ 75%; the phylum were inferred when sequence identity ≥ 70%). The tag sequence with the highest abundance was selected as a representative sequence within each cluster. The representative sequences were classified into organisms by a naive Bayesian model using the RDP classifier (version 2.2) with confidence threshold values ranging from 0.8 to 1.

We evaluated the adequacy of sequencing by plotting the rarefaction curve for each index and indirectly reflected the richness of species in the sample. The rarefaction curve calculates the expected value of each index by sampling n tags several times and then makes a curve according to a set of n values (i.e., the number of samplings, which is generally a group of arithmetic sequences less than the total number of sequences) and the corresponding expected value of the index. When the curve flattens or reaches a plateau, it can be considered that the increase in sequencing depth has no effect on species diversity. This indicated that the amount of sequencing was sufficient. The OTU rarefaction curve was plotted in the R project ggplot2 package (version 2.2.1).

Sequences were rarefied prior to calculation of alpha and beta diversity statistics. Alpha diversity indexes were calculated in QIIME (version 1.9.1) from rarefied samples using the Shannon index for diversity and the Chao1 index for richness. Beta diversity was calculated using the unweighted pair group method using the arithmetic average (UPGMA) clustering method and principal component analysis (PCA). The total OTUs are shown in Supplementary Table 1.

Antibiotic treatment and fecal microbiota transplantation

The mice were treated with antibiotics for 7 d before transplantation experiments. Six-week-old PCOS C57BL/6 mice were supplemented with either omega-3 PUFAs or an equal volume of corn oil at 2 g/kg in the presence of the antibiotic vancomycin (0.5 g/liter), neomycin sulfate (1 g/liter), metronidazole (1 g/liter), and ampicillin (1 g/liter) [39].

Briefly, 6-week-old female donor PCOS mice were treated with omega-3 PUFAs or an equal volume of corn oil (2 g/kg). The recipient mice were treated with fresh transplant material from either PCOS mice or omega-3 PUFA-treated mice. Stool was collected daily from donor mice and pooled. Donor stool (100 mg) was diluted with saline, homogenized for 1 min using a vortex to achieve a liquid slurry, and then centrifuged at 500 × g for 3 min to remove particulate matter to facilitate administration. A total of 100 µl of the fecal suspension from either PCOS mice or omega-3 PUFA-treated mice was administered to mice by oral gavage. Fresh transplant material was prepared within 10 min before oral gavage to prevent changes in bacterial composition. Oral gavage with fecal transplant material was conducted every 2 days throughout the 8-week experiment.

Statistical analysis

Statistical analysis was performed using GraphPad Prism version 8 for Windows. The normality of the data was analyzed first, and then statistical tests were performed. Student’s t test, the Mann–Whitney test, and one-way analysis of variance (ANOVA) followed by Tukey’s post hoc test were used. All other data are presented as the mean ± standard error of the mean. The statistical significance threshold: alpha = 0.05 for hypothesis testing, as p value < alpha, we conclude that the null hypothesis should be rejected. And p values are denoted in figures and indicate the following: *P < 0.05; **P < 0.01; ***P < 0.001, ****P < 0.0001.

Results

Omega-3 PUFA supplementation attenuates endocrine and metabolic disorders in DHEA-induced PCOS mice

Because PCOS is closely associated with endocrine and metabolic disorders, to investigate the effect of omega-3 PUFAs on metabolism and endocrine function, we established a PCOS model (Fig. 1A) and assessed serum hormone levels, glucose homeostasis and insulin sensitivity in diverse groups (Fig. 1). During the 8-week treatment, the body weight of the PCOS mice was similar to that of the control group. In comparison with mice treated with DHEA alone, omega-3 PUFA treatment did not change body weight (Fig. 1B). No differences were observed in FSH levels among the three groups (Fig. 1C). The levels of LH, E2, T and the LH/FSH ratio were elevated in the PCOS mice, but these effects were reversed by omega-3 PUFA treatment (Fig. 1D-G). In addition, compared with control mice, the PCOS mice displayed higher fasting blood glucose levels, in which omega-3 PUFAs significantly decreased (Fig. 1H). Compared with healthy controls, mice treated with DHEA displayed insulin resistance, as revealed by glucose tolerance tests (GTTs) and insulin tolerance tests (ITTs). This impairment in insulin resistance was improved by omega-3 PUFA treatment (Fig. 1I, J). Together, these results demonstrate that omega-3 PUFA supplementation improved insulin resistance and sex hormone disorders in DHEA-induced PCOS mice.

Fig. 1
figure 1

Omega-3 PUFA supplementation attenuates metabolic and endocrine disorders in DHEA-induced PCOS mice. A Experimental design. B Body weights (n = 6). C-G The serum levels of FSH (C), LH (D), E2 (E), T (F) and LH/FSH (G) were measured by ELISA (n = 6). H Fasting blood glucose (n = 10). I GTT (n = 10). J ITT (n = 10), * represents the control group compared to the PCOS group, and # represents PCOS mice compared to the omega-3 group. The data are shown as the mean ± SEM with individual values. Statistical analyses were carried out using one-way ANOVA followed by Dunnett’s multiple comparisons test (*P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001). FSH, follicle-stimulating hormone; LH, luteinizing hormone; E2, estradiol 2; T, testosterone; GTT, glucose tolerance test; ITT, insulin tolerance test

Omega-3 PUFAs attenuate ovarian dysfunction in DHEA-induced PCOS mice

Infertility and metabolic syndrome are major concerns of women with PCOS. Next, we measured the effects of omega-3 PUFAs on ovarian function in PCOS-like mouse models. H&E staining was used to determine the alteration of ovarian pathology in diverse groups (Fig. 2A). The PCOS group exhibited a higher number of cystic follicles and a lower number of corpus lutea than the control group. On the other hand, omega-3 PUFA supplementation decreased the number of cystic follicles and increased the formation of corpus lutea (Fig. 2A). Compared with the PCOS group, omega-3 PUFA-treated mice displayed a decreased ovarian index and reversed disrupted estrous cycles (Fig. 2B-G). Together, these results suggest that omega-3 PUFAs ameliorated ovarian dysfunction in PCOS mice.

Fig. 2
figure 2

Omega-3 PUFAs attenuate ovarian dysfunction in DHEA-induced PCOS mice. A Representative images of ovarian sections. Scale bars: 100 μm. The asterisk (*) represents the corpus luteum, and the pound sign (#) represents the ovary vacuoles. B Estrous cycle changes. C Ovarian index. D-G Representative estrous cycles in representative mice from diverse groups. The data are shown as the mean ± SEM with individual values. Statistical analyses were carried out using one-way ANOVA followed by Dunnett’s multiple comparisons test (**P < 0.01; ***P < 0.001). P, proestrus stage; E, estrus stage; M, metestrus stage; D, diestrus stage.

Omega-3 PUFAs alleviate gut dysbiosis in PCOS mice

To explore the effects of omega-3 PUFAs on gut microbiota, we harvested fecal samples from the control, PCOS and omega-3 PUFA-treated mice and then performed 16 S rDNA analysis. Subsequently, the abundance and diversity of the bacterial community were assessed by the Shannon curve (Fig. 3A) and three α-diversity indexes, the Sob index, Chao 1 index and abundance-based coverage estimator (ACE) index. The Sob index, Chao1 and Ace index of the gut microbiota were significantly altered by omega-3 PUFAs (Fig. 3B-D), which indicates that omega-3 PUFA treatment has an obvious effect on the α-diversity of gut microbiota in PCOS mice. Next, we used the UPGMA clustering method and PCA to assess the β-diversity of the gut microbiota across diverse groups. The UPGMA clustering method showed that omega-3 PUFA supplementation affected the gut microbial profile of the PCOS mice (Fig. 3E). However, the distinction of PCA was not much different between the PCOS and omega-3 groups (Fig. 3F). Together, these results suggest that the β-diversity of the gut microbiota is not affected by omega-3 treatment.

Fig. 3
figure 3

Omega-3 PUFA treatment has an obvious effect on the α-diversity but not β-diversity of the gut microbiota in PCOS mice. The microbiota composition of control, PCOS and omega-3 mice was analyzed by 16 S rDNA pyrosequencing (n = 3 for each group). A Shannon curve. B Sob index. C Chao1 index. D Ace index. E UPGMA clustering method. F PCA of gut microbiota based on the OTU data of the control, PCOS, and omega-3 groups. PCA, principal component analysis; UPGMA, unweighted pair group method using arithmetic average. The data are shown as the mean ± SEM with individual values. Statistical analyses were carried out using one-way ANOVA followed by Dunnett’s multiple comparisons test (*P < 0.05; **P < 0.01)

To assess the overall composition of the bacterial community in different groups, we analyzed the degree of bacterial taxonomic similarity at the genus level (Fig. 4A). Compared to control mice, PCOS mice displayed an increase in the relative abundance of Alloprevotella and a decrease in the relative abundance of Akkermansia and Alistipes, while omega-3 PUFA treatment protected against these effects (Fig. 4B-K). A heatmap was further used to identify the specific bacterial phylotypes that were altered by omega-3 PUFAs (Fig. 4L). In control mice, treatment with DHEA significantly increased 6 operational taxonomic units (OTUs) and decreased 15 OTUs (Fig. 4L). Among these 21 OTUs, omega-3 PUFA treatment regulated 20 OTUs, which were altered in PCOS mice compared with control mice (Fig. 4L). These results indicate that omega-3 PUFAs modulated the gut microbiota of PCOS mice.

Fig. 4
figure 4

Omega-3 PUFAs modulate the gut microbiota of PCOS mice. A Bacterial taxonomic profiling at the genus level of intestinal bacteria from different mouse groups. B-K Relative abundance of Lachnospiraceae_NK4A136_group, Oscillibacter, Alistipes, Eubacterium_xylanophilum_group, Alloprevotella, Intestinimonas, Akkermansia, Blautia, Lachnospiraceae_UCG-006, and Roseburia. L Heatmap of the 21 OTUs in the control group altered in PCOS in response to omega-3 PUFA treatment. The phylum, family, and genus names of the OTUs are shown in the right panel. The color of the spots in the left panel represents the relative abundance at the genus level in each group. The analyses were conducted using R software version 3.3.1

Gut microbiota mediates part of the beneficial effects of omega-3 PUFAs on the ovaries

To further investigate whether the beneficial effects of omega-3 PUFAs were mediated by the gut microbiota, we transferred the microbiota from omega-3 PUFA-treated mice to PCOS recipient mice (Fig. 5A). After 8 weeks of colonization, horizontal fecal transfer from omega-3 PUFA-treated mice (omega-3→PCOS) demonstrated some protective effects similar to those observed in the omega-3 PUFA-treated group, such as fasting blood glucose (Fig. 5C) and LH/FSH ratio (Fig. 5F). However, the body weight (Fig. 5B), ovarian index, serum FSH, LH, E2 and T levels were not significantly different between the t-PCOS and t-Omega-3 groups (Fig. 5D-I). Compared with t-PCOS mice, t-Omega-3 mice also displayed disrupted estrous cycles (Fig. 5J, K). To further confirm whether the protective effects of omega-3 were conducted by microbes, we treated omega-3 PUFA-treated PCOS mice with a cocktail of antibiotics, which included vancomycin, neomycin sulfate, metronidazole, and ampicillin. When the gut microbiota was suppressed by the antibiotic cocktail, only the protective effects of LH levels were abolished (Fig. 1). Collectively, these results demonstrated that the gut microbiota mediates part of the beneficial effects of omega-3 PUFAs on the ovaries.

Fig. 5
figure 5

Gut microbiota mediates part of the beneficial effects of omega-3 PUFAs on the ovaries. Mice were randomly divided into four groups. PCOS mice were orally administered corn oil (PCOS) or omega-3 PUFAs (2 g/kg every 2 days). Horizontal fecal transfer from saline–treated PCOS mice is referred to as PCOS receivers (PCOS→PCOS). Horizontal fecal transfer from omega-3 PUFA-treated mice is referred to as omega-3 receivers (omega-3→PCOS). A Study design of the fecal transplantation experiment. B Body weight of the above four groups of mice. C Fasting blood glucose (n = 6). D-H Serum FSH, LH, LH/FSH ratio, E2 and T levels (n = 5). I Ovarian index (n = 4). J, K Representative estrous cycles. Error bars are expressed as the means ± SEMs. Statistical significance was determined by P values determined by two-tailed paired-samples t test (*P < 0.05). P, proestrus stage; E, estrus stage; M, metestrus stage; D, diestrus stage; FSH, follicle-stimulating hormone; LH, luteinizing hormone; E2, estradiol 2; T, testosterone

Omega-3 PUFAs improve the function and morphology of adipose tissues in PCOS in a gut microbiota–dependent manner

The possible mechanisms by which omega-3 regulates insulin resistance in PCOS were investigated. White adipose tissue browning increases the metabolic rate and improves insulin resistance [40, 41]. Transplantation of brown adipose tissue reversed anovulation, polycystic ovaries, insulin resistance and infertility in PCOS rats [42]. Therefore, the morphology of adipose tissues and thermogenic markers were measured in subcutaneous and brown adipose tissues from PCOS mice. Omega-3 PUFA-treated mice showed a decrease in multilocular cells within adipose tissues (Fig. 6A). PCOS mice exhibited significantly lower levels of Ucp1, Pgc1a, Cited1 and Cox8b mRNAs than controls in subcutaneous adipose tissues. Notably, the relative expression of these thermogenic markers in subcutaneous fat was dramatically elevated after omega-3 administration in the PCOS mice (Fig. 6B-E). However, although the expression of Pgc1a and Cited1 in brown fat tissues was decreased in PCOS mice, omega-3 PUFA treatment did not change these thermogenic markers (Fig. 6F-I). Interestingly, different results were observed in omega-3 + ABX mice, that is, browning of adipose tissues was decreased, as revealed by the reduced expression of Ucp1, Cited and Cox8b in subcutaneous fat tissues and the increase in multilocular cells within the adipose tissues (Fig. 6A-E). However, transferring feces from omega-3 PUFA-treated mice to PCOS mice only increased Ucp1 and Pgc1a expression in both subcutaneous and brown adipose tissues, and other thermogenic markers did not change significantly (Fig. 7A-I). These data indicated that the mechanisms underlying omega-3 PUFA-regulated improvements in the insulin resistance associated with PCOS likely involve effects on adipose tissue to decrease thermogenic markers and multilocular cells within the subcutaneous adipose tissues. Furthermore, the gut microbiota played a key role in the improvement of adipose tissue function and morphology by omega-3 in PCOS mice.

Fig. 6
figure 6

Omega-3 PUFAs improve the function and morphology of adipose tissues of PCOS in a gut microbiota–dependent manner. A Representative images of subcutaneous adipose tissues from control, PCOS, omega-3, and omega-3 + ABX mice. Scale bars: 50 μm. B-E Thermogenic markers in subcutaneous adipose tissues. F-I Thermogenic markers in brown adipose tissues. Error bars are expressed as the means ± SEMs. Statistical significance was determined by P values determined by one-way ANOVA followed by Dunnett’s multiple comparisons test (*P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001)

Fig. 7
figure 7

Transferring feces from omega-3 PUFA-treated mice to PCOS mice changed the function and morphology of adipose tissues. A Representative images of subcutaneous adipose tissues from t-PCOS and t-Omega-3 mice. Scale bars: 50 μm. B-E Thermogenic markers in subcutaneous adipose tissues. F-I Thermogenic markers in brown adipose tissues. Error bars are expressed as the means ± SEMs. Statistical significance was determined by P values determined by two-tailed paired-samples t test (*P < 0.05)

Omega-3 PUFAs ameliorate ovarian dysfunction associated with PCOS and likely involve direct effects on inhibiting ovarian inflammation

The possible mechanisms by which omega-3 regulates ovarian function in PCOS were investigated. Inflammation and oxidative damage play an essential role in the pathogenesis of PCOS [43]. We next investigated whether omega-3 plays an anti-inflammatory role in the ovaries of PCOS mice. Supplementation with omega-3 PUFAs significantly reversed the enhanced expression of IL-1b, TNF-a and IL-18 mRNAs in PCOS mouse ovaries (Fig. 8A-C). However, the mRNA levels of CCL2 did not increase in PCOS mice (Fig. 8D). There was no difference between the omega-3 and omega-3 + ABX groups. Together, RT‒qPCR analyses confirmed that t-omega-3 mice showed decreased expression of Il1b and Ccl2 (Fig. 8E, H). The mRNA levels of TNF-α did not change between t-PCOS and t-Omega-3 groups (Fig. 8F). There was a trend toward decreased Il18 (Fig. 8G), but this difference was not statistically significant. Moreover, we investigated some antioxidants, such as Ho1, Nqo1, Sod3 and Cat. However, they did not change (Fig. 8I-L). Transferring feces from omega-3 PUFA-treated mice to PCOS mice only increased Ho1 expression in PCOS mouse ovaries, but other antioxidants did not change significantly (Fig. 8M-P). Our data indicated that the mechanisms underlying omega-3 PUFA-regulated improvements in the ovarian dysfunction associated with PCOS likely involve direct effects on the ovary to inhibit inflammation.

Fig. 8
figure 8

Omega-3 PUFAs ameliorate ovarian dysfunction associated with PCOS, likely involving direct effects on inhibiting ovarian inflammation. A-D The mRNA levels of IL-1β, TNF-α, IL-18 and CCL2 in the control, PCOS, omega-3 and omega-3 + ABX groups. E-H The mRNA levels of IL-1β, TNF-α, IL-18 and CCL2 in the t-PCOS and t-Omega-3 groups. Data are shown as the mean ± SEM. Statistical significance was determined by P values determined by one-way ANOVA followed by Dunnett’s multiple comparisons test and a two-tailed Mann–Whitney U test (*P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001)

Discussion

Although PCOS is a serious health concern worldwide, the limited treatment options primarily focus on the symptoms and therefore yield unsatisfactory therapeutic outcomes, and long-term metabolic complications such as obesity, type 2 diabetes mellitus (T2DM), and cardiovascular disease (CVD) exert a far-reaching influence on their quality of life [44]. Therefore, there remains an urgent need to develop effective therapies. Studies have shown that gut dysbiosis is characteristic of PCOS patients and rodent models. The manipulation of gut microbiota by FMT, probiotics, prebiotics or symbiotics ameliorates the PCOS phenotype [9]. FMT is a treatment that involves administration of a minimally manipulated microbial community from the stool of a healthy donor into the patient’s intestinal tract [45], which is a method to directly change the recipient’s gut microbiota to normalize the composition and gain a therapeutic benefit. Probiotics (e.g., Lactobacillus, Bifidobacterium, and Streptococcus) are live beneficial bacteria that, when ingested, may confer a health benefit on the host [46]. Prebiotics (e.g., lactulose and fructooligosaccharides, mainly inulin) are “nondigestible food ingredients that beneficially affect the host by selectively stimulating the growth and/or activity of one or a limited number of beneficial bacteria in the colon, and thus improve host health” [47]. Symbiotics are the combination of prebiotics and probiotics. It has been suggested that prebiotics, probiotics, and symbiotics could be a potential therapy for gastrointestinal disorders because of their beneficial effects on modifying the gut microflora [48]. As an anti-inflammatory and microbiota-regulating supplement, the potential effect of omega-3 PUFAs on PCOS is worth investigating. First, we conducted a detailed phenotypic assessment of PCOS and omega-3 PUFA-treated mice, and we found that omega-3 PUFA supplementation could ameliorate endocrine disorders, ovarian dysfunction and insulin resistance. Then, we showed that omega-3 PUFAs could alleviate gut dysbiosis in PCOS mice. The mechanisms underlying omega-3 benefits likely involve direct effects on the ovary to inhibit inflammation. Furthermore, the mechanism underlying omega-3 alleviation of IR involving the gut microbiota improved adipose tissue function and morphology by decreasing thermogenic markers and multilocular cells within the subcutaneous adipose tissues in DHEA-induced PCOS mice.

Omega-3 PUFAs are powerful antioxidative stress and anti-inflammatory agents. In addition, omega-3 PUFAs play a major role in male and female reproduction. Emerging evidence suggests that inflammation and oxidative stress contribute greatly to PCOS pathophysiology. In the present study, we observed that omega-3 PUFAs ameliorated estrous cycle disorders and ovarian morphology disruption and decreased serum testosterone and fasting blood glucose levels in DHEA-induced mice, which suggests that omega-3 PUFAs could improve ovarian dysfunction and insulin resistance in PCOS mice.

The gut microbiota is considered to be a full-fledged endocrine organ and plays an important role in the female reproductive endocrine system [49]. Growing evidence has demonstrated that the gut microbiota and its metabolites are closely associated with the occurrence and development of PCOS. Qiao X. et al. reported that Bacteroides vulgatus was markedly elevated in the gut of individuals with PCOS, and transplantation of fecal microbiota from women with PCOS or B. vulgatus-colonized recipient mice resulted in increased disruption of ovarian functions and infertility [9]. Liu et al. found a decrease in Ruminococcaceae in women with PCOS, which was consistent with our research [30]. α-diversity refers to the taxonomic diversity of bacteria present within a single sample [50]. α-diversity measures diversity within communities, and β-diversity measures diversity between communities. Here, we found that omega-3 PUFA treatment has an obvious effect on the α-diversity but not β-diversity of the gut microbiota in PCOS mice. In addition, omega-3 PUFA treatment displayed a decrease in the relative abundance of Alloprevotella and an increase in the relative abundance of Akkermansia and Alistipes. In general, these studies raised the possibility that Akkermansia might be important for the establishment of beneficial intestinal microbiota and could be developed into a probiotic therapy. However, whether Akkermansia could protect against ovary dysfunction was not determined in our study. It will be better to further replenish Akkermansia by oral gavage to accurately assess their effect. The gut microbiota affects host metabolism by interacting with host signaling pathways; therefore, further studies are required to elucidate the precise mechanism by which omega-3 PUFAs affect metabolism in PCOS mice. To our knowledge, the results of the current study suggest for the first time that omega-3 PUFAs can protect PCOS mice by improving white adipose tissue browning and are associated with the gut microbiota. Numerous studies have demonstrated that a chronic low-grade degree of inflammation plays a critical role in the development of PCOS. In our study, we indicated that omega-3 PUFA supplementation alleviated ovarian inflammation by suppressing the mRNA expression of IL-1b, TNF-a and IL-18 in PCOS mouse ovaries. MCP-1 (monocyte chemoattractant protein-1), also known as chemokine (CC-motif) ligand 2 (CCL2), is from the family of CC chemokines [51]. PCOS females have significantly increased levels of MCP-1 [14], and clinical studies have shown that supplementation with omega-3 PUFAs is beneficial in decreasing MCP-1 expression in macrophages and the levels of endothelial chemokines [52, 53]. However, the mRNA levels of CCL2 did not increase in PCOS mice in our study. Numerous studies have demonstrated that the protective effect of omega-3 PUFAs is associated with their ability to scavenge oxygen free radicals and reduce oxidative stress in target organs [54]. Interestingly, we observed no effect of omega-3 PUFA administration on the levels of Ho1, Nqo1, Sod3 and Cat in the ovaries of DHEA-treated mice. Therefore, the protective effect of omega-3 PUFAs on DHEA-induced ovarian dysfunction was not attributed to reduced oxidative stress in ovaries. Our data indicate that the mechanisms underlying omega-3 PUFA-regulated improvements in the ovarian dysfunction associated with PCOS likely involve mainly direct effects on the ovary to inhibit inflammation. Ovarian granulosa cells (GCs) are critical in shaping follicular development. Evidence has also shown that the apoptosis rates of ovarian GCs in antral follicles in PCOS are significantly increased compared with those in healthy controls [55]. The follicles are comprised of three essential cells: theca cells, granulosa cells, and the oocyte. Together, these cells interact in a synergistic manner to secrete sex steroids and protein hormones that contribute to oocyte maturation for successful fertilization [56]. Gonadotropin-releasing hormone (GnRH) is released in a pulsatile fashion from the hypothalamus, inducing the release of the gonadotropins FSH and LH from the anterior pituitary gland. FSH acts directly on granulosa cells of the developing ovarian follicle to stimulate growth of the follicle, secretion of estrogen and expression of the LH receptor (LHCGR) [57,58,59]. LH stimulates androgen production by the theca cells of developing follicles and triggers ovulation when follicular maturation is complete [60]. During the ovulatory process, LH causes the final maturation and release of the ovum from preovulatory follicles and stimulates the differentiation of the theca and granulosa cells of the preovulatory follicle into the small and large steroidogenic cells of the corpus luteum, respectively [61, 62]. Defects in angiogenesis of the corpus luteum are at the origin of PCOS. In PCOS ovulatory cycles, infertility could result from a dysfunctional corpus luteum [63]. Recent research has focused on the function of the corpus luteum, theca cells and granulosa cells, which play a key role in ovarian androgen production and conversion. The specific cell molecular mechanisms underlying omega-3 PUFA-mediated improvement of PCOS ovarian functions remain to be further explored.

The findings that androgens can induce PCOS phenotypes and gut microbiota disturbance in rodents [64, 65] were validated in DHEA-induced PCOS-like mice. Clinical studies found that the gut microbial composition and relative abundance of taxa were related to PCOS phenotypes [66]. Zarrinpar et al. found that depletion of the microbiome by antibiotics reduced FBG levels and the GTT glucose surge in normal mice [67]. Han et al. found that the blood glucose curves showed similarly high levels in the DHEA and DHEA + antibiotics groups, suggesting that hyperandrogenism leads to glucose intolerance independent of the gut microbiome [44]. The clearance of gut microbiota did not avert polycystic morphology induced by DHEA, suggesting that the ovarian anomalies resulting from androgen administration were independent of the gut microbiota, which was consistent with our research. Here, we found that omega-3 PUFAs could alleviate gut dysbiosis in PCOS mice. When the gut microbiota was cleared with antibiotics, most of the protective effects of omega-3 PUFAs on ovarian dysfunction remained, and FMT showed few protective effects of colonized microflora on PCOS. These findings indicate that although omega-3 PUFAs can alter gut microbiota dysbiosis in PCOS mice, omega-3 PUFAs protect against endocrine disorders and ovarian dysfunctions in a gut microbiota-independent manner. However, the gut microbiota plays a key role in improving white adipose tissue browning.

Conclusion

In conclusion, we found that omega-3 PUFA supplementation could ameliorate endocrine disorders, ovarian dysfunction and insulin resistance in PCOS mice. Omega-3 PUFAs could alleviate gut dysbiosis in PCOS mice. The mechanisms underlying omega-3 benefits likely involve direct effects on inhibiting ovarian inflammation. On the other hand, the mechanism underlying omega-3 alleviation of IR involving the gut microbiota improved adipose tissue function and morphology by decreasing thermogenic markers and multilocular cells within the subcutaneous adipose tissues in DHEA-induced PCOS mice.

Availability of data and materials

The datasets supporting the conclusions of this article are included within the additional files.

Abbreviations

PCOS:

Polycystic ovary syndrome

PUFA:

Polyunsaturated fatty acid

IR:

Insulin resistance

DHEA:

Dehydroepiandrosterone

16S rDNA:

16S ribosomal DNA

GCs:

Granulosa cells

FMT:

Fecal microbiota transplantation

SCFAs:

Short-chain fatty acids

FSH:

Follicle-stimulating hormone

LH:

Luteinizing hormone

E2:

Estradiol 2

T:

Testosterone

GTT:

Glucose tolerance test

ITT:

Insulin tolerance test

PCA:

Principal component analysis

UPGMA:

Unweighted pair group method using arithmetic average

TNF-α:

Tumor necrosis factor-α

IL-1β:

Interleukin-1 beta

References

  1. Norman RJ, Dewailly D, Legro RS, Hickey TE. Polycystic ovary syndrome. Lancet. 2007;370(9588):685–97.

    Article  CAS  PubMed  Google Scholar 

  2. Teede HJ, Misso ML, Costello MF, Dokras A, Laven J, Moran L, Piltonen T, Norman RJ. Recommendations from the international evidence-based guideline for the assessment and management of polycystic ovary syndrome. Hum Reprod. 2018;33(9):1602–18.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Azziz R, Carmina E, Chen Z, Dunaif A, Laven JS, Legro RS, Lizneva D, Natterson-Horowtiz B, Teede HJ, Yildiz BO. Polycystic ovary syndrome. Nat Rev Dis Primers. 2016;2:16057.

    Article  PubMed  Google Scholar 

  4. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Baptiste CG, Battista MC, Trottier A, Baillargeon JP. Insulin and hyperandrogenism in women with polycystic ovary syndrome. J Steroid Biochem Mol Biol. 2010;122(1–3):42–52.

    Article  CAS  PubMed  Google Scholar 

  6. Dokras A. Mood and anxiety disorders in women with PCOS. Steroids. 2012;77(4):338–41.

    Article  CAS  PubMed  Google Scholar 

  7. Dumesic DA, Lobo RA. Cancer risk and PCOS. Steroids. 2013;78(8):782–5.

    Article  CAS  PubMed  Google Scholar 

  8. Grigoryan OR, Zhemaite NS, Volevodz NN, Andreeva EN, Melnichenko GA, Dedov II. Long-term consequences of polycystic ovary syndrome. Ter Arkh. 2017;89(10):75–9.

    CAS  PubMed  Google Scholar 

  9. Qi X, Yun C, Sun L, Xia J, Wu Q, Wang Y, Wang L, Zhang Y, Liang X, Wang L, et al. Gut microbiota-bile acid-interleukin-22 axis orchestrates polycystic ovary syndrome. Nat Med. 2019;25(8):1225–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Balen A. The pathophysiology of polycystic ovary syndrome: trying to understand PCOS and its endocrinology. Best Pract Res Clin Obstet Gynaecol. 2004;18(5):685–706.

    Article  PubMed  Google Scholar 

  11. Ehrmann DA. Polycystic ovary syndrome. N Engl J Med. 2005;352(12):1223–36.

    Article  CAS  PubMed  Google Scholar 

  12. Goodarzi MO, Dumesic DA, Chazenbalk G, Azziz R. Polycystic ovary syndrome: etiology, pathogenesis and diagnosis. Nat Rev Endocrinol. 2011;7(4):219–31.

    Article  CAS  PubMed  Google Scholar 

  13. Zhang X, Zhang C, Shen S, Xia Y, Yi L, Gao Q, Wang Y. Dehydroepiandrosterone induces ovarian and uterine hyperfibrosis in female rats. Hum Reprod. 2013;28(11):3074–85.

    Article  CAS  PubMed  Google Scholar 

  14. Liu M, Gao J, Zhang Y, Li P, Wang H, Ren X, Li C. Serum levels of TSP-1, NF-κB and TGF-β1 in polycystic ovarian syndrome (PCOS) patients in northern China suggest PCOS is associated with chronic inflammation. Clin Endocrinol (Oxf). 2015;83(6):913–22.

    Article  CAS  PubMed  Google Scholar 

  15. Escobar-Morreale HF. Polycystic ovary syndrome: definition, aetiology, diagnosis and treatment. Nat Rev Endocrinol. 2018;14(5):270–84.

    Article  PubMed  Google Scholar 

  16. Faghfoori Z, Fazelian S, Shadnoush M, Goodarzi R. Nutritional management in women with polycystic ovary syndrome: a review study. Diabetes Metab Syndr. 2017;11(Suppl 1):429-s432.

    Article  Google Scholar 

  17. Szczuko M, Malarczyk I, Zapałowska-Chwyć M. Improvement in anthropometric parameters after rational dietary intervention in women with polycystic ovary syndrom as the best method to support treatment. Rocz Panstw Zakl Hig. 2017;68(4):409–17.

    CAS  PubMed  Google Scholar 

  18. Che X, Chen Z, Liu M, Mo Z. Dietary interventions: a promising treatment for polycystic ovary syndrome. Ann Nutr Metab. 2021;77(6):313–23.

    Article  CAS  PubMed  Google Scholar 

  19. Çıtar Dazıroğlu ME, Acar Tek N. The effect on inflammation of adherence to the Mediterranean Diet in Polycystic Ovary Syndrome. Curr Nutr Rep. 2023;12(1):191–202.

    Article  PubMed  Google Scholar 

  20. Osibogun O, Ogunmoroti O, Michos ED. Polycystic ovary syndrome and cardiometabolic risk: opportunities for cardiovascular disease prevention. Trends Cardiovasc Med. 2020;30(7):399–404.

    Article  PubMed  Google Scholar 

  21. Phelan N, O’Connor A, Kyaw Tun T, Correia N, Boran G, Roche HM, Gibney J. Hormonal and metabolic effects of polyunsaturated fatty acids in young women with polycystic ovary syndrome: results from a cross-sectional analysis and a randomized, placebo-controlled, crossover trial. Am J Clin Nutr. 2011;93(3):652–62.

    Article  CAS  PubMed  Google Scholar 

  22. Santos HO, Price JC, Bueno AA. Beyond Fish Oil supplementation: the Effects of Alternative Plant sources of Omega-3 polyunsaturated fatty acids upon lipid indexes and cardiometabolic Biomarkers-An overview. Nutrients. 2020;12(10):3159.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Oner G, Muderris II. Efficacy of omega-3 in the treatment of polycystic ovary syndrome. J Obstet Gynaecol. 2013;33(3):289–91.

    Article  CAS  PubMed  Google Scholar 

  24. Kabir M, Skurnik G, Naour N, Pechtner V, Meugnier E, Rome S, Quignard-Boulangé A, Vidal H, Slama G, Clément K, et al. Treatment for 2 mo with n 3 polyunsaturated fatty acids reduces adiposity and some atherogenic factors but does not improve insulin sensitivity in women with type 2 diabetes: a randomized controlled study. Am J Clin Nutr. 2007;86(6):1670–9.

    Article  CAS  PubMed  Google Scholar 

  25. Wathes DC, Abayasekara DR, Aitken RJ. Polyunsaturated fatty acids in male and female reproduction. Biol Reprod. 2007;77(2):190–201.

    Article  CAS  PubMed  Google Scholar 

  26. Franasiak JM, Scott RT Jr. Introduction: microbiome in human reproduction. Fertil Steril. 2015;104(6):1341–3.

    Article  PubMed  Google Scholar 

  27. Xie F, Anderson CL, Timme KR, Kurz SG, Fernando SC, Wood JR. Obesity-dependent increases in oocyte mRNAs are associated with increases in proinflammatory signaling and gut microbial abundance of Lachnospiraceae in female mice. Endocrinology. 2016;157(4):1630–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Pelzer ES, Allan JA, Waterhouse MA, Ross T, Beagley KW, Knox CL. Microorganisms within human follicular fluid: effects on IVF. PLoS One. 2013;8(3): e59062.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Lindheim L, Bashir M, Münzker J, Trummer C, Zachhuber V, Leber B, Horvath A, Pieber TR, Gorkiewicz G, Stadlbauer V, et al. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Liu R, Zhang C, Shi Y, Zhang F, Li L, Wang X, Ling Y, Fu H, Dong W, Shen J, et al. Dysbiosis of Gut Microbiota Associated with Clinical Parameters in Polycystic Ovary Syndrome. Front Microbiol. 2017;8:324.

    PubMed  PubMed Central  Google Scholar 

  31. 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):1502–11.

    Article  PubMed  PubMed Central  Google Scholar 

  32. He FF, Li YM. Role of gut microbiota in the development of insulin resistance and the mechanism underlying polycystic ovary syndrome: a review. J Ovarian Res. 2020;13(1):73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Thackray VG. Sex, microbes, and polycystic ovary syndrome. Trends Endocrinol Metab. 2019;30(1):54–65.

    Article  CAS  PubMed  Google Scholar 

  34. Wang T, Sha L, Li Y, Zhu L, Wang Z, Li K, Lu H, Bao T, Guo L, Zhang X, et al. Dietary α-Linolenic acid-rich Flaxseed Oil exerts Beneficial Effects on polycystic ovary syndrome through sex steroid hormones-microbiota-inflammation Axis in rats. Front Endocrinol (Lausanne). 2020;11:284.

    Article  PubMed  Google Scholar 

  35. Costantini L, Molinari R, Farinon B, Merendino N. Impact of omega-3 fatty acids on the gut microbiota. Int J Mol Sci. 2017;18(12):2645.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Watson H, Mitra S, Croden FC, Taylor M, Wood HM, Perry SL, Spencer JA, Quirke P, Toogood GJ, Lawton CL, et al. A randomised trial of the effect of omega-3 polyunsaturated fatty acid supplements on the human intestinal microbiota. Gut. 2018;67(11):1974–83.

    Article  CAS  PubMed  Google Scholar 

  37. van Houten EL, Visser JA. Mouse models to study polycystic ovary syndrome: a possible link between metabolism and ovarian function? Reprod Biol. 2014;14(1):32–43.

    Article  PubMed  Google Scholar 

  38. McLean AC, Valenzuela N, Fai S, Bennett SA. Performing vaginal lavage, crystal violet staining, and vaginal cytological evaluation for mouse estrous cycle staging identification. J Vis Exp. 2012;(67):e4389.

  39. Zeng SL, Li SZ, Xiao PT, Cai YY, Chu C, Chen BZ, Li P, Li J, Liu EH. Citrus polymethoxyflavones attenuate metabolic syndrome by regulating gut microbiome and amino acid metabolism. Sci Adv. 2020;6(1): eaax6208.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Brestoff JR, Artis D. Immune regulation of metabolic homeostasis in health and disease. Cell. 2015;161(1):146–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Bartelt A, Heeren J. Adipose tissue browning and metabolic health. Nat Rev Endocrinol. 2014;10(1):24–36.

    Article  CAS  PubMed  Google Scholar 

  42. Yuan X, Hu T, Zhao H, Huang Y, Ye R, Lin J, Zhang C, Zhang H, Wei G, Zhou H, et al. Brown adipose tissue transplantation ameliorates polycystic ovary syndrome. Proc Natl Acad Sci U S A. 2016;113(10):2708–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Zhao Y, Zhang C, Huang Y, Yu Y, Li R, Li M, Liu N, Liu P, Qiao J. Up-regulated expression of WNT5a increases inflammation and oxidative stress via PI3K/AKT/NF-κB signaling in the granulosa cells of PCOS patients. J Clin Endocrinol Metab. 2015;100(1):201–11.

    Article  CAS  PubMed  Google Scholar 

  44. Han Q, Wang J, Li W, Chen ZJ, Du Y. Androgen-induced gut dysbiosis disrupts glucolipid metabolism and endocrinal functions in polycystic ovary syndrome. Microbiome. 2021;9(1):101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Khoruts A, Sadowsky MJ. Understanding the mechanisms of faecal microbiota transplantation. Nat Rev Gastroenterol Hepatol. 2016;13(9):508–16.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Dhiman RK. Gut microbiota and hepatic encephalopathy. Metab Brain Dis. 2013;28(2):321–6.

    Article  CAS  PubMed  Google Scholar 

  47. Chow J. Probiotics and prebiotics: a brief overview. J Ren Nutr. 2002;12(2):76–86.

    Article  PubMed  Google Scholar 

  48. Prasad S, Dhiman RK, Duseja A, Chawla YK, Sharma A, Agarwal R. Lactulose improves cognitive functions and health-related quality of life in patients with cirrhosis who have minimal hepatic encephalopathy. Hepatology. 2007;45(3):549–59.

    Article  PubMed  Google Scholar 

  49. Qi X, Yun C, Pang Y, Qiao J. The impact of the gut microbiota on the reproductive and metabolic endocrine system. Gut Microbes. 2021;13(1):1–21.

    Article  PubMed  Google Scholar 

  50. Jiménez RR, Sommer S. The amphibian microbiome: natural range of variation, pathogenic dysbiosis, and role in conservation. Biodivers Conserv. 2017;26(4):763–86.

    Article  Google Scholar 

  51. Singh S, Anshita D, Ravichandiran V. MCP-1: function, regulation, and involvement in disease. Int Immunopharmacol. 2021;101(Pt B):107598.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Hung AM, Booker C, Ellis CD, Siew ED, Graves AJ, Shintani A, Abumrad NN, Himmelfarb J, Ikizler TA. Omega-3 fatty acids inhibit the up-regulation of endothelial chemokines in maintenance hemodialysis patients. Nephrol Dial Transplant. 2015;30(2):266–74.

    Article  CAS  PubMed  Google Scholar 

  53. Spencer M, Finlin BS, Unal R, Zhu B, Morris AJ, Shipp LR, Lee J, Walton RG, Adu A, Erfani R, et al. Omega-3 fatty acids reduce adipose tissue macrophages in human subjects with insulin resistance. Diabetes. 2013;62(5):1709–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Ge Z, Wang C, Zhang J, Li X, Hu J. Tempol protects against acetaminophen Induced Acute Hepatotoxicity by inhibiting oxidative stress and apoptosis. Front Physiol. 2019;10: 660.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Wang D, Weng Y, Zhang Y, Wang R, Wang T, Zhou J, Shen S, Wang H, Wang Y. Exposure to hyperandrogen drives ovarian dysfunction and fibrosis by activating the NLRP3 inflammasome in mice. Sci Total Environ. 2020;745: 141049.

    Article  CAS  PubMed  Google Scholar 

  56. Przygrodzka E, Plewes MR, Davis JS. Luteinizing Hormone Regulation of Inter-Organelle Communication and Fate of the Corpus Luteum. Int J Mol Sci. 2021;22(18):9972.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Casarini L, Crépieux P. Molecular Mechanisms of Action of FSH. Front Endocrinol (Lausanne). 2019;10:305.

    Article  PubMed  Google Scholar 

  58. Gutiérrez CG, Campbell BK, Webb R. Development of a long-term bovine granulosa cell culture system: induction and maintenance of estradiol production, response to follicle-stimulating hormone, and morphological characteristics. Biol Reprod. 1997;56(3):608–16.

    Article  PubMed  Google Scholar 

  59. Riccetti L, Sperduti S, Lazzaretti C, Casarini L, Simoni M. The cAMP/PKA pathway: steroidogenesis of the antral follicular stage. Minerva Ginecol. 2018;70(5):516–24.

    Article  PubMed  Google Scholar 

  60. Abedel-Majed MA, Romereim SM, Davis JS, Cupp AS. Perturbations in lineage specification of Granulosa and Theca cells may alter Corpus Luteum formation and function. Front Endocrinol (Lausanne). 2019;10:832.

    Article  PubMed  Google Scholar 

  61. Meidan R, Girsh E, Blum O, Aberdam E. In vitro differentiation of bovine theca and granulosa cells into small and large luteal-like cells: morphological and functional characteristics. Biol Reprod. 1990;43(6):913–21.

    Article  CAS  PubMed  Google Scholar 

  62. Sanders SL, Stouffer RL. Localization of steroidogenic enzymes in macaque luteal tissue during the menstrual cycle and simulated early pregnancy: immunohistochemical evidence supporting the two-cell model for estrogen production in the primate corpus luteum. Biol Reprod. 1997;56(5):1077–87.

    Article  CAS  PubMed  Google Scholar 

  63. Boutzios G, Karalaki M, Zapanti E. Common pathophysiological mechanisms involved in luteal phase deficiency and polycystic ovary syndrome. Impact on fertility. Endocrine. 2013;43(2):314–7.

    Article  CAS  PubMed  Google Scholar 

  64. Zhang F, Ma T, Cui P, Tamadon A, He S, Huo C, Yierfulati G, Xu X, Hu W, Li X, et al. Diversity of the gut microbiota in Dihydrotestosterone-Induced PCOS rats and the Pharmacologic Effects of Diane-35, Probiotics, and Berberine. Front Microbiol. 2019;10: 175.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. 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):400–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Jobira B, Frank DN, Pyle L, Silveira LJ, Kelsey MM, Garcia-Reyes Y, Robertson CE, Ir D, Nadeau KJ, Cree-Green M. Obese adolescents with PCOS have altered biodiversity and relative abundance in gastrointestinal microbiota. J Clin Endocrinol Metab. 2020;105(6):e2134-2144.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Zarrinpar A, Chaix A, Xu ZZ, Chang MW, Marotz CA, Saghatelian A, Knight R, Panda S. Antibiotic-induced microbiome depletion alters metabolic homeostasis by affecting gut signaling and colonic metabolism. Nat Commun. 2018;9(1):2872.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the other members of Prof. Yao’s laboratory for their discussion and insights. We are grateful to Guangzhou Genedenovo Biotechnology Co., Ltd. for assisting in sequencing and/or bioinformatics analysis.

Funding

This work was supported by the National Natural Science Foundation of China (grants 82274651, 81973965, 82271687, 32000583 and 81904043). Jiangsu Provincial Key Research and Development Program (grant BE2022712) and the Key Medical Research Projects of Jiangsu Provincial Health Commission (grant ZD2022004).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: BY, LC, RJM; Methodology: HZ, LZ, CWL, JJ; Investigation: HZ, LZ; Data Analysis: HZ, LZ, CWL; Visualization: HZ; Supervision: BY, LC, RJM; Writing – original draft: HZ. All authors reviewed the manuscript.

Corresponding authors

Correspondence to Rujun Ma, Li Chen or Bing Yao.

Ethics declarations

Ethics approval and consent to participate

Animal subjects: All animal protocols were approved by the Animal Care and Use Committee of Jinling Hospital.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, H., Zheng, L., Li, C. et al. Effects of gut microbiota on omega-3-mediated ovary and metabolic benefits in polycystic ovary syndrome mice. J Ovarian Res 16, 138 (2023). https://doi.org/10.1186/s13048-023-01227-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13048-023-01227-w

Keywords