Abstract
Global fertility rates continue to decline despite advancements in assisted reproductive technologies, highlighting a significant gap in our understanding of the mechanisms underlying preconception physiology. In this commentary, we review a growing body of work demonstrating that the microbiome plays a crucial yet underexplored role in women’s reproductive health. This work has shown that microbial communities produce substrates that support metabolic, immune, and hormonal functions during this critical period, affecting fertility, pregnancy outcomes, and offspring health. Women with reproductive disorders, including endometriosis, polycystic ovarian syndrome, primary ovarian insufficiency, and recurrent pregnancy loss, harbor distinct microbial signatures. Animal studies provide key mechanistic insights, showing that disruption of microbiota accelerates ovarian aging, but translating these findings to human preconception health requires careful consideration. While these findings are compelling, this emerging field currently lacks a clear understanding of how microbial signals affect reproductive tissues through metabolites, immune responses, or hormonal pathways. We outline criteria for establishing microbial causation in preconception health, including sufficiency, necessity, specificity, and timing. Moving beyond correlations involves selecting appropriate models, focusing on key developmental windows, conducting longitudinal studies before conception, and investigating how specific microbial metabolites influence reproductive outcomes. Incorporating microbiome research into preconception care could lead to the development of new therapies and interventions. While the principles outlined here primarily address preconception reproductive health, they also offer a framework for microbiome research in general, emphasizing the need for a mechanistic understanding, timely interventions, and progress from association to causation in this rapidly evolving field.
Background
Despite significant technological progress in reproductive medicine over recent decades, fecundity rates continue to decline worldwide [1–7]. We can now manipulate reproduction at the cellular and molecular levels in ways that were previously impossible, including freezing oocytes for decades [8, 9], genetically screening embryos for chromosomal abnormalities [10, 11], and even editing disease-causing variants using CRISPR technology [12–14]. These innovations have transformed the available menu of fertility treatments for patients, but they have not addressed the primary determinant of female fertility: the ovarian reserve [15, 16]. This finite pool of primordial follicles is established before birth and represents a woman’s total reproductive potential [17–19]. Throughout life, primordial follicles are continuously recruited, activated, and depleted, a process that is inevitable but varies significantly between individuals [20, 21]. Environmental factors, such as diet, toxicant exposure, antibiotics, and stress, can influence the rate of follicle depletion, but our understanding of which factors are essential and how they exert their effects remains limited [22–25]. The consequences of this incomplete understanding are clear in clinical trials focusing on obesity-related fertility complications, which show a puzzling paradox: weight-loss interventions improve cardiovascular and metabolic health but, in most cases, fail to improve fertility outcomes and may even increase the risk of adverse pregnancy outcomes [26–30].
This gap between systemic and reproductive health suggests that we are still missing key factors that connect environmental influences on ovarian biology. We propose that the microbiome is one such missing link. The microbiota—referring to the trillions of microorganisms living in and on our bodies—produces tens of thousands of bioactive metabolites that regulate essential aspects of host physiology, effectively representing the critical link between diet, metabolism, immunity, and health outcomes [31–37]. While these microbial communities are influenced by age, genetics, environment, and lifestyle factors, diet stands out as the most direct and modifiable factor shaping their composition, function, and activity [38–43]. Within days, dietary changes can alter microbial community structure and metabolite production, particularly short-chain fatty acids (SCFAs) [41, 42, 44]. This rapid responsiveness has profound implications: Western nutritional patterns that are high in fat and ultra-processed foods but low in fiber disrupt the intestinal microbiota, reducing SCFA production and triggering intestinal permeability and low-grade inflammation even before weight gain occurs [43, 45–49]. These microbiome-mediated effects may help explain why lifestyle interventions focused on caloric restriction, rather than diet composition and quality, often fail to improve fertility outcomes despite improving metabolic health [28, 29, 50, 51].
Given its sensitivity to external inputs and its systemic reach, the gut microbiome is uniquely positioned to influence reproductive physiology. Distinct gut microbial signatures characterize women with reproductive disorders, including primary ovarian insufficiency (POI) [52], polycystic ovarian syndrome (PCOS) [53, 54], decreased ovarian reserve [55], endometriosis [56], and early menopause [57, 58], who exhibit alterations in gut microbiota composition and function. Such gut microbial imbalances (dysbiosis) are also associated with infertility [59, 60], poor responses to assisted reproductive technologies [55], recurrent implantation failure [59], and adverse pregnancy outcomes [61]. Animal studies have demonstrated that the gut microbiota and its metabolites influence both the quantity and quality of oocytes [62–65], as well as the modulation of cisplatin-induced POI to help preserve fertility [66]. While cervicovaginal and endometrial microbiota undoubtedly influence reproductive outcomes and have been extensively reviewed elsewhere [67–71], this commentary focuses on the gut microbiome, which has received less attention despite its potential for systemic effects through dietary and nutritional strategies [35, 39, 41, 72–74]. This expanding body of evidence suggests that the gut microbiome represents a mechanistic link connecting environmental influences on reproductive health.
Thus, in this commentary, we review existing evidence linking gut microbial communities to reproductive outcomes, propose mechanisms of microbial dependence, and set standards for establishing causality in this emerging field. We also emphasize the importance of timing, context, and model selection in evaluating host-microbiota interactions during the preconception period and their impact on fertility, as well as how to apply basic research to translate these findings into clinical practice. We propose that integrating microbiome science into reproductive medicine presents a unique opportunity to reconceptualize fertility not just as an isolated endocrine process but as one intricately embedded within a broader ecological system. As the field advances, a unified framework for research will be crucial to unlock these possibilities and bridge the gap between systemic and reproductive health. The same experimental challenges and conceptual questions arise in other fields of microbiome research, including metabolism [75], immunity [42, 76], neuroscience and neuropsychiatry [77–81], and oncology [82–85]. By framing sufficiency, necessity, specificity, and timing within the context of reproductive biology, we aim to provide a transferable blueprint that can guide discovery across different organ systems.
The microbiota and reproduction: from initial observations to associations
Researchers first discovered a link between gut microbiota and reproduction when they observed that germ-free mice had smaller litter sizes [86]. This phenotype was reversed by accidental bacterial contamination of the isolators, where germ-free mice are typically maintained under axenic conditions [86, 87]. Subsequent studies in multiple species, including flies, mosquitoes, nematodes, and honeybees, have shown that the loss of commensal intestinal bacteria prevents the onset of sexual maturity [88–95]. These processes are reversible with the reintroduction of commensal bacteria. Almost half a century after the initial observation in germ-free mice, our group discovered that germ-free female mice also exhibit hallmarks of accelerated reproductive aging, including depletion of the primordial follicle pool, which constitutes the ovarian reserve, excessive collagen buildup, and a shortened reproductive lifespan, ultimately leading to secondary infertility [64]. Although germ-free mice are born with normal numbers of primordial follicles, primordial follicle loss begins to occur rapidly during the weaning transition, a conserved period of maturation during which the microbiota shifts from milk-associated to solid food-associated communities [96]. This weaning transition represents a crucial window of rapid expansion in the diversity of microbiota and microbial genes, which are necessary for producing metabolites that have lasting consequences for epigenetic programming, immunity, metabolism, and health outcomes [97–101]. Together with reports on microbiota functions in other developmental processes, such as sexual maturity and aging [102–104], these findings suggest that microbiota play previously underexplored roles in regulating the timing of key life stages in their hosts, with significant implications for ovarian biology and reproductive medicine.
They also raise an important question: how do environmental factors influence the crosstalk between the intestinal microbiota and ovarian tissue? One key example is Western dietary patterns, which disrupt the gut microbiota by altering microbial composition and reducing the production of key metabolites, including SCFAs. These microbial changes occur before obesity-induced ovarian dysfunction, including oocyte lipid accumulation, mitochondrial impairment, oxidative stress, disrupted meiosis, decreased fertilization rates, subfertility, poorer embryo quality, and weaker responses to superovulation during in vitro fertilization [105, 106]. Diet-induced changes in the microbiota can occur even without obvious obesity or insulin resistance [45, 49, 107, 108]. This work further highlights the limitations of body mass index, which is widely used to determine eligibility for fertility treatment and to guide clinical decisions [109, 110], as a marker of reproductive health. For instance, metabolically healthy individuals with obesity (as defined by BMI) might have normal reproductive function, while those with a normal BMI may experience metabolic dysfunction that affects fertility [111, 112]. It also suggests that dietary modifications that affect the microbiota and its metabolites, rather than just lowering calorie intake, may be crucial for improving oocyte quality and fertility. Intriguingly, colonizing germ-free mice with intestinal microbiota during the weaning transition is sufficient to rescue the premature ovarian aging phenotype we have observed [64], as is treatment with microbial-derived SCFAs alone [64], pointing to a direct, metabolite-mediated pathway through which the intestinal microbiota can influence ovarian longevity, independent of systemic metabolic status.
Antibiotic exposure is another significant disruptor of the microbiota with effects on reproductive health outcomes. Broad-spectrum antibiotics have a potent impact on gut microbiota, resulting in decreased microbial diversity, reduced SCFA availability, and dysbiosis with damaging effects on host immune and metabolic processes [113]. Some human studies have linked preconception exposure to antibiotics with increased interaction effects with oral contraceptives, risk of infertility, miscarriage, and congenital anomalies [113–115]. However, some caution is necessary when interpreting these results, as (chronic) antibiotic use may be indicated for unrelated comorbid health conditions that can have unintended reproductive consequences.
Connecting microbial-derived signals to reproductive tissues: exploring mechanisms
Having established that microbiota influences female reproductive outcomes, the mechanistic pathways that may link intestinal microbiota to ovarian function warrant examination. The influence of microbes on immune populations in tissues, such as the intestinal tract, is well-documented, and the microbiota and its metabolites may also shape the ovarian tissue immune compartment. Once touted as an immune-privileged site [116], recent single-cell analyses revealed that the ovary maintains a dynamic immune environment that includes infiltrating and tissue-resident macrophages, monocytes, neutrophils, dendritic cells, CD4⁺ and CD8⁺ T cells, γδ T cells, mucosal-associated invariant T cells (MAIT), innate lymphoid cells (ILCs), natural killer (NK) cells, and B cells [117–126]. These populations are part of a somatic cellular network that includes granulosa and stromal cells, which control follicle development, ovulation, and luteal remodeling [127–132]. This immune landscape also changes with age, as the number of inflammatory cells accumulates in the ovaries and the surrounding adipose tissue [117, 133, 134]. These changes are thought to contribute to follicular dysfunction and eventual age-related ovarian fibrosis. It is important to note that work in other tissues has shown that the microbiota influences these immune populations in some capacity, including IL-17-producing effector T cells and regulatory T cells [135–138], as well as ILCs, NK cells, and MAIT cells [139–141]. However, whether the ovarian immune compartment similarly depends on microbial signals remains unexplored, representing a fundamental gap in our understanding of ovarian-microbiome crosstalk.
Germ-free mice show disrupted follicle quiescence gene expression before significant follicle loss occurs [64], suggesting that microbial signals influence transcriptional networks that regulate follicle quiescence, activation, and atresia [64]. Transcriptomic analyses reveal that genes related to SCFA transporter activity are altered in germ-free ovaries [38]. This suggests that the ovary may directly sense and respond to SCFAs, but further validation is required. SCFAs act as key mediators of host-microbe communication [40, 142], primarily influencing gene expression through two mechanisms: (1) binding to G protein-coupled receptors (GPR41, GPR43, GPR109A) to initiate signaling pathways that regulate cellular metabolism, inflammation, and hormone secretion, and (2) inhibition of class I and IIa histone deacetylases (HDACs), which increases chromatin accessibility [143–146]. Stable isotope tracing has confirmed that microbe-derived substrates are incorporated into histone tails, demonstrating direct epigenetic modification [147–153]. In immune contexts, microbial-derived SCFAs promote the growth and activity of regulatory T cells through similar mechanisms [113–115]. While SCFAs promote cell cycle arrest and quiescence through histone deacetylase inhibition in other tissues [102–104], whether this specific pathway operates in the ovary to maintain follicle quiescence remains to be determined. This represents a critical knowledge gap in understanding how microbial metabolites contribute to preserving ovarian reserve.
While transcriptional and immune mechanisms await confirmation in ovarian tissue, the microbial regulation of reproductive hormones offers more established links. One of the best-known roles of the intestinal microbiota is the recycling and recirculation of estrogens. Specific bacterial species, such as Bacteroides thetaiotaomicron, Phocaeicola vulgatus, and Clostridium perfringens, produce β-glucuronidase enzymes that deconjugate estrogens in the gut, allowing for their reabsorption and subsequent return to the systemic circulation [154, 155]. This “estrobolome” influences circulating estrogen levels and has been linked to conditions like PCOS, endometriosis, and early menopause [156, 157]. Elevated fecal β-glucuronidase activity correlates with higher serum estradiol and testosterone levels in women with PCOS [158]. Commensal microbes also impact neuroactive steroid levels, including the synthesis and metabolism of progesterone derivatives, such as allopregnanolone [159, 160]. Recent research has identified Eggerthella lenta and Gordonibacter pamelaeae as microbial producers of allopregnanolone from glucocorticoids via a hydrogen-dependent 21-dehydroxylation pathway, demonstrating that commensal bacteria can produce bioactive progestins in vivo [159, 160]. Stable isotope tracing and infusion studies demonstrate that microbial-derived SCFAs build up in regions of the hypothalamus responsible for regulating fertility, reproduction, and pregnancy [161]. Whether these metabolites play a causal role in regulating the hypothalamic circuits that control fertility and reproduction remains to be investigated [162].
While we have focused on the intestinal microbiota and its impact on ovarian function, it is essential to recognize that other microbial niches also play crucial roles in reproductive health. The cervicovaginal and endometrial microbiota have a significant impact on fertility and pregnancy outcomes [163–165]. Reproductive niches dominated by Lactobacillus, particularly L. crispatus, have been associated with higher rates of embryo implantation and live births after in vitro fertilization. In contrast, women with Lactobacillus-depleted or anaerobe-rich communities have increased rates of poor IVF response, failed embryo transfer, and miscarriage [67, 166–170]. A systematic review and meta-analysis of 25 studies confirmed this pattern, showing an 18% lower pregnancy rate and a 50% higher risk of early pregnancy loss in women harboring a non-optimal cervicovaginal microbiota [171]. However, results on live births were inconsistent [171]. In the first multicenter randomized placebo-controlled IVF trial, women with non-optimal cervicovaginal microbiota dominated by Gardnerella and Fannyhessea received 7 days of oral clindamycin followed by intravaginal L. crispatus CTV 05 (LACTIN V), clindamycin alone, or double placebo [172]. None of the treatment arms showed improvement in any IVF endpoint compared with the placebo, including hormone levels, implantation rates, pregnancy rates, live birth rates, perinatal outcomes, or safety indices [172]. The investigators noted that approximately one-quarter of participants had already shifted to Lactobacillus dominance before randomization, and more than one-third of the placebo group exhibited a spontaneous transition toward Lactobacillus dominance by the time of embryo transfer [172]. Thus, the authors recommend that future trials enroll women with persistent non-optimal communities at the time of transfer and analyze both vaginal and endometrial niches to test mechanism-based therapies [172].
Emerging evidence also suggests that the paternal microbiome may influence fertility and offspring health by affecting sperm quality and epigenetic programming [173–176], highlighting another critical area for future research. These diverse microbial influences underscore the need for comprehensive approaches to understanding microbiome–reproduction interactions.
Benchmarks for studying microbiota on reproductive outcomes: establishing standards
As interest in the role of the microbiota in reproductive outcomes continues to grow, this emerging field must develop the criteria needed to establish microbial causation. These standards are already in place in many other fields [177–183], and we draw inspiration from them to create a framework specifically designed for reproductive biology (sufficiency, necessity, specificity, timing, and mechanism). This framework is particularly crucial as clinical tests used in infertility care without proper validation can lead to misdiagnosis, unnecessary interventions, and increased emotional and financial burden for patients.
First, model selection influences interpretations. Germ-free mice are a powerful model for demonstrating the effects of microbiota; however, they exhibit inherent reproductive deficits, including accelerated follicle loss and a shorter reproductive lifespan [38]. Using these models without proper controls risks circular reasoning. For example, transplanting microbiota from patients with reproductive pathologies like POI or PCOS into germ-free mice may produce disease-like traits. We might interpret this as evidence that the microbiota causes the reproductive disorder. However, this reasoning is circular, as reproductive phenotypes in germ-free mice could arise from two sources: (1) the baseline reproductive deficits inherent to the germ-free state itself, or (2) the specific effects of the transplanted microbiota. Without proper controls, we cannot distinguish between these possibilities. For instance, if both the microbiota of healthy donors and PCOS patients exhibit similar reproductive deficits when transplanted into germ-free mice, this would suggest that the issue lies with the germ-free model rather than the disease-associated microbiota. This distinction is key: showing microbiota influences a particular endpoint or phenotype is different from proving they cause disease. Rigorous experimental design includes multiple controls, such as conventionally raised mice and gnotobiotic mice that are colonized with known, defined microbial communities, as well as detailed baseline phenotyping to differentiate host susceptibility from microbial causation.
Second, timing is crucial in depletion and reconstitution experiments. Microbial signals May have different effects before versus after conception or during critical windows of reproductive, immune, or endocrine development. Antibiotic-based depletion studies, which investigate the role of the microbiome by disrupting or reducing microbial populations with broad-spectrum antibiotics, can help determine microbial necessity and sufficiency. Recolonization methods help establish sufficiency, but both must consider the unique timing of female reproductive transitions and microbial signaling. For example, mouse strains exhibit differences in the rate of follicle depletion, which is further compounded by age; pubertal mice Maintain a larger ovarian reserve than 6-month-old mice that have undergone repeated ovulations and significant follicle loss. Initiating antibiotic depletion in older mice thus involves assessing the effects of microbes on an already diminished reserve, potentially missing critical preservation mechanisms that operate earlier in life (e.g., developmental programming of follicle survival pathways, early immune-ovarian crosstalk). Defined consortia, such as the altered Schaedler flora [184] or Oligo-Mouse Microbiota [185], provide tractable systems. These simplified, well-characterized microbial communities enable researchers to manipulate microbial composition with precision, reduce biological variability, and more easily link specific microbial signals to host outcomes. This approach could be beneficial for teasing out complex interactions in reproductive biology, where both host physiology and microbial ecology undergo changes over time. Regardless of the approach used—whether germ-free, antibiotic-depleted, or gnotobiotic models—reproductive outcomes must be measured explicitly and rigorously. Variables such as estrous cyclicity, hormone levels, follicle counts, ovulation patterns, and oocyte quality provide critical context for interpreting the effects of the microbiome on fertility. Without directly assessing these endpoints, studies risk overgeneralizing or missing reproductive-specific phenotypes, limiting both mechanistic insight and translational potential.
Third, human studies must focus on timing and context as important a priori factors. Most microbiome research involving humans is retrospective or cross-sectional and usually includes patients who are already diagnosed. These approaches limit the ability to draw causal conclusions and make it hard to determine whether changes are causes or effects. We advise against overinterpreting fecal microbiota transplantation (FMT) from clinical donors into germ-free mice as evidence of causality. While FMT can reveal whether a disease-associated microbiota can transfer specific phenotypes to a recipient, it cannot disentangle the contributions of underlying host factors, comorbidities, or environmental exposures that may have shaped the donor microbiome in the first place. Moreover, germ-free mice are not immunologically or hormonally identical to humans, and their responses to human-derived microbiota may reflect exaggerated or artificial effects. Without appropriate controls and contextual understanding, these models risk reinforcing associations rather than revealing actual mechanisms. To advance the field, we advocate for longitudinal, prospective studies that track microbiome changes in individuals before the onset of reproductive dysfunction. Populations at elevated risk, such as daughters of women with reproductive disorders, adolescent girls with menstrual irregularities, or patients pursuing fertility preservation for non-infertility indications, offer unique windows for observing how microbial shifts might precede or predict reproductive decline. These designs are better positioned to identify early microbial signatures of disease, uncover modifiable risk factors, and lay the groundwork for preventive interventions. Crucially, embedding microbiome assessments into fertility care pathways and life course studies may help reframe reproductive aging as not only hormonally driven but also ecologically shaped.
Fourth, studies must link microbial signatures to functional outputs and their target cells in reproductive tissues. Relying solely on microbial relative abundance offers a limited understanding of the mechanisms involved. Instead, functional outputs of the microbiota are more likely to mediate direct effects on reproductive physiology. Integrated multi-omic methods, including transcriptomics, metabolomics, and in vitro assays, can connect microbial signals to specific cellular responses. Stable isotope tracing provides an additional layer by directly tracking microbial metabolites in ovarian tissues and mapping their incorporation into host metabolic or epigenetic pathways. We propose that novel insights into mechanisms will arise when specific microbial products are associated with changes in inflammation, endocrine signaling, mitochondrial function, or fibrosis within relevant ovarian cell types, such as oocytes, granulosa cells, stromal cells, or immune populations. Such granularity is necessary for identifying which microbial signals interact with specific host targets to influence reproductive outcomes.
Modulating the human microbiome to improve reproductive outcomes
Dietary interventions hold great promise For improving fertility and reproductive outcomes. Recent work has shown that a plant-based diet, which involves doubling fiber intake and eliminating ultra-processed Foods, leads to significant changes in gut microbiota within 4 days, characterized by fewer pro-inflammatory taxa and increased SCFA production compared to a calorie-matched control diet [186]. These shifts are accompanied by clinical improvements, including weight loss, a 17% reduction in LDL cholesterol, a 14% decrease in inflammatory markers, and improved glucose control [186]. These effects are eliminated within weeks of participants returning to their pre-intervention dietary habits [186], suggesting that continued compliance is likely required to sustain microbiota-driven improvements in reproductive and metabolic health.
In contrast, although calorie-restricted diets can improve weight and metabolic markers, patients often start by reducing their intake of carbohydrate-rich foods. This inadvertently reduces dietary fiber because it eliminates sources of microbiota-accessible carbohydrates, which in turn decreases the production of beneficial microbial metabolites [187–189]. Food processing, regardless of caloric content, promotes metabolic dysfunction, with ultra-processed diets leading to weight gain compared to unprocessed diets, even when the calorie content is matched [190]. Emphasizing high-protein intake during caloric restriction can also cause colonic fermentation to shift from saccharolytic to proteolytic processes, resulting in the overproduction of metabolites that are detrimental to colonic health, such as phenolic compounds and N-nitroso compounds [191]. Relying on ultra-processed “diet” foods and artificial sweeteners during calorie restriction may further disrupt the microbiome [192, 193]. These findings reinforce an important concept that “a calorie is not a calorie” when it comes to its biological effects [50, 194], suggesting that food quality and processing may influence reproductive outcomes through microbiome-mediated mechanisms. Additionally, the impact of dietary approaches may vary depending on the reproductive outcome being targeted, the timing of the intervention, and the specific population studied.
Indeed, it is essential to distinguish between interventions that aim to preserve oocyte quantity (i.e., the ovarian reserve) and those designed to improve or maintain oocyte quality, as these rely on distinct biological processes and require different timing strategies. Animal studies have identified early-life windows, such as the weaning period and the onset of puberty, as critical periods when the pool of primordial follicles appears to be especially sensitive to microbial and dietary cues. While the exact timing may differ in humans, analogous critical windows likely include early childhood dietary transitions (ages 2–5) [96, 97, 195–197], the pubertal period [198, 199], and adolescence [200], representing periods when both the microbiome and reproductive system undergo significant developmental changes. Since this follicle pool is non-renewable once depleted, early interventions may be key for preserving long-term ovarian reserve. In contrast, oocyte quality may remain modifiable beyond these early developmental windows. Folliculogenesis and the maturation of high-quality oocytes require sustained metabolic and hormonal support, which occurs over several months [201]. In contrast, the microbiome can shift within days in response to nutritional changes. Therefore, interventions with microbiota-directed complementary foods might need to be sustained over extended periods to support the full duration of the follicular maturation process. Filling critical knowledge gaps—such as identifying the key microbial metabolites and their mechanisms of action—will be essential to optimizing the design, timing, and personalization of these interventions.
A call to integrate microbiome science into reproductive medicine
The time is now to integrate microbiome science into reproductive medicine. We propose three immediate priorities for the field: First, establish rigorous standards for causality using our proposed framework to move beyond correlative studies. Second, develop longitudinal cohort studies that monitor microbiome-reproduction interactions, with a focus on key developmental periods. Third, establish interdisciplinary research networks that integrate expertise in microbiology, reproductive biology, nutrition, immunology, and clinical medicine to accelerate the translation of findings.
As evidence accumulates, several practical steps can help bridge the gap between research and clinical care. Fertility specialists can collect more detailed dietary and antibiotic histories during consultations, with a particular focus on fiber intake and other nutritional factors known to influence the microbiota. Clinicians may also advise patients planning to conceive to adopt microbiota-directed complementary diets several months in advance, with the caveat that while such strategies are biologically plausible and low-risk, they have not yet been validated through clinical trials to improve fertility outcomes. To advance from current knowledge to validated interventions, we recommend focusing on (1) prospective cohort studies tracking individuals from preconception through pregnancy; (2) randomized controlled trials testing microbiome-targeted dietary interventions on reproductive outcomes; and (3) development of validated biomarkers linking microbial function to oocyte quality and fertility potential. Achieving these goals will require strong interdisciplinary collaboration among reproductive endocrinologists, microbiome scientists, nutrition researchers, immunologists, and systems biologists to translate emerging insights into actionable clinical protocols.
Conclusions
Ultimately, the microbiome offers a powerful, systems-level lens through which to understand and improve reproductive health by linking diet, immunity, metabolism, and hormonal signaling. Going forward, targeted investment in basic and translational research, the development of standardized tools and protocols, and the training of a new generation of interdisciplinary scientists will be essential to realizing this potential. We anticipate that the rigor and mechanistic focus advocated in this commentary will accelerate the transition from correlation to causation, not only in reproductive medicine but also in cardiovascular disease, metabolic disorders, neurologic conditions, cancer, and other fields exploring how microbial communities and their metabolites impact health and disease.
Acknowledgements
EJ acknowledges funding by the National Institutes of Health grant K01DK1121734 (EJ), National Institutes of Health grant P50HD096723 (EJ), Burroughs Wellcome Fund Next Gen Pregnancy Initiative Award (EJ), Magee Auxiliary Research Scholars Award (EJ), MWRI Internal Pilot Award (EJ).
Authors’ contributions
SKM and EJ both wrote the initial draft, and NV, JMR, and AK contributed to writing the final commentary. The authors read and approved the final manuscript.
Funding
National Institute of Diabetes and Digestive and Kidney Diseases,K01DK1121734,Burroughs Wellcome Fund,Eunice Kennedy Shriver National Institute of Child Health and Human Development,P50HD096723
Data availability
No datasets were generated or analysed during the current study.
Declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- 1.Inhorn MC, Patrizio P. Infertility around the globe: new thinking on gender, reproductive technologies and global movements in the 21st century. Hum Reprod Update. 2015;21:411–26. [DOI] [PubMed] [Google Scholar]
- 2.Levine H, Jørgensen N, Martino-Andrade A, Mendiola J, Weksler-Derri D, Jolles M, et al. Temporal trends in sperm count: a systematic review and meta-regression analysis of samples collected globally in the 20th and 21st centuries. Hum Reprod Update. 2023;29:157–76. [DOI] [PubMed] [Google Scholar]
- 3.Borumandnia N, Alavi Majd H, Khadembashi N, Alaii H. Worldwide trend analysis of primary and secondary infertility rates over past decades: A cross-sectional study. Int J Reprod Biomed. 2022;20:37–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Aitken RJ. The Global Decline in Human Fertility: The Post-Transition Trap Hypothesis. Life (Basel). 2024;14:369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Aitken RJ. What is driving the global decline of human fertility? Need for a multidisciplinary approach to the underlying mechanisms. Front Reprod Health . 2024 [cited 2025 Jun 25];6. Available from: 10.3389/frph.2024.1364352/full [DOI] [PMC free article] [PubMed]
- 6.Bhattacharjee NV, Schumacher AE, Aali A, Abate YH, Abbasgholizadeh R, Abbasian M, et al. Global fertility in 204 countries and territories, 1950–2021, with forecasts to 2100: a comprehensive demographic analysis for the Global Burden of Disease Study 2021. The Lancet. 2024;403:2057–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Fauser BCJM, Adamson GD, Boivin J, Chambers GM, de Geyter C, Dyer S, et al. Declining global fertility rates and the implications for family planning and family building: an IFFS consensus document based on a narrative review of the literature. Hum Reprod Update. 2024;30:153–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Stigliani S, Moretti S, Anserini P, Casciano I, Venturini PL, Scaruffi P. Storage time does not modify the gene expression profile of cryopreserved human metaphase II oocytes. Hum Reprod. 2015;30:2519–26. [DOI] [PubMed] [Google Scholar]
- 9.Cobo A, García-Velasco JA, Coello A, Domingo J, Pellicer A, Remohí J. Oocyte vitrification as an efficient option for elective fertility preservation. Fertil Steril. 2016;105:755-764.e8. [DOI] [PubMed] [Google Scholar]
- 10.Viotti M. Preimplantation Genetic Testing for Chromosomal Abnormalities: Aneuploidy, Mosaicism, and Structural Rearrangements. Genes (Basel). 2020;11:602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Munné S, Kaplan B, Frattarelli JL, Child T, Nakhuda G, Shamma FN, et al. Preimplantation genetic testing for aneuploidy versus morphology as selection criteria for single frozen-thawed embryo transfer in good-prognosis patients: a multicenter randomized clinical trial. Fertil Steril. 2019;112:1071-1079.e7. [DOI] [PubMed] [Google Scholar]
- 12.Reddy P, Ocampo A, Suzuki K, Luo J, Bacman SR, Williams SL, et al. Selective elimination of mitochondrial mutations in the germline by genome editing. Cell. 2015;161:459–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Adashi EY, Cohen IG. Heritable Human Genome Editing: The International Commission Report. JAMA. 2020;324:1941–2. [DOI] [PubMed] [Google Scholar]
- 14.Ma H, Marti-Gutierrez N, Park S-W, Wu J, Lee Y, Suzuki K, et al. Correction of a pathogenic gene mutation in human embryos. Nature. 2017;548:413–9. [DOI] [PubMed] [Google Scholar]
- 15.Sunkara SK, Rittenberg V, Raine-Fenning N, Bhattacharya S, Zamora J, Coomarasamy A. Association between the number of eggs and live birth in IVF treatment: an analysis of 400 135 treatment cycles. Hum Reprod. 2011;26:1768–74. [DOI] [PubMed] [Google Scholar]
- 16.Practice Committee of the American Society for Reproductive Medicine. Electronic address: asrm@asrm.org, Practice Committee of the American Society for Reproductive Medicine. Testing and interpreting measures of ovarian reserve: a committee opinion. Fertil Steril. 2020;114:1151–7. [DOI] [PubMed]
- 17.Kallen A, Polotsky AJ, Johnson J. Untapped Reserves: Controlling Primordial Follicle Growth Activation. Trends Mol Med. 2018;24:319–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Telfer EE, Grosbois J, Odey YL, Rosario R, Anderson RA. Making a good egg: human oocyte health, aging, and in vitro development. Physiol Rev. 2023;103:2623–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Findlay JK, Hutt KJ, Hickey M, Anderson RA. How Is the Number of Primordial Follicles in the Ovarian Reserve Established?1. Biol Reprod. 2015;93(111):1–7. [DOI] [PubMed] [Google Scholar]
- 20.McGee EA, Hsueh AJ. Initial and cyclic recruitment of ovarian follicles. Endocr Rev. 2000;21:200–14. [DOI] [PubMed] [Google Scholar]
- 21.Nelson SM, Telfer EE, Anderson RA. The ageing ovary and uterus: new biological insights. Hum Reprod Update. 2013;19:67–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Armstrong DT. Environmental stress and ovarian function. Biol Reprod. 1986;34:29–39. [DOI] [PubMed] [Google Scholar]
- 23.Silvestris E, de Pergola G, Rosania R, Loverro G. Obesity as disruptor of the female fertility. Reprod Biol Endocrinol. 2018;16:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Silvestris E, Lovero D, Palmirotta R. Nutrition and Female Fertility: An Interdependent Correlation. Front Endocrinol (Lausanne). 2019;10:346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Richardson MC, Guo M, Fauser BCJM, Macklon NS. Environmental and developmental origins of ovarian reserve. Hum Reprod Update. 2014;20:353–69. [DOI] [PubMed] [Google Scholar]
- 26.Best D, Avenell A, Bhattacharya S. How effective are weight-loss interventions for improving fertility in women and men who are overweight or obese? A systematic review and meta-analysis of the evidence. Hum Reprod Update. 2017;23:681–705. [DOI] [PubMed] [Google Scholar]
- 27.Gaskins AJ, Chavarro JE. Diet and fertility: a review. Am J Obstet Gynecol. 2018;218:379–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Legro RS, Hansen KR, Diamond MP, Steiner AZ, Coutifaris C, Cedars MI, et al. Effects of preconception lifestyle intervention in infertile women with obesity: The FIT-PLESE randomized controlled trial. PLoS Med. 2022;19: e1003883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Mutsaerts MAQ, van Oers AM, Groen H, Burggraaff JM, Kuchenbecker WKH, Perquin DAM, et al. Randomized Trial of a Lifestyle Program in Obese Infertile Women. N Engl J Med. 2016;374:1942–53. [DOI] [PubMed] [Google Scholar]
- 30.Carson SA, Kallen AN. Diagnosis and Management of Infertility: A Review. JAMA. 2021;326:65–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol. 2009;9:313–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hooper LV, Gordon JI. Commensal Host-Bacterial Relationships in the Gut. Science. 2001;292:1115–8. [DOI] [PubMed] [Google Scholar]
- 33.Hooper LV, Midtvedt T, Gordon JI. How Host-Microbial Interactions Shape the Nutrient Environment of the Mammalian Intestine. Annu Rev Nutr. 2002;22:283–307. [DOI] [PubMed] [Google Scholar]
- 34.Sender R, Fuchs S, Milo R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016;14: e1002533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Bishai JD, Palm NW. Small Molecule Metabolites at the Host-Microbiota Interface. J Immunol. 2021;207:1725–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Donia MS, Fischbach MA. Small Molecules from the Human Microbiota. Science. 2015;349:1254766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zierer J, Jackson MA, Kastenmüller G, Mangino M, Long T, Telenti A, et al. The fecal metabolome as a functional readout of the gut microbiome. Nat Genet. 2018;50:790–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Zmora N, Suez J, Elinav E. You are what you eat: diet, health and the gut microbiota. Nat Rev Gastroenterol Hepatol. 2019;16:35–56. [DOI] [PubMed] [Google Scholar]
- 40.Koh A, De Vadder F, Kovatcheva-Datchary P, Bäckhed F. From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites. Cell. 2016;165:1332–45. [DOI] [PubMed] [Google Scholar]
- 41.Alexander M, Turnbaugh PJ. Deconstructing Mechanisms of Diet-Microbiome-Immune Interactions. Immunity. 2020;53:264–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell. 2014;157:121–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Albenberg LG, Wu GD. Diet and the intestinal microbiome: associations, functions, and implications for health and disease. Gastroenterology. 2014;146:1564–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Koh TJ, DiPietro LA. Inflammation and wound healing: the role of the macrophage. Expert Rev Mol Med. 2011;13: e23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Hildebrandt MA, Hoffmann C, Sherrill-Mix SA, Keilbaugh SA, Hamady M, Chen Y, et al. High-Fat Diet Determines the Composition of the Murine Gut Microbiome Independently of Obesity. Gastroenterology. 2009;137:1716-1724.e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kim K-A, Gu W, Lee I-A, Joh E-H, Kim D-H. High fat diet-induced gut microbiota exacerbates inflammation and obesity in mice via the TLR4 signaling pathway. PLoS ONE. 2012;7: e47713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, et al. Metabolic Endotoxemia Initiates Obesity and Insulin Resistance. Diabetes. 2007;56:1761–72. [DOI] [PubMed] [Google Scholar]
- 48.Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet–induced obesity and diabetes in mice. Diabetes. 2008;57:1470–81. [DOI] [PubMed] [Google Scholar]
- 49.Dalby MJ, Ross AW, Walker AW, Morgan PJ. Dietary Uncoupling of Gut Microbiota and Energy Harvesting from Obesity and Glucose Tolerance in Mice. Cell Rep. 2017;21:1521–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Sonnenburg JL, Bäckhed F. Diet-microbiota interactions as moderators of human metabolism. Nature. 2016;535:56–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Zeevi D, Korem T, Zmora N, Israeli D, Rothschild D, Weinberger A, et al. Personalized Nutrition by Prediction of Glycemic Responses. Cell. 2015;163:1079–94. [DOI] [PubMed] [Google Scholar]
- 52.Wang J, Luo R, Zhao X, Xia D, Liu Y, Shen T, et al. Association between gut microbiota and primary ovarian insufficiency: a bidirectional two-sample Mendelian randomization study. Front Endocrinol. 2023 [cited 2025 Mar 18];14. Available from: h 10.3389/fendo.2023.1183219/full [DOI] [PMC free article] [PubMed]
- 53.Torres PJ, Siakowska M, Banaszewska B, Pawelczyk L, Duleba AJ, Kelley ST, et al. Gut Microbial Diversity in Women With Polycystic Ovary Syndrome Correlates With Hyperandrogenism. J Clin Endocrinol Metab. 2018;103:1502–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Yurtdaş G, Akdevelioğlu Y. A New Approach to Polycystic Ovary Syndrome: The Gut Microbiota. J Am Coll Nutr. 2020;39:371–82. [DOI] [PubMed] [Google Scholar]
- 55.Fo X, Pei M, Liu P, Zhu F, Zhang Y, Mu X. Metagenomic analysis revealed the association between gut microbiota and different ovary responses to controlled ovarian stimulation. Sci Rep. 2024;14:14930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Ata B, Yildiz S, Turkgeldi E, Brocal VP, Dinleyici EC, Moya A, et al. The Endobiota Study: Comparison of Vaginal, Cervical and Gut Microbiota Between Women with Stage 3/4 Endometriosis and Healthy Controls. Sci Rep. 2019;9:2204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Nieto MR, Rus MJ, Areal-Quecuty V, Lubián-López DM, Simon-Soro A. Menopausal shift on women’s health and microbial niches. npj Womens Health. 2025;3:3.
- 58.Peters BA, Santoro N, Kaplan RC, Qi Q. Spotlight on the Gut Microbiome in Menopause: Current Insights. Int J Womens Health. 2022;14:1059–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Patel N, Patel N, Pal S, Nathani N, Pandit R, Patel M, et al. Distinct gut and vaginal microbiota profile in women with recurrent implantation failure and unexplained infertility. BMC Women’s Health. 2022;22:113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Chopra C, Kumar V, Kumar M, Bhushan I. Role of vaginal microbiota in idiopathic infertility: a prospective study. Microbes Infect. 2024;26: 105308. [DOI] [PubMed] [Google Scholar]
- 61.Koren O, Konnikova L, Brodin P, Mysorekar IU, Collado MC. The maternal gut microbiome in pregnancy: implications for the developing immune system. Nat Rev Gastroenterol Hepatol. 2024;21:35–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Kim M, Benayoun BA. The microbiome: An emerging key player in aging and longevity. Translational Medicine of Aging. 2020;4:103–16. [PMC free article] [PubMed] [Google Scholar]
- 63.Qi X, Yun C, Pang Y, Qiao J. The impact of the gut microbiota on the reproductive and metabolic endocrine system. Gut Microbes. 2021;13:1–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Munyoki SK, Goff JP, Reshke A, Wilderoter E, Mafarachisi N, Kolobaric A, et al. The microbiota extends the reproductive lifespan by safeguarding the ovarian reserve. bioRxiv; 2024 [cited 2024 Dec 17]. p. 2024.09.13.612929. Available from: https://www.biorxiv.org/content/10.1101/2024.09.13.612929v1
- 65.Ju S, Kang ZY, Yang LY, Xia YJ, Guo YM, Li S, et al. Gut microbiota and ovarian diseases: a new therapeutic perspective. Journal of Ovarian Research. 2025;18:105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Feng Y, Zheng H, Yin C, Liang D, Zhang S, Chen J, et al. β-resorcylic acid released by Limosilactobacillus reuteri protects against cisplatin-induced ovarian toxicity and infertility. CR Med. 2024 [cited 2024 Sep 21];5. Available from: https://www.cell.com/cell-reports-medicine/abstract/S2666-3791(24)00399-9 [DOI] [PMC free article] [PubMed]
- 67.Moreno I, Simon C. Deciphering the effect of reproductive tract microbiota on human reproduction. Reproductive Medicine and Biology. 2019;18:40–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Kroon SJ, Ravel J, Huston WM. Cervicovaginal microbiota, women’s health, and reproductive outcomes. Fertil Steril. 2018;110:327–36. [DOI] [PubMed] [Google Scholar]
- 69.Schoenmakers S, Laven J. The vaginal microbiome as a tool to predict IVF success. Curr Opin Obstet Gynecol. 2020;32:169–78. [DOI] [PubMed] [Google Scholar]
- 70.Toson B, Simon C, Moreno I. The Endometrial Microbiome and Its Impact on Human Conception. Int J Mol Sci. 2022;23:485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Moosa Y, Kwon D, de Oliveira T, Wong EB. Determinants of Vaginal Microbiota Composition. Front Cell Infect Microbiol. 2020;10:467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Sanz Y, Cryan JF, Deschasaux-Tanguy M, Elinav E, Lambrecht R, Veiga P. The gut microbiome connects nutrition and human health. Nat Rev Gastroenterol Hepatol. 2025;22:534–55. [DOI] [PubMed] [Google Scholar]
- 73.Chen RY, Mostafa I, Hibberd MC, Das S, Lynn HM, Webber DM, et al. Melding microbiome and nutritional science with early child development. Nat Med. 2021;27:1503–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Spivak I, Fluhr L, Elinav E. Local and systemic effects of microbiome-derived metabolites. EMBO Rep. 2022;23: e55664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Zhao L. The gut microbiota and obesity: from correlation to causality. Nat Rev Microbiol. 2013;11:639–47. [DOI] [PubMed] [Google Scholar]
- 76.Lynn DJ, Benson SC, Lynn MA, Pulendran B. Modulation of immune responses to vaccination by the microbiota: implications and potential mechanisms. Nat Rev Immunol. 2022;22:33–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Cryan JF, O’Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, et al. The Microbiota-Gut-Brain Axis. Physiol Rev. 2019;99:1877–2013. [DOI] [PubMed] [Google Scholar]
- 78.Foster JA, McVey Neufeld K-A. Gut-brain axis: how the microbiome influences anxiety and depression. Trends Neurosci. 2013;36:305–12. [DOI] [PubMed] [Google Scholar]
- 79.McGuinness AJ, Loughman A, Foster JA, Jacka F. Mood Disorders: The Gut Bacteriome and Beyond. Biol Psychiatry. 2024;95:319–28. [DOI] [PubMed] [Google Scholar]
- 80.Schneider E, O’Riordan KJ, Clarke G, Cryan JF. Feeding gut microbes to nourish the brain: unravelling the diet-microbiota-gut-brain axis. Nat Metab. 2024;6:1454–78. [DOI] [PubMed] [Google Scholar]
- 81.Yu LW, Agirman G, Hsiao EY. The Gut Microbiome as a Regulator of the Neuroimmune Landscape. Annu Rev Immunol. 2022;40:143–67. [DOI] [PubMed] [Google Scholar]
- 82.Elinav E, Garrett WS, Trinchieri G, Wargo J. The cancer microbiome. Nat Rev Cancer. 2019;19:371–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Gopalakrishnan V, Helmink BA, Spencer CN, Reuben A, Wargo JA. The Influence of the Gut Microbiome on Cancer, Immunity, and Cancer Immunotherapy. Cancer Cell. 2018;33:570–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Khan MdAW, Ologun G, Arora R, McQuade JL, Wargo JA. The gut microbiome modulates response to cancer immunotherapy. Dig Dis Sci. 2020;65:885–96. [DOI] [PMC free article] [PubMed]
- 85.Matson V, Chervin CS, Gajewski TF. Cancer and the Microbiome-Influence of the Commensal Microbiota on Cancer, Immune Responses, and Immunotherapy. Gastroenterology. 2021;160:600–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Shinoda M, Tamura H, Maejima K. Watarai S [Reproductive ability of germfree ICR female mice (author’s transl)]. Jikken Dobutsu. 1980;29:55–9. [DOI] [PubMed] [Google Scholar]
- 87.Shimizu K, Muranaka Y, Fujimura R, Ishida H, Tazume S, Shimamura T. Normalization of reproductive function in germfree mice following bacterial contamination. Exp Anim. 1998;47:151–8. [DOI] [PubMed] [Google Scholar]
- 88.Elgart M, Stern S, Salton O, Gnainsky Y, Heifetz Y, Soen Y. Impact of gut microbiota on the fly’s germ line. Nat Commun. 2016;7:11280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Gnainsky Y, Zfanya N, Elgart M, Omri E, Brandis A, Mehlman T, et al. Systemic Regulation of Host Energy and Oogenesis by Microbiome-Derived Mitochondrial Coenzymes. Cell Reports . 2021 [cited 2025 Jun 25];34. Available from: https://www.cell.com/cell-reports/abstract/S2211-1247(20)31572-2 [DOI] [PubMed]
- 90.Sharon G, Segal D, Ringo JM, Hefetz A, Zilber-Rosenberg I, Rosenberg E. Commensal bacteria play a role in mating preference of Drosophila melanogaster. Proc Natl Acad Sci. 2010;107:20051–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Dedeine F, Vavre F, Fleury F, Loppin B, Hochberg ME, Boulétreau M. Removing symbiotic Wolbachia bacteria specifically inhibits oogenesis in a parasitic wasp. Proc Natl Acad Sci. 2001;98:6247–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Coon KL, Vogel KJ, Brown MR, Strand MR. Mosquitoes rely on their gut microbiota for development. Mol Ecol. 2014;23:2727–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Cabreiro F, Gems D. Worms need microbes too: microbiota, health and aging in Caenorhabditis elegans. EMBO Mol Med. 2013;5:1300–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Zheng H, Powell JE, Steele MI, Dietrich C, Moran NA. Honeybee gut microbiota promotes host weight gain via bacterial metabolism and hormonal signaling. Proc Natl Acad Sci. 2017;114:4775–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Zumkhawala-Cook A, Gallagher P, Raymann K. Diet affects reproductive development and microbiota composition in honey bees. Animal Microbiome. 2024;6:64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Koenig JE, Spor A, Scalfone N, Fricker AD, Stombaugh J, Knight R, et al. Succession of microbial consortia in the developing infant gut microbiome. Proc Natl Acad Sci U S A. 2011;108(Suppl 1):4578–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Flores JN, Lubin J-B, Silverman MA. The case for microbial intervention at weaning. Gut Microbes. 2024;16:2414798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Al Nabhani Z, Eberl G. Imprinting of the immune system by the microbiota early in life. Mucosal Immunol. 2020;13:183–9. [DOI] [PubMed] [Google Scholar]
- 99.Al Nabhani Z, Dulauroy S, Marques R, Cousu C, Al Bounny S, Déjardin F, et al. A Weaning Reaction to Microbiota Is Required for Resistance to Immunopathologies in the Adult. Immunity. 2019;50:1276-1288.e5. [DOI] [PubMed] [Google Scholar]
- 100.Knoop KA, Gustafsson JK, McDonald KG, Kulkarni DH, Coughlin PE, McCrate S, et al. Microbial antigen encounter during a preweaning interval is critical for tolerance to gut bacteria. Sci Immunol. 2017;2:eaao1314. [DOI] [PMC free article] [PubMed]
- 101.Lubin J-B, Green J, Maddux S, Denu L, Duranova T, Lanza M, et al. Arresting microbiome development limits immune system maturation and resistance to infection in mice. Cell Host Microbe. 2023;31:554-570.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Metcalf CJE, Henry LP, Rebolleda-Gómez M, Koskella B. Why Evolve Reliance on the Microbiome for Timing of Ontogeny? mBio. 10:e01496–19. [DOI] [PMC free article] [PubMed]
- 103.Kim M, Wang J, Pilley SE, Lu RJ, Xu A, Kim Y, et al. Estropausal gut microbiota transplant improves measures of ovarian function in adult mice. bioRxiv. 2025;2024.05.03.592475.
- 104.Xu L, Zhang Q, Dou X, Wang Y, Wang J, Zhou Y, et al. Fecal microbiota transplantation from young donor mice improves ovarian function in aged mice. J Genet Genomics. 2022;49:1042–52. [DOI] [PubMed] [Google Scholar]
- 105.Wu LL, Norman RJ, Robker RL. The impact of obesity on oocytes: evidence for lipotoxicity mechanisms. Reprod Fertil Dev. 2011;24:29–34. [DOI] [PubMed] [Google Scholar]
- 106.Meulders B, Marei WFA, Loier L, Leroy JLMR. Lipotoxicity and Oocyte Quality in Mammals: Pathogenesis, Consequences, and Reversibility. Annual Review of Animal Biosciences. 2025;13:233–54. [DOI] [PubMed] [Google Scholar]
- 107.Jašarević E, Hill EM, Kane PJ, Rutt L, Gyles T, Folts L, et al. The composition of human vaginal microbiota transferred at birth affects offspring health in a mouse model. Nat Commun. 2021;12:6289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Morrison KE, Jašarević E, Howard CD, Bale TL. It’s the fiber, not the fat: significant effects of dietary challenge on the gut microbiome. Microbiome. 2020;8:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Kaye L, Sueldo C, Engmann L, Nulsen J, Benadiva C. Survey assessing obesity policies for assisted reproductive technology in the United States. Fertil Steril. 2016;105:703-706.e2. [DOI] [PubMed] [Google Scholar]
- 110.Pandey S, Maheshwari A, Bhattacharya S. Should access to fertility treatment be determined by female body mass index? Hum Reprod. 2010;25:815–20. [DOI] [PubMed] [Google Scholar]
- 111.Broughton DE, Jungheim ES. A Focused Look at Obesity and the Preimplantation Trophoblast. Semin Reprod Med. 2016;34:5–10. [DOI] [PubMed] [Google Scholar]
- 112.Schon SB, Cabre HE, Redman LM. The impact of obesity on reproductive health and metabolism in reproductive-age females. Fertil Steril. 2024;122:194–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Alemu BK, Wang CC, Li L, Zhu Z, Li Q, Wang Y. Effect of preconception antibiotics exposure on female reproductive health and pregnancy outcomes: a systematic review and meta-analysis. EClinicalMedicine. 2024;78: 102935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Dickinson BD, Altman RD, Nielsen NH, Sterling ML. Drug interactions between oral contraceptives and antibiotics. Obstet Gynecol. 2001;98:853–60. [DOI] [PubMed] [Google Scholar]
- 115.Norwitz ER, Greenberg JA. Antibiotics in Pregnancy: Are They Safe? Rev Obstet Gynecol. 2009;2:135–6. [PMC free article] [PubMed] [Google Scholar]
- 116.Chasse AY, Bandyadka S, Wertheimer MC, Serizier SB, McCall K. Professional phagocytes are recruited for the clearance of obsolete nonprofessional phagocytes in the Drosophila ovary. Front Immunol. 2024;15:1389674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Ben Yaakov T, Wasserman T, Aknin E, Savir Y. Single-cell analysis of the aged ovarian immune system reveals a shift towards adaptive immunity and attenuated cell function. Hägg S, Harper DM, editors. eLife. 2023;12:e74915. [DOI] [PMC free article] [PubMed]
- 118.Chai C, Liang L, Mikkelsen NS, Wang W, Zhao W, Sun C, et al. Single-cell transcriptome analysis of epithelial, immune, and stromal signatures and interactions in human ovarian cancer. Commun Biol. 2024;7:131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Brodeur TY, Lanser TB, Salter L, Sessions K, Hogan T, Bonney EA, et al. Single cell analysis of ovarian immune cells during homeostasis and hormonal flux reveals dynamic changes in NK and B cell populations in the periovulatory phase. bioRxiv; 2025 [cited 2025 Jun 25]. p. 2025.04.18.649580. Available from: 10.1101/2025.04.18.649580v1 [DOI]
- 120.Garcia-Flores V, Romero R, Peyvandipour A, Galaz J, Pusod E, Panaitescu B, et al. A single-cell atlas of murine reproductive tissues during preterm labor. Cell Rep. 2023;42: 111846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Isola JVV, Ocañas SR, Hubbart CR, Ko S, Mondal SA, Hense JD, et al. A single-cell atlas of the aging mouse ovary. Nat Aging. 2024;4:145–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Jones ASK, Hannum DF, Machlin JH, Tan A, Ma Q, Ulrich ND, et al. Cellular atlas of the human ovary using morphologically guided spatial transcriptomics and single-cell sequencing. Science Advances. 2024;10:eadm7506. [DOI] [PMC free article] [PubMed]
- 123.Wagner M, Yoshihara M, Douagi I, Damdimopoulos A, Panula S, Petropoulos S, et al. Single-cell analysis of human ovarian cortex identifies distinct cell populations but no oogonial stem cells. Nat Commun. 2020;11:1147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Winkler I, Tolkachov A, Lammers F, Lacour P, Daugelaite K, Schneider N, et al. The cycling and aging mouse female reproductive tract at single-cell resolution. Cell. 2024;187:981-998.e25. [DOI] [PubMed] [Google Scholar]
- 125.Fan X, Bialecka M, Moustakas I, Lam E, Torrens-Juaneda V, Borggreven NV, et al. Single-cell reconstruction of follicular remodeling in the human adult ovary. Nat Commun. 2019;10:3164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Wang S, Zheng Y, Li J, Yu Y, Zhang W, Song M, et al. Single-Cell Transcriptomic Atlas of Primate Ovarian Aging. Cell. 2020;180:585-600.e19. [DOI] [PubMed] [Google Scholar]
- 127.Bukovsky A, Caudle MR. REVIEW ARTICLE: Immune Physiology of the Mammalian Ovary – A Review. Am J Reprod Immunol. 2008;59:12–26. [DOI] [PubMed] [Google Scholar]
- 128.Robker RL, Wu LL, Yang X. Inflammatory pathways linking obesity and ovarian dysfunction. J Reprod Immunol. 2011;88:142–8. [DOI] [PubMed] [Google Scholar]
- 129.Wu R, Van der Hoek KH, Ryan NK, Norman RJ, Robker RL. Macrophage contributions to ovarian function. Hum Reprod Update. 2004;10:119–33. [DOI] [PubMed] [Google Scholar]
- 130.Richards JS, Liu Z, Shimada M. Immune-like mechanisms in ovulation. Trends Endocrinol Metab. 2008;19:191–6. [DOI] [PubMed] [Google Scholar]
- 131.Duffy DM, Ko C, Jo M, Brannstrom M, Curry TE. Ovulation: Parallels With Inflammatory Processes. Endocr Rev. 2018;40:369–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Espey LL. Ovulation as an Inflammatory Reaction—A Hypothesis. Biol Reprod. 1980;22:73–106. [DOI] [PubMed] [Google Scholar]
- 133.Zhang Z, Schlamp F, Huang L, Clark H, Brayboy L. Inflammaging is associated with shifted macrophage ontogeny and polarization in the aging mouse ovary. Reproduction. 2020;159:325–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Lliberos C, Liew SH, Mansell A, Hutt KJ. The Inflammasome Contributes to Depletion of the Ovarian Reserve During Aging in Mice. Front Cell Dev Biol. 2020;8: 628473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009;139:485–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Ivanov II, Frutos RDL, Manel N, Yoshinaga K, Rifkin DB, Sartor RB, et al. Specific Microbiota Direct the Differentiation of IL-17-Producing T-Helper Cells in the Mucosa of the Small Intestine. Cell Host Microbe. 2008;4:337–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011;331:337–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Narushima S, Sugiura Y, Oshima K, Atarashi K, Hattori M, Suematsu M, et al. Characterization of the 17 strains of regulatory T cell-inducing human-derived Clostridia. Gut Microbes. 2014;5:333–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Ganal SC, Sanos SL, Kallfass C, Oberle K, Johner C, Kirschning C, et al. Priming of natural killer cells by nonmucosal mononuclear phagocytes requires instructive signals from commensal microbiota. Immunity. 2012;37:171–86. [DOI] [PubMed] [Google Scholar]
- 140.Gury-BenAri M, Thaiss CA, Serafini N, Winter DR, Giladi A, Lara-Astiaso D, et al. The Spectrum and Regulatory Landscape of Intestinal Innate Lymphoid Cells Are Shaped by the Microbiome. Cell. 2016;166:1231-1246.e13. [DOI] [PubMed] [Google Scholar]
- 141.Constantinides MG, Link VM, Tamoutounour S, Wong AC, Perez-Chaparro PJ, Han S-J, et al. MAIT cells are imprinted by the microbiota in early life and promote tissue repair. Science. 2019;366:eaax6624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Tan J, McKenzie C, Potamitis M, Thorburn AN, Mackay CR, Macia L. The role of short-chain fatty acids in health and disease. Adv Immunol. 2014;121:91–119. [DOI] [PubMed] [Google Scholar]
- 143.Brown AJ, Goldsworthy SM, Barnes AA, Eilert MM, Tcheang L, Daniels D, et al. The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J Biol Chem. 2003;278:11312–9. [DOI] [PubMed] [Google Scholar]
- 144.Singh N, Gurav A, Sivaprakasam S, Brady E, Padia R, Shi H, et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity. 2014;40:128–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Davie JR. Inhibition of histone deacetylase activity by butyrate. J Nutr. 2003;133:2485S-2493S. [DOI] [PubMed] [Google Scholar]
- 146.Waldecker M, Kautenburger T, Daumann H, Busch C, Schrenk D. Inhibition of histone-deacetylase activity by short-chain fatty acids and some polyphenol metabolites formed in the colon. J Nutr Biochem. 2008;19:587–93. [DOI] [PubMed] [Google Scholar]
- 147.Krautkramer KA, Kreznar JH, Romano KA, Vivas EI, Barrett-Wilt GA, Rabaglia ME, et al. Diet-Microbiota Interactions Mediate Global Epigenetic Programming in Multiple Host Tissues. Mol Cell. 2016;64:982–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Fellows R, Varga-Weisz P. Chromatin dynamics and histone modifications in intestinal microbiota-host crosstalk. Molecular Metabolism. 2020;38: 100925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Fellows R, Denizot J, Stellato C, Cuomo A, Jain P, Stoyanova E, et al. Microbiota derived short chain fatty acids promote histone crotonylation in the colon through histone deacetylases. Nat Commun. 2018;9:105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Gates LA, Reis BS, Lund PJ, Paul MR, Leboeuf M, Djomo AM, et al. Histone butyrylation in the mouse intestine is mediated by the microbiota and associated with regulation of gene expression. Nat Metab. 2024;6:697–707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Lund PJ, Gates LA, Leboeuf M, Smith SA, Chau L, Lopes M, et al. Stable isotope tracing in vivo reveals a metabolic bridge linking the microbiota to host histone acetylation. Cell Rep. 2022;41: 111809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Xiao X, Zhou Y, Li X, Jin J, Durham J, Ye Z, et al. 13C-Stable isotope resolved metabolomics uncovers dynamic biochemical landscape of gut microbiome-host organ communications in mice. Microbiome. 2024;12:90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Thomas SP, Denu JM. Short-chain fatty acids activate acetyltransferase p300. Shi X, Cole PA, Khochbin S, editors. eLife. 2021;10:e72171. [DOI] [PMC free article] [PubMed]
- 154.Ervin SM, Li H, Lim L, Roberts LR, Liang X, Mani S, et al. Gut microbial β-glucuronidases reactivate estrogens as components of the estrobolome that reactivate estrogens. J Biol Chem. 2019;294:18586–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Flores R, Shi J, Fuhrman B, Xu X, Veenstra TD, Gail MH, et al. Fecal microbial determinants of fecal and systemic estrogens and estrogen metabolites: a cross-sectional study. J Transl Med. 2012;10:253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Tetel MJ, de Vries GJ, Melcangi RC, Panzica G, O’Mahony SM. Steroids, stress and the gut microbiome-brain axis. J Neuroendocrinol. 2018;30. [DOI] [PMC free article] [PubMed]
- 157.Baker JM, Al-Nakkash L, Herbst-Kralovetz MM. Estrogen-gut microbiome axis: Physiological and clinical implications. Maturitas. 2017;103:45–53. [DOI] [PubMed] [Google Scholar]
- 158.Patel J, Chaudhary H, Rajput K, Parekh B, Joshi R. Assessment of gut microbial β-glucuronidase and β-glucosidase activity in women with polycystic ovary syndrome. Sci Rep. 2023;13:11967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.McCurry MD, D’Agostino GD, Walsh JT, Bisanz JE, Zalosnik I, Dong X, et al. Gut bacteria convert glucocorticoids into progestins in the presence of hydrogen gas. Cell. 2024;187:2952-2968.e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Doden H, Sallam LA, Devendran S, Ly L, Doden G, Daniel SL, et al. Metabolism of Oxo-Bile Acids and Characterization of Recombinant 12α-Hydroxysteroid Dehydrogenases from Bile Acid 7α-Dehydroxylating Human Gut Bacteria. Appl Environ Microbiol. 2018;84:e00235-e318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Frost G, Sleeth ML, Sahuri-Arisoylu M, Lizarbe B, Cerdan S, Brody L, et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat Commun. 2014;5:3611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Qi X, Yun C, Sun L, Xia J, Wu Q, Wang Y, et al. Gut microbiota-bile acid-interleukin-22 axis orchestrates polycystic ovary syndrome. Nat Med. 2019;25:1225–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Ravel J, Gajer P, Abdo Z, Schneider GM, Koenig SSK, McCulle SL, et al. Vaginal microbiome of reproductive-age women. Proc Natl Acad Sci U S A. 2011;108(Suppl 1):4680–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Ravel J, Brotman RM, Gajer P, Ma B, Nandy M, Fadrosh DW, et al. Daily temporal dynamics of vaginal microbiota before, during and after episodes of bacterial vaginosis. Microbiome. 2013;1:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.García-Velasco JA, Menabrito M, Catalán IB. What fertility specialists should know about the vaginal microbiome: a review. Reprod Biomed Online. 2017;35:103–12. [DOI] [PubMed] [Google Scholar]
- 166.Haahr T, Jensen JS, Thomsen L, Duus L, Rygaard K, Humaidan P. Abnormal vaginal microbiota may be associated with poor reproductive outcomes: a prospective study in IVF patients. Hum Reprod. 2016;31:795–803. [DOI] [PubMed] [Google Scholar]
- 167.Koedooder R, Singer M, Schoenmakers S, Savelkoul PHM, Morré SA, de Jonge JD, et al. The vaginal microbiome as a predictor for outcome of in vitro fertilization with or without intracytoplasmic sperm injection: a prospective study. Hum Reprod. 2019;34:1042–54. [DOI] [PubMed] [Google Scholar]
- 168.Moreno I, Codoñer FM, Vilella F, Valbuena D, Martinez-Blanch JF, Jimenez-Almazán J, et al. Evidence that the endometrial microbiota has an effect on implantation success or failure. Am J Obstet Gynecol. 2016;215:684–703. [DOI] [PubMed] [Google Scholar]
- 169.Bar O, Vagios S, Barkai O, Elshirbini J, Souter I, Xu J, et al. Harnessing vaginal inflammation and microbiome: a machine learning model for predicting IVF success. NPJ Biofilms Microbiomes. 2025;11:95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Moreno I, Garcia-Grau I, Perez-Villaroya D, Gonzalez-Monfort M, Bahçeci M, Barrionuevo MJ, et al. Endometrial microbiota composition is associated with reproductive outcome in infertile patients. Microbiome. 2022;10:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Maksimovic Celicanin M, Haahr T, Humaidan P, Skafte-Holm A. Vaginal dysbiosis – the association with reproductive outcomes in IVF patients: a systematic review and meta-analysis. Curr Opin Obstet Gynecol. 2024;36:155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Haahr T, Freiesleben NL, Jensen MB, Elbaek HO, Alsbjerg B, Laursen R. Efficacy of clindamycin and LACTIN-V for in vitro fertilization patients with vaginal dysbiosis: a randomised double-blind, placebo-controlled multicentre trial. Nature Communications. 2025;16:5166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Argaw-Denboba A, Schmidt TSB, Di Giacomo M, Ranjan B, Devendran S, Mastrorilli E, et al. Paternal microbiome perturbations impact offspring fitness. Nature. 2024;629:652–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Harris JC, Trigg NA, Goshu B, Yokoyama Y, Dohnalová L, White EK, et al. The microbiota and T cells non-genetically modulate inherited phenotypes transgenerationally. Cell Reports . 2024 [cited 2025 Mar 18];43. Available from: https://www.cell.com/cell-reports/abstract/S2211-1247(24)00357-7 [DOI] [PMC free article] [PubMed]
- 175.Trigg NA, Zhou SK, Harris JC, Lamonica MN, Nelson MA, Silverman MA, et al. A lack of commensal microbiota influences the male reproductive tract intergenerationally in mice. 2025 [cited 2025 Mar 18]; Available from: https://rep.bioscientifica.com/view/journals/rep/169/4/REP-24-0204.xml [DOI] [PMC free article] [PubMed]
- 176.Al-Asmakh M, Stukenborg J-B, Reda A, Anuar F, Strand M-L, Hedin L, et al. The Gut Microbiota and Developmental Programming of the Testis in Mice. PLoS ONE. 2014;9: e103809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Walter J, Armet AM, Finlay BB, Shanahan F. Establishing or Exaggerating Causality for the Gut Microbiome: Lessons from Human Microbiota-Associated Rodents. Cell. 2020;180:221–32. [DOI] [PubMed] [Google Scholar]
- 178.Schloss PD. Identifying and Overcoming Threats to Reproducibility, Replicability, Robustness, and Generalizability in Microbiome Research. mBio. 2018;9:e00525-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Vonaesch P, Anderson M, Sansonetti PJ. Pathogens, microbiome and the host: emergence of the ecological Koch’s postulates. FEMS Microbiol Rev. 2018;42:273–92. [DOI] [PubMed] [Google Scholar]
- 180.Neville BA, Forster SC, Lawley TD. Commensal Koch’s postulates: establishing causation in human microbiota research. Curr Opin Microbiol. 2018;42:47–52. [DOI] [PubMed] [Google Scholar]
- 181.Fischbach MA. Microbiome: Focus on causation and mechanism. Cell. 2018;174:785–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Hill AB. The Environment and Disease: Association or Causation? Proc R Soc Med. 1965;58:295–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Evans AS. Causation and Disease: The Henle-Koch Postulates Revisited. Yale J Biol Med. 1976;49:175–95. [PMC free article] [PubMed] [Google Scholar]
- 184.Dewhirst FE, Chien CC, Paster BJ, Ericson RL, Orcutt RP, Schauer DB, et al. Phylogeny of the defined murine microbiota: altered Schaedler flora. Appl Environ Microbiol. 1999;65:3287–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Brugiroux S, Beutler M, Pfann C, Garzetti D, Ruscheweyh H-J, Ring D, et al. Genome-guided design of a defined mouse microbiota that confers colonization resistance against Salmonella enterica serovar Typhimurium. Nat Microbiol. 2016;2:16215. [DOI] [PubMed] [Google Scholar]
- 186.Li F, Armet AM, Korpela K, Liu J, Quevedo RM, Asnicar F, et al. Cardiometabolic benefits of a non-industrialized-type diet are linked to gut microbiome modulation. Cell. 2025;188:1226-1247.e18. [DOI] [PubMed] [Google Scholar]
- 187.Howarth NC, Saltzman E, Roberts SB. Dietary fiber and weight regulation. Nutr Rev. 2001;59:129–39. [DOI] [PubMed] [Google Scholar]
- 188.Thompson T, Dennis M, Higgins LA, Lee AR, Sharrett MK. Gluten-free diet survey: are Americans with coeliac disease consuming recommended amounts of fibre, iron, calcium and grain foods? J Hum Nutr Diet. 2005;18:163–9. [DOI] [PubMed] [Google Scholar]
- 189.Miketinas DC, Bray GA, Beyl RA, Ryan DH, Sacks FM, Champagne CM. Fiber Intake Predicts Weight Loss and Dietary Adherence in Adults Consuming Calorie-Restricted Diets: The POUNDS Lost (Preventing Overweight Using Novel Dietary Strategies) Study. J Nutr. 2019;149:1742–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Hall KD, Ayuketah A, Brychta R, Cai H, Cassimatis T, Chen KY, et al. Ultra-Processed Diets Cause Excess Calorie Intake and Weight Gain: An Inpatient Randomized Controlled Trial of Ad Libitum Food Intake. Cell Metab. 2019;30:67-77.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Russell WR, Gratz SW, Duncan SH, Holtrop G, Ince J, Scobbie L, et al. High-protein, reduced-carbohydrate weight-loss diets promote metabolite profiles likely to be detrimental to colonic health. Am J Clin Nutr. 2011;93:1062–72. [DOI] [PubMed] [Google Scholar]
- 192.Suez J, Cohen Y, Valdés-Mas R, Mor U, Dori-Bachash M, Federici S, et al. Personalized microbiome-driven effects of non-nutritive sweeteners on human glucose tolerance. Cell. 2022;185:3307-3328.e19. [DOI] [PubMed] [Google Scholar]
- 193.Suez J, Korem T, Zeevi D, Zilberman-Schapira G, Thaiss CA, Maza O, et al. Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature. 2014;514:181–6. [DOI] [PubMed] [Google Scholar]
- 194.Why Calories Don’t Count: How we got the science of weight loss wrong - Dr Giles Yeo: 9781398704329 - AbeBooks. [cited 2025 Jul 16]. Available from: https://www.abebooks.com/9781398704329/Why-Calories-Count-got-science-1398704326/plp
- 195.Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486:222–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Roswall J, Olsson LM, Kovatcheva-Datchary P, Nilsson S, Tremaroli V, Simon M-C, et al. Developmental trajectory of the healthy human gut microbiota during the first 5 years of life. Cell Host Microbe. 2021;29:765-776.e3. [DOI] [PubMed] [Google Scholar]
- 197.Stewart CJ, Ajami NJ, O’Brien JL, Hutchinson DS, Smith DP, Wong MC, et al. Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature. 2018;562:583–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Korpela K, Kallio S, Salonen A, Hero M, Kukkonen AK, Miettinen PJ, et al. Gut microbiota develop towards an adult profile in a sex-specific manner during puberty. Sci Rep. 2021;11:23297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Calcaterra V, Rossi V, Massini G, Regalbuto C, Hruby C, Panelli S, et al. Precocious puberty and microbiota: The role of the sex hormone–gut microbiome axis. Front Endocrinol. 2022 [cited 2025 Jul 16];13. Available from: 10.3389/fendo.2022.1000919/full [DOI] [PMC free article] [PubMed]
- 200.Carson MD, Westwater C, Novince CM. Adolescence and the Microbiome. Am J Pathol. 2023;193:1900–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Gougeon A. Dynamics of follicular growth in the human: a model from preliminary results. Hum Reprod. 1986;1:81–7. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No datasets were generated or analysed during the current study.
