Skip to main content

Obesity and recurrent spontaneous abortion: the crucial role of weight management in pregnancy

Abstract

Recurrent spontaneous abortion (RSA), characterized by the loss of two or more pregnancies, impacts approximately 1–2% of couples and poses a significant challenge for individuals of childbearing age. The precise mechanisms underlying RSA remain incompletely understood. Concurrently, the global prevalence of obesity is on the rise, with obesity being closely associated with female reproductive disorders and infertility. This study initially examines the pathways through which obesity contributes to RSA, encompassing factors such as embryonic euploid miscarriage, endometrial development, immune function, among others. Furthermore, adipokines and the fat mass and obesity-related (FTO) are identified as potential contributors to RSA. The study also explores the enhancement of pregnancy outcomes through various weight management strategies, with a particular focus on the roles of dietary interventions, physical activity, and weight control during pregnancy. Obesity is closely related to RSA in multiple aspects. Additional clinical prospective and experimental studies are required to explore its precise pathogenesis. Through this review, we aim to provide strategies for improvement and treatment approaches for RSA related to obesity. Through this review, we suggest potential clinical management strategies and research avenues aimed at offering enhancements and therapeutic insights for miscarriages linked to obesity and its associated risk factors.

Introduction

The American Society for Reproductive Medicine (ASRM) defines RSA as a condition characterized by the occurrence of two or more clinical pregnancies resulting in loss [1]. This condition impacts approximately 1–2% of couples [2]. Major factors contributing to RSA include chromosomal abnormalities, anatomical structural irregularities, infections, immune system dysregulation, and endocrine disorders. Nevertheless, a significant proportion of RSA cases, approximately 50%, remain of unknown origin, termed as unexplained recurrent pregnancy loss. Despite extensive scholarly research in this area, challenges persist in fully understanding the causes and management of RSA.

As societal conditions advance, there is a global increase in the prevalence of obesity. This widespread issue has become a significant concern, impacting more than 2 billion individuals worldwide and resulting in over 3 million fatalities annually [3]. Projections based on current data suggest that by 2030, nearly 40% of the global population will be overweight, with approximately one-fifth falling into the obese category [4]. Obesity is recognized as a key risk factor for various non-communicable diseases and is closely linked to conditions such as type 2 diabetes, hypertension, coronary heart disease, acute pancreatitis, premature aging, and neurodegenerative disorders [5,6,7].

In the realm of reproductive health, researchers have noted that women with overweight may encounter a range of pregnancy complications, including birth defects, miscarriages, and neonatal illnesses, which can ultimately impact fertility rates. Studies indicate that obesity have detrimental effects on fertility by impeding ovulation, menstrual regularity, and natural conception. Obesity stands as an independent risk factor for recurrent miscarriages [8], with women with obesity exhibiting a notably higher rate of miscarriage compared to their counterparts with non-obesity [9]. As Body Mass Index (BMI) rises in pregnant individuals, there is a significant decrease in live birth rates [10].

This review aims to explore the mechanisms through which obesity contributes to RSA and heightens the risk of miscarriage by examining current scientific evidence on the adverse effects of maternal obesity on RSA. While male obesity receives less attention in this context due to the gender-specific nature of RSA, efforts have been made to synthesize existing research on the influence of male obesity on RSA. Additionally, the review outlines the positive effects of weight management on pregnancy outcomes for women with obesity, underscoring the advantages of controlling weight gain during pregnancy for the well-being of both mothers and fetuses.

The mechanism of obesity leading to RSA

Focusing on early embryo development, the review delves into six key aspects through which female obesity impacts the initiation and progression of RSA: (i) obesity and embryo euploid miscarriage; (ii) obesity and oocyte quality; (iii) obesity and endometrial development; (iv) obesity and maternal immune imbalance; (v) obesity and disrupted adipokine secretion; (vi) obesity-related genes (Fig. 1).

Fig. 1
figure 1

Risk factors for obesity leading to RSA. In female obese patients, RSA may be related to six aspects: embryo euploid miscarriage, poor oocyte quality, endometrial dysplasia, maternal immune imbalance, adipokine secretion disorder, and obesity-related genetic abnormalities

Obesity and embryo euploid miscarriage

Chromosomal abnormalities in embryos are a common factor contributing to early miscarriage. Research indicates a link between obesity and a heightened occurrence of euploid miscarriages in women with RSA [11]. Tremellen et al. conducted an analysis on pregnancy outcomes post-transfer of confirmed euploid embryos in cryopreserved In Vitro Fertilization (IVF) cycles, noting a significantly elevated rate of pregnancy failures in obese individuals (41.9%) compared to lean (14.2%) and super-recombinant (29.1%) groups [12]. A recent multicenter review in 2021, involving 3,480 women categorized by weight, reported a 22.7% miscarriage rate in women with obesity, with 13.5% classified as clinical abortion [13]. Furthermore, in a separate study involving embryo transfers with known chromosomal outcomes, Among the 117 eligible patients who underwent abortions, women with obesity experienced a 58% frequency of euploid miscarriages, in contrast to 37% in women with non-obese, indicating a relative risk of 1.63 (95% confidence interval 1.08–2.47) [9].

Compared to miscarriage in euploid pregnancies, the relationship between obesity and aneuploid pregnancy is less significant. The adverse effects of being overweight or obese on IVF and reproductive outcomes may not be directly linked to aneuploidy [14]. A study that looked back at a group of patients found that without taking maternal age into account, maternal BMI was significantly linked to a higher occurrence of aneuploidy, with rates of 51.5% in the obese category and 39.3% in the non-obese category. However, once age was factored in, BMI was not a reliable predictor of aneuploidy incidence [15]. This suggests that the detrimental effects of excess weight on IVF and reproductive outcomes may not be specifically tied to aneuploidy, a perspective supported by another retrospective study conducted by the Urian’ team [16].

The dominant perspective posits that obesity has a detrimental effect on embryonic chromosomes, primarily by influencing the quantity of chromosomal euploids, consequently elevating the risk of miscarriage. Nevertheless, the exact mechanism behind this phenomenon remains ambiguous, underscoring the need for additional research to explore the correlation between obesity and the quantity and configuration of both oocytes and embryonic chromosomes.

Obesity leading to abnormal oocytes quality

Oocytes are crucial in the embryonic development process, with the quality of oocytes significantly influencing the subsequent development of embryos. Leary et al. compared the morphology and developmental dynamics of embryos from healthy individuals with those from women with obesity and overweight. They found that oocytes from women with obesity and overweight were significantly smaller than those from women with a healthy BMI [17]. Furthermore these embryos often exhibit metabolic abnormalities, which could potentially jeopardize the health of the fetus and future generations. Additionally, embryos derived from oocytes of women with obesity and overweight also demonstrate lower developmental potential after IVF, with reduced blastocyst formation rates [18]. These results indicate that obesity increases the likelihood of reproductive system abnormalities by influencing the quality of oocytes. Subsequently, we will explore the precise mechanisms implicated in this phenomenon. We will discuss the impact and mechanisms of obesity on oocytes quality from four aspects: (i) Abnormal extracellular environment of oocytes; (ii) Telomere dysfunction; (iii) Stella levels decrease; (iv) Mitochondrial dysfunction (Fig. 2).

Fig. 2
figure 2

Obesity leading to abnormal oocytes quality. A A high concentration of palmitic acid in follicular fluid induces ceramide accumulation and downregulates the AMPK/SIRT3 pathway, resulting in oocytes mitochondrial protein hyperacetylation and dysfunction. B Obesity can shorten the telomere length of oocytes, affect the development of embryos and fetuses, and consequently shorten neonatal telomeres. C Female mice knocked out for Stella could produce oocytes that appear normal, but the embryos were unable to reach the blastocyst stage. D Obesity leads to decreased mtDNA content and increased mitochondrial single nucleotide variations in oocytes, which can also result in mitochondrial dysfunction. Abbreviations: AMPK: adenosine 5’-monophosphate (AMP)-activated protein kinase; SIRT3: sirtuin 3; ATP: adenosine triphosphate; ROS: reactiveoxygenspecies; Stella KO: stella knockout mice; mtDNA: mitochondrial DNA; mtSNVs: mitochondrial nucleotide variations; MMP: membrane potential

Abnormal extracellular environment of oocytes

The maturation process of oocytes is a multifaceted phenomenon governed by various intrinsic and extrinsic factors within the ovarian environment. External elements such as follicular fluid (FF) and granulosa cells play a crucial role in providing essential nutrients and growth factors that facilitate the growth, maturation, and overall health of oocytes [19]. Granulosa cells contribute a diverse array of energy sources and nutrients to support the development and fertilization of oocytes [20]. Findings from in vitro fertilization-embryo transfer experiments demonstrated a decline in mitochondrial membrane potential, adenosine triphosphate levels, and mitochondrial DNA (mtDNA) copy number in granulosa cells of patients with obesity with polycystic ovary syndrome (PCOS) alongside an increase in reactive oxygen species levels. Notably, the formation of high-quality embryos, usable embryos, and high-quality blastocysts was significantly reduced in these patients [21]. Studies on mice subjected to a high-fat diet (HFD) have revealed an augmentation in corpus luteum count and heightened levels of estradiol and progesterone in the early stages of pregnancy. Treatment with oleic acid (OA) and palmitic acid (PA) resulted in a notable increase in estradiol and progesterone levels in luteinized KGN cells, suggesting that HFD-induced obesity during early pregnancy may compromise ovarian function and impact oocytes development [22]. Furthermore, consumption of a high-fat diet has been shown to upregulate the expression of miR-133a in adipose tissue and ovaries. miR-133a functions as a regulator, promoting apoptosis in granulosa cells by modulating the expression of anti-apoptotic and pro-apoptotic proteins, consequently leading to aberrant follicular development in obese mice [23].

The metabolites present in follicular fluid play a critical role in the maturation and growth of oocytes. In individuals with PCOS who are overweight or obese, a cluster of proteins associated with inflammation, immune response, and metabolic alterations has been identified in follicular fluid, which has a direct impact on oocytes development [24]. Elevated levels of inflammatory cytokines such as IL-6 and TNF-α have been observed in the follicular fluid of women with obesity, indicating an immune imbalance that could potentially harm developing oocytes [25]. Additionally, research has demonstrated a strong positive relationship between maternal BMI and the concentration of fatty acids in follicular fluid [26]. Increased levels of PA in follicular fluid have been found to trigger ceramide accumulation and suppress the AMPK/SIRT3 pathway in endometrial epithelial cells, resulting in heightened acetylation and functional impairment of mitochondrial proteins in oocytes [27] (Fig. 2A). Obesity has the capacity to disrupt oocytes metabolism and signaling pathways by influencing the functionality and metabolic status of granulosa cells, as well as the chemical composition of follicular fluid, potentially leading to developmental irregularities and reduced reproductive function. These discoveries underscore the significance of maintaining a healthy weight and metabolic profile for optimal fertility, and offer promising avenues for future therapeutic interventions.

Telomere dysfunction

Telomeres are essential in various stages of gametogenesis and embryogenesis, playing critical roles in chromosome orientation, synapsis, and segregation, all of which are vital for preserving genomic integrity and stability [28]. Research indicates a significant correlation between the length of telomeres in oocytes and embryonic development. Maternal obesity has been linked to telomere dysfunction, leading to chromosomal instability, reduced oocytes quality, and compromised embryonic developmental potential [29]. Studies by Antunes have demonstrated that women with overweight tend to have shorter telomeres in their oocytes compared to those with a normal BMI [30], underscoring the association between maternal weight and oocytes quality. Furthermore, investigations suggest a connection between the length of telomeres in newborns and the mother’s pre-pregnancy BMI. An increase in the mother’s pre-pregnancy BMI may result in a decrease in telomerse length in newborns, potentially impacting their future health and development [31]. In conclusion, these findings highlight the significance of telomeres in reproductive and developmental processes, as well as the influence of maternal obesity on oocytes quality and embryonic development through telomeres mechanisms (Fig. 2B).

Stella levels decrease

Stella, a well-known maternal factor, is present in primordial germ cells (PGCs) and serves a critical function during the initial stages of embryogenesis and cleavage [32]. It safeguards the distinct oocytes epigenome by preventing aberrant DNA methylation facilitated by DNMT1 and UHRF1 [33]. In mice subjected to a HFD, there was a notable decrease in Stella protein levels within oocytes. Introducing excess Stella in oocytes derived from mothers with obesity mitigated the developmental abnormalities observed in embryos. Remarkably, female mice lacking Stella produced seemingly normal oocytes that seldom progressed into blastocysts, displaying a phenotype akin to embryos from HFD mice [34] (Fig. 2C). In essence, obesity results in diminished Stella levels in oocytes, impeding embryonic development. Stella plays a safeguarding role in the epigenetic control of oocytes, and changes in Stella expression levels, prompted by obesity, have significant implications for embryonic development.

Mitochondrial dysfunction

Mitochondrial mass and mtDNA content massively increase during oocyte growth. They are highly dynamic organelles and oocyte maturation is accompanied by mitochondrial trafficking around subcellular compartments. Due to their key roles in generation of ATP and reactive oxygen species (ROS), mitochondrial defects have largely been linked with oocyte dysfunction [35]. Research studies have demonstrated that obese mouse oocytes cells exhibit various mitochondrial functional abnormalities, including decreased mitochondrial membrane potential, altered mitochondrial distribution, and structural irregularities, as well as defects in spindle/chromosome organization [36]. Marei et al. have reported that a high-fat die leads to a notable increase in mitochondrial inner membrane potential (MMP), reactive oxygen species levels, mitochondrial structural abnormalities, and endoplasmic reticulum (ER) swelling, while decreasing mitochondrial DNA (mtDNA) copy number [37]. These changes suggest that obesity may contribute to impaired mitochondrial function in egg cells. Furthermore, obesity can reduce mtDNA content in fully developed oocytes cells and cause mitochondrial nucleotide variations (mtSNVs), which can negatively affect mitochondrial function and oocytes cell quality [38] (Fig. 2D). Conversely, treatment with IGF2 in mature oocytes cells has been shown to enhance oocytes cell maturation and mitochondrial function, thereby alleviating the negative effects of obesity [36]. In conclusion, These animal studies highlight the detrimental influence of obesity on oocytes cell mitochondrial function, which can compromise oocytes cell maturation and female fertility.

Suboptimal oocytes quality is a notable element influencing adverse reproductive results in females with obesity. Efforts are underway to investigate approaches aimed at enhancing oocytes quality in this demographic. Antioxidant therapy is a prevalent intervention utilized to mitigate obesity-related oxidative stress and enhance oocytes quality. While existing studies predominantly concentrate on animal models, the Table 1 delineates prospective therapeutic avenues that could be relevant to human subjects. Despite the promising outcomes observed with current treatment modalities, additional research is imperative to substantiate their efficacy in clinical settings. The enhancement of oocytes quality in women with obesity necessitates further investigation and scholarly inquiry.

Table 1 Drug therapy to improve oocytes quality

Obesity leading to impaired endometrial development

After the fusion of sperm and eggs, the zygotes must successfully implant within a specific timeframe. This process requires the endometrium, which is influenced by estrogen and progesterone from the corpus luteum, to exhibit receptivity. This critical phase is essential for the initiation of pregnancy. Endometrial receptivity, known as the “window of implantation”, denotes a hormonally regulated stage where the endometrial tissue temporarily transitions into a functional state to facilitate blastocyst implantation and the subsequent establishment of pregnancy [50]. The largest single cause of pregnancy loss is implantation error, with defective implantation rates in humans reaching up to 78%. Successful pregnancy establishment hinges on proper implantation, which requires a complex interplay between the endometrium and the blastocyst [51]. In addition, during pregnancy, the endometrium undergoes decidualization, a vital process for embryo implantation, pregnancy maintenance, and labor initiation [52]. The uterine decidua plays various roles, such as aiding in gas and nutrient exchange between the mother and fetus, immune regulation, trophoblast invasion, and placental development [53]. Deficiencies in endometrial decidualization can disrupt the interaction between the embryo and mother, leading to recurrent miscarriages [54]. Comstock et al. noted significant differences in endometrial transcriptomes between individuals with obesity and non-obesity, suggesting that these variances may contribute to a decreased implantation rate and a higher risk of miscarriage in women with obesity [55].

Bellver et al.’ study demonstrated that obesity negatively impacts the receptivity of the endometrium, potentially influencing the endometrial environment and resulting in a delayed Window of Implantation (WOI) and consequently poorer outcomes in Assisted Reproductive Technology (ART) [56]. A prospective cohort study demonstrated a correlation between BMI levels and the displacement rate of WOI, with percentages of 4.2% for normal weight individuals, 17.2% for those who are overweight, and 24.2% for patients with obesity. Following the clinical correction of displaced WOI through personalized embryo transfer (pET), there were no significant variations in pregnancy rate, implantation rate, or rates of pregnancy loss (6.7% vs. 13.2%) [57]. Weight reduction in obese women with polycystic ovary syndrome (PCOS) who are experiencing infertility could have a positive impact on endometrial receptivity [58]. At the animal level, studies have shown compromised endometrial receptivity in early-pregnancy mice due to maternal hyperinsulinemia on mouse models with hyperinsulinemia [59]. Additionally, obesity has been found to alter the transcriptional profile of sheep endometrium, leading to changes in gene expression during implantation, potentially contributing to obesity-related pregnancy complications [60] These findings suggest that obesity may increase miscarriage rates by affecting the duration of the implantation window. Strategies such as weight loss or interventions to correct the WOI may help mitigate some of the adverse pregnancy effects associated with obesity.

Furthermore, obesity has the potential to influence the decidualization process, which could result in miscarriage. Research conducted by Antoniotti et al. revealed that elevated levels of inflammation in the uterine cavity of individuals with obesity may trigger inflammatory signaling in endometrial epithelial cells. This activation can lead to endoplasmic reticulum stress, hindering decidualization and blastocyst implantation, as well as impeding trophoblast invasion, ultimately culminating in miscarriage and pregnancy complications [61]. In comparison to individuals with lower body weight, patients with obesity exhibited a significant reduction in the gene expression of PRL and IGFBP1, two markers associated with endometrial decidualization. Impaired autophagy flux in patients with obesity was also observed, prompting the authors to suggest that decreased autophagy might be accountable for compromised in vitro decidualization [62]. Studies involving animals and cells have demonstrated that the HFD can induce impaired decidualization in mice with obesity and metabolic syndrome [63]. HFD mice impair decidualization by disrupting glycolysis in the endometrium during early pregnancy [64]. Obesity can impede decidualization through various mechanisms, including inflammation, autophagy, fatty acids, and glycolysis. By affecting processes such as inflammation signaling, alterations in gene expression, impaired autophagy flux, and interference with metabolic pathways, obesity can disrupt the normal decidualization of the endometrium, potentially heightening the risk of RSA.

In brief, Obesity may shift the window of implantation and alter proper decidualization, which could potentially impact pregnancy outcomes.

Obesity leading to maternal immune imbalance

Obesity is commonly associated with chronic inflammation, characterized by significant alterations in the typical energy storage and endocrine functions of adipocytes. This condition is marked by heightened pro-inflammatory signaling, adipokine secretion, and cellular apoptosis [65]. Table 2 summarizes the changes in immune cells and their mechanisms in individuals with obesity. At a cellular level, obesity results in a reduction in anti-inflammatory Treg and Th2 cells, alongside an elevation in pro-inflammatory Th1 cells and CD8 + T cells. Furthermore, the balance between M2 and M1 macrophages is disrupted, with an increase in pro-inflammatory M1 macrophages [66]. Research indicates that the accumulation of pro-inflammatory macrophages is a central characteristic of obesity and a significant contributor to adipose tissue inflammation and associated complications [67].

During pregnancy, maintaining a delicate equilibrium between pro-inflammatory and anti-inflammatory cytokines is essential for successful implantation [68]. A review of immune cell alterations in individuals experiencing recurrent miscarriage is presented in Table 3. For instance, in cases of RSA, there is an elevated presence of Th17 cells and a reduced number of Treg cells in the decidua, suggesting a correlation between the Th17/Treg cells ratio modification at the maternal-fetal interface and the development of RSA [69]. Furthermore, obesity in pregnant women can lead to an upsurge in pro-inflammatory cells and factors, disrupting the balance between pro-inflammatory and anti-inflammatory cytokines, consequently leading to unfavorable pregnancy outcomes.

Table 2 Changes in immune cells in obese individual
Table 3 Immune cells changes associated with RSA

uNK cells

Uterine natural killer (uNK) cells represent a majority of early pregnancy uterine leukocytes, comprising 60–70% of the total population [109]. These cells are pivotal in regulating neovascularization, uterine artery remodeling, placental development, and immune responses during pregnancy [110]. Research indicates that IFN-γ, originating from uNK cells, plays a crucial role in facilitating uterine spiral artery remodeling [111]. Obesity is further associated with a notable decline in uNK cell numbers, resulting in impaired uterine artery remodeling. Additionally, in cases of obesity, uNK cells demonstrate an exaggerated response to platelet-derived growth factor (PDGF), causing excessive decorin expression that hampers trophoblast survival and impedes placental development, potentially causing placental damage [70]. In summary, obesity can diminish uNK cells quantities and disrupt uterine artery remodeling, thereby influencing pregnancy outcomes. The results suggest a relationship between obesity and the dysfunction of uNK cells, which may have negative implications for pregnancy outcomes.

T cells

During the early stages of embryo implantation, T cells constitute 3–10% of the white blood cells at the maternal-fetal interface [112]. Th17 cells primarily produce pro-inflammatory cytokines, while Treg cells mediate immunosuppression [113]. In the early stage of embryo implantation, the endometrium is in a pro-inflammatory environment, and the peripheral blood and decidua of RSA women exibit an increase in pro-inflammatory Th17 cells and a decrease in anti-inflammatory Treg cells [114]. In the third trimester, immunosuppressive T cells dominate, and Tregs are usually significantly increased at the maternal-fetal interface and in peripheral blood during this period [115]. Liao et al. found that women with a low Treg cells group in unexplained RSA (URSA) exhibited approximately five times the risk of RSA compared to women with normal Treg levels [116]. Research has also found that, compared with normal patients, the expression of Treg cells in RSA patients was decreased, while the expression of Th17 cells was increased [117]. Hence, the imbalance between Th17 and Treg cells is closely related to RSA. Both obese individuals and obese mouse models have shown an increase in Th17 cells and a decrease in Treg cells, indicating a similar trend in T cells alterations between individuals with obesity and RSA patients [76, 78, 79]. This implies that obesity may contribute to the progression of RSA by elevating the number of pro-inflammatory T cells.

Macrophages

Macrophages are the second-largest group of immune cells, comprising 20% of total leukocytes at the maternal-fetal interface [118]. They are involved in various functions such as antigen presentation, coordinating tissue remodeling and angiogenesis, inducing apoptosis of damaged cells, promoting trophoblast invasion, and inhibiting inflammation. Decidual macrophages are essential for fostering maternal and fetal immune tolerance [118, 119]. In pregnancies affected by obesity, there is a notable increase in the number of macrophages compared to those in normal-weight women, along with heightened expression of IL-6 and GRO-α proteins in the decidua [74]. Moreover, individuals with obesity demonstrate elevated levels of placental resident macrophages and increased expression of macrophage-related cytokines and specific factors like CD14, CD68, and EMR-1 [120], which are crucial for proinflammatory processes. In essence, macrophages play a critical role in obesity-induced RSA, as the obesogenic environment impacts pregnancy outcomes by enhancing pro-inflammatory macrophages and stimulating the release of pro-inflammatory cytokines.

Under the state of obesity, alterations in immune cell numbers and functions are associated with adverse pregnancy outcomes, such as RSA. NK cells, T cells, and macrophages can influence placental function and alter intrauterine inflammation, contributing to RSA. Understanding the physiological processes related to immune cells and obesity during pregnancy is vital.

Obesity leads to disrupted secretion of adipokines

Adipokines, which are bioactive peptides secreted by adipocytes, play a vital role in the regulation of energy, inflammation, and immune responses [121]. In cases of obesity, there is an upregulation of pro-inflammatory adipokines and a downregulation of anti-inflammatory adipokines. Optimal adipokine levels are fundamental for maintaining the integrity of the hypothalamic-pituitary-gonadal axis, regulating ovulation processes, achieving successful embryo implantation, and ensuring physiological pregnancies [122]. Pro-inflammatory adipokines consist of leptin, resistin, visfatin, and chemerin, while anti-inflammatory adipokines include adiponectin, omentin, and isthmin 1 [108, 123, 124]. This discussion will specifically focus on three adipokines that are closely associated with RSA.

Leptin, a significant hormone involved in the process of implantation, plays a crucial role in the regulation of placental development and endometrial receptivity [125]. Research conducted by Plowden revealed that elevated levels of leptin prior to pregnancy in women with a history of miscarriage were linked to decreased fertility, pregnancy, and live birth rates. This correlation was closely associated with BMI and leptin concentrations [126]. Additionally, Müller et al. discovered that the GTCC haplotype of the leptin receptor (LEPR) gene locus in patients with RSA was notably lower compared to that in healthy individuals [127]. Consequently, deviations in leptin levels and leptin receptors may result in impaired trophoblastic cell proliferation, leading to unfavorable pregnancy outcomes.

Adiponectin plays a crucial role in pregnancy and placental development by interacting with adiponectin receptors present in various tissues, which are vital for embryonic development and endometrial receptivity [128]. Research has demonstrated that women experiencing RSA exhibit significantly elevated serum adiponectin levels compared to a control group, while maintaining similar serum leptin levels [129]. At the animal level, studies have revealed that mice with adiponectin haploinsufficiency or homozygous mutations exhibit a notably reduced litter rate in comparison to wild-type mice [130]. Furthermore, the administration of recombinant adiponectin (rAPN) therapy has been shown to enhance pregnancy outcomes in aborted mouse models by augmenting the quantity and functionality of Treg cells and suppressing the quantity and functionality of Th17 cells [131]. These findings collectively underscore the critical role of adiponectin in preserving normal fertility in female mice.

Fatty Acid-Binding Protein 4 (FABP4), another adipokine, coordinates lipid transport in mature adipocytes. FABP4 regulates embryo implantation by altering uterine receptivity, and decreased expression of FABP4 in the endometrium may be related to pregnancy loss [132]. Animal experiments showed that FABP4 can also block blood vessel formation and lead to the obstruction of trophoblast vascular remodeling in early pregnancy [133]. Inhibition of FABP4 can reduce BMI and improve insulin resistance in obese mice [134]. Therefore, FABP4 is closely associated with obesity and pregnancy loss.

In brief, the roles and significance of leptin and adiponectin in pregnancy are crucial. Dysregulation of their levels and functions can result in trophoblast dysfunction and negative pregnancy consequences. Notably, deviations in leptin and adiponectin receptors have been linked to issues such as infertility and RSA. Furthermore, the presence of FABP4 has been correlated with conditions like obesity, insulin resistance, and pregnancy loss, suggesting that these elements could serve as potential targets for addressing infertility and enhancing pregnancy outcomes. Consequently, these discoveries offer valuable biological insights into the management and treatment of reproductive disorders related to obesity, underscoring the importance of weight control in enhancing fertility outcomes.

Obesity related genes FTO and RSA

In 2007, Dina et al. discovered that single nucleotide polymorphisms (SNPs) in the FTO gene region were associated with BMI and obesity risk, establishing FTO as the first locus explicitly linked to obesity [135]. The protein encoded by the FTO gene is an m6A demethylase, which can regulate m6A modifications on mRNA and thereby affect gene expression and metabolic pathways [136]. The activity of the FTO gene is associated with the signal transduction in the appetite control center, and its variation may cause increased appetite, which in turn influences the risk of obesity [137]. Experimental data obtained from animal models suggest that knockout of the FTO gene inhibited obesity and promoted energy expenditure, while overexpression of FTO increased food intake and obesity [138, 139]. Furthermore, overexpression of FTO can promote adipogenesis in 3T3-L1 preadipocytes and porcine intramuscular adipocytes [140]. Thus, the findings from both in vitro and in vivo studies indicate that FTO can facilitate fat accumulation and contribute to obesity. Recent studies have elucidated a relationship between the FTO and RSA. Andraweera et al. analyzed the peripheral blood of 202 patients with RSA in early pregnancy and found that the FTO rs9939609 SNPs may be associated with RSA [141]. Qiu et al. discovered that the downregulation of FTO in chorionic villi disrupts immune tolerance and angiogenesis at the maternal-fetal interface, leading to abnormal methylation, oxidative stress, and eventually RSA [142]. Another study revealed that FTO promotes MEG3 degradation by inhibiting MEG3 m6A modification in the trophoblast, thereby disrupting the MEG3-TGF-β regulatory axis. This disruption results in abnormal activation of the TGF-β signaling pathway, inhibiting cell invasion, and causing RSA [143].

The function of the FTO is not solely linked to the initiation of obesity but may also impact the susceptibility to RSA through its effects on embryonic development and immune function at the interface between the mother and fetus. Consequently, the FTO could potentially act as a crucial molecular bridge connecting obesity and RSA. Investigation into the FTO has the potential to enhance our comprehension of the molecular pathways involved in obesity and provide innovative approaches for the prevention and management of RSA.

The association between male obesity and RSA

In early pregnancy, the mother’s influence on the embryo is more pronounced than the father’s, but sperm also plays a crucial role in embryonic development, and abnormal sperm may lead to miscarriage. Hence, in this section, we briefly summarize some mechanisms by which obesity affects male reproductive health and may causes recurrent miscarriage. A retrospective multicenter case-control study suggests an association between increased sperm DNA fragmentation and idiopathic recurrent miscarriage [144], and the weight of the father is inversely related to the prognosis of future live births in couples with RSA [145]. Sperm DNA damage measured by the alkaline Comet has promise as a robust biomarker for sporadic and recurrent miscarriage after spontaneous or assisted conception, and may provide novel diagnoses and guidance for future fertility pathways [146]. Analysis of gene expression profiles has revealed a sex-specific response of the transcriptome to maternal and paternal obesity. Changes in the male blastocyst transcriptome were more pronounced than those in female blastocysts, and the impact of paternal obesity was greater than maternal obesity [147]. Paternal obesity can induce epigenetic and gene expression changes in sperm and the placenta. Epigenetic abnormalities in sperm can affect the expression of imprinted genes in the placenta and genes related to placental endocrine pathways [148]. Additionally, male obesity is linked to alterations in sperm DNA methylation profiles, which may compromise the reprogramming fidelity of sperm subsets and impede normal embryonic development [149]. Animal experiments have shown that the expression of SETD2 in the testis and sperm of male mice fed a high-fat diet for two months significantly increased. This alteration led to changes in DNA methylation levels, leading to increased apoptosis of blastocysts and a reduced total cell count in blastocysts [150]. FTO knockout mice exhibited an age-dependent decline in sperm concentration and serum testosterone levels [151]. Furthermore, the abundance of the MC4R gene in sperm was negatively correlated with sperm motility and positively correlated with biochemical pregnancy. The abundance of MC4R and GNPDA2 transcripts in sperm from men with asthenospermia and teratoospermia was higher than in normospermia [152].

Pregnancy is not solely the mother’s responsibility, the father also plays a significant role. In cases of RSA, alterations in DNA methylation levels may manifest in the sperm of fathers with obesity. These changes can result in placental epigenetic modifications and abnormal embryonic development. Obesity-related genes, such as FTO, MC4R, and GNPDA2, can also impact pregnancy outcomes by affecting sperm count and motility. For men with obesity, weight loss is a crucial step when seeking to improve fertility.

The significance of maintaining appropriate weight levels throughout pregnancy

Potential benefits of weight loss for patients with RSA

For infertile women with obesity who want to conceive, weight loss may be a solution to improving fertility. A systematic review and meta-analysis showed that bariatric surgery significantly reduced the rate of miscarriage in treated women (p = 0.01), RR 0.51,95% CI 0.30, 0.86 [153]. In addition, A retrospective cohort study showed that overweight infertile patients who lost 10% of their body weight had significantly higher pregnancy and live birth rates compared to those who did not lose weight [154]. However, this finding is not consistently supported by randomized controlled trials (RCTs). In obese women with PCOS who are infertile, weight loss positively affects endometrial receptivity, with 25% of women who successfully lose weight seeing an increased rate of natural pregnancy [58]. These data suggest that meaningful weight loss can improve the pregnancy and live birth rates among overweight infertile patients. Women who conceive naturally avoid IVF procedures, which alleviates significant psychological, physical, and financial burdens. Moreover, weight loss before pregnancy can also decrease the risk of adverse conditions during gestation. It has been observed that as the weight loss ratio increases, the incidence of hypertensive disorders during pregnancy, preterm delivery, stillbirth, and low birth weight tends to decline [155, 156]. Although some studies suggest that strengthening pre-pregnancy lifestyle intervention for weight loss does not improve fertility or birth outcomes [157, 158], some studies have shown that weight loss before pregnancy increases the risk of miscarriage [159], most current studies believe that if pregnant women control their weight at a healthy level before pregnancy, it is beneficial for both themselves and the fetus [160, 161]. Prepregnancy weight loss should be supervised by a clinician with specific expertise in obesity treatment. Weight loss may be required for longer intervals than those used in existing studies before attempting to conceive [162]. We advocate for women with obesity to lose weight before and during pregnancy, and to control their weight within a relatively normal range.

Managing weight during pregnancy through diet and exercise

Weight loss can help restore fertility in individuals with obesity and reduce the risk of RSA. Weight loss may improve fertility in individuals with obesity, although its effect on reducing the risk of RSA is not yet fully established. In addition to focusing on RSA, we also need to consider the impact of weight management on other pregnancy complications. Being overweight before pregnancy can increase the incidence of gestational diabetes, pregnancy complications, macrosomia, and preterm birth. Measures such as adopting healthier eating habits and engaging in regular physical exercise can enhance the mother’s health and optimize fetal development.

For patients with obesity, adjusting weight through diet is a fundamental aspect of disease management. For pregnant women with a pre-pregnancy BMI of ≥ 35 kg/m², individual assessment and dietary consultation by dietitians can help achieve and limit gestational weight gain, reduce the incidence of gestational diabetes mellitus, and decrease the likelihood of cesarean Sect. [163]. Following the implementation of a low glycemic index diet during pregnancy and a subsequent five-year follow-up, evidence emerged indicating a causal relationship between reduced gestational weight gain (GWG) during pregnancy and enhanced maternal well-being, as well as a decrease in overall weight gain. Pregnant women also experienced a change in their usual diet after delivery, showing a preference for foods rich in protein, with lower GI and lower GL energy [164]. Studies have shown that the Mediterranean diet can help improve diabetes, inflammatory diseases, cancer, and cognitive decline [165]. It can also reduce maternal anxiety and stress and improve sleep quality throughout pregnancy [166, 167]. There is evidence suggesting that diet is linked to inflammation, oxidative stress, and brain function, factors that may be associated with mental disorders. A study examining the relationship between dietary patterns and mental disorders in pregnant women in southern Brazil discovered that excessive consumption of sweets and sugars, as well as inadequate intake of fruits, were strongly linked to depression. Generalized anxiety, on the other hand, was associated with low consumption of beans [168]. These pieces of evidence clearly demonstrate that dietary habits and nutritional intake during pregnancy are crucial. Moderate energy intake and a balanced diet not only impact the mothe’s health and reduce the occurrence of complications during pregnancy but also benefit the growth and development of the fetus. Additionally, they can have long-term positive effects on the mother.

Exercise during pregnancy has positive effects on both the mother and the fetus. Engaging in an exercise program has psychological benefits for pregnant women, such as reduced anxiety and depression, as well as improved well-being. Moderate-intensity aerobic exercise and exercise interventions have been shown to mitigate weight gain during pregnancy [169]. Moderate-intensity exercise and exercising during the second trimester can help control blood sugar levels [170]. In addition to reducing pregnancy weight and preventing gestational diabetes, researchers have found that pregnant women are more likely to regain their pre-pregnancy weight within six months after delivery [171]. From a psychological perspective, physical exercise during pregnancy and the postpartum period can improve the mental health of pregnant women and reduce the probability of postpartum depression [172]. In the absence of medical or obstetric complications, the American College of Obstetricians and Gynecologists recommends 30 min of aerobic exercise per day for physical and mental health during pregnancy [173]. In the third trimester of pregnancy, pregnant women often experience poor sleep due to symptoms such as uterine enlargement, bladder compression, and back pain. Engaging in prenatal yoga practice has been shown to effectively improve the sleep quality of pregnant women in the third trimester [174]. The main benefits of exercise for pregnant women include improved physical condition, weight control, shorter labor time, faster postpartum recovery, and prevention of gestational diabetes, gestational hypertension, preeclampsia, and preterm birth.

However, these studies primarily rely on association analysis, without having clearly established causal relationships. Future research should delve deeper into the causal relationships and their intensity, aiming to provide more definitive evidence.

A women-centered approach is essential to advance weight management strategies. In clinical practice, we have found women seek practical advice delivered in a sensitive and respectful manner, rather than a sole focus on the risks of obesity during pregnancy. Weight control becomes more challenging, particularly for individuals who need to improve their nutrition during pregnancy. Women with obesity must manage their weight gain within a certain range during pregnancy, necessitating strict monitoring. Whether through dietary intervention, intensive exercise, or a combination of both, these strategies positively impact pregnancy outcomes.

How clinicians should evaluate and manage patients with obesity and RSA

RSA is a complex disease that often involves multiple medical factors and lifestyle influences. For patients with obesity, clinicians should evaluate and manage RSA by considering a combination of the following factors. For patients with obesity, clinicians should evaluate and manage RSA by considering a combination of factors while being mindful of the stigma and bias that these patients may face within the healthcare system. It is crucial to ensure a full and unbiased evaluation, not just assuming that miscarriage is secondary to body size. It is well-established that obesity is associated with an increased risk of miscarriage. Therefore, patients with obesity should actively take steps to lose weight to reduce this risk. The endocrine environment of obese patients may affect the risk of miscarriage. PCOS, a gynecological disease closely related to obesity, is closely associated with RSA [175]. Additionally, the evaluation of markers of glucose/insulin metabolism as well as the levels of vitamin D as important step to be included in the diagnostic work-up of RPL women [176]. Immune factors may also play a pivotal role in RSA. Clinicians should identify patients with or without immune system diseases during clinical consultations and treatments and actively address the primary diseases [177]. In addition, genetic factors may cause RSA, and clinicians should conduct genetic tests on both spouses to rule out possible genetic factors [178]. In conclusion, clinicians should develop personalized evaluation and management programs tailored to the specific conditions of patients, taking into account genetic, endocrine, immune, and other factors to reduce the risk of RSA.

Summary and future perspectives

This review delves into the association between obesity and RSA, highlighting that obesity is more likely to result in euploid miscarriages rather than aneuploid miscarriages. Obesity can negatively impact oocytes quality, influencing embryo implantation, and hinder decidualization, consequently affecting the rate of embryo implantation. Moreover, obesity can disrupt the balance of endometrial immune cells and cytokines, perturb adipocytokines released by adipocytes, and disturb the expression of specific obesity-related genes. These factors collectively contribute to the initiation and progression of RSA. Additionally, male obesity can compromise sperm quality, thereby diminishing fertility. Therefore, the influence of male obesity on fertility necessitates consideration. Given the escalating concern of obesity, interventions aimed at reducing the weight of women with obesity through dietary modifications, physical activity, and potentially medication are anticipated to enhance fertility outcomes in RSA patients.

In clinical practice, it is imperative for physicians to conduct a thorough assessment of the patien’s weight and ascertain potential causes of previous miscarriages. Tailored evaluation and management strategies should be devised to mitigate the risk of RSA. The correlation between obesity and RSA is multifaceted, necessitating further investigation through clinical and experimental research to elucidate its precise pathogenesis. Individuals with obesity encounter challenges in conception and are predisposed to unfavorable pregnancy outcomes. Additional research is warranted to elucidate the involvement of obesity in the pathogenesis of RSA and the associated regulatory mechanisms, with the goal of implementing prospective therapeutic interventions.

As a systemic disease, obesity triggers numerous mechanisms that impact female reproductive health. At present, there is no ideal treatment for obesity and obesity-induced RSA. Studying the effects of obesity on RSA and its underlying mechanisms holds significant social and clinical value for improving pregnancy outcomes. This article reviews the impact of obesity on female RSA and fertility. It examines the association between obesity and RSA as well as the influence of weight management on pregnancy. The aim is to provide insights for research, prevention, and treatment of RSA in women with obesity.

Data availability

No datasets were generated or analysed during the current study.

References

  1. Cavalcante MB, Sarno M, Peixoto AB, Araujo Júnior E, Barini R. Obesity and recurrent miscarriage: a systematic review and meta-analysis. J Obstet Gynaecol Res. 2019;45:30–8.

    Article  PubMed  Google Scholar 

  2. Bender Atik R, Christiansen OB, Elson J, Kolte AM, Lewis S, Middeldorp S, Nelen W, Peramo B, Quenby S, Vermeulen N, Goddijn M. ESHRE guideline: recurrent pregnancy loss. Hum Reprod Open. 2018;2018:004.

    Google Scholar 

  3. Stefan N, Schick F, Häring HU. Causes, characteristics, and consequences of metabolically unhealthy normal weight in humans. Cell Metab. 2017;26:292–300.

    Article  PubMed  CAS  Google Scholar 

  4. Kelly T, Yang W, Chen CS, Reynolds K, He J. Global burden of obesity in 2005 and projections to 2030. Int J Obes (Lond). 2008;32:1431–7.

    Article  PubMed  CAS  Google Scholar 

  5. Elagizi A, Kachur S, Lavie CJ, Carbone S, Pandey A, Ortega FB, Milani RV. An overview and update on obesity and the obesity Paradox in Cardiovascular diseases. Prog Cardiovasc Dis. 2018;61:142–50.

    Article  PubMed  Google Scholar 

  6. Mazon JN, de Mello AH, Ferreira GK, Rezin GT. The impact of obesity on neurodegenerative diseases. Life Sci. 2017;182:22–8.

    Article  PubMed  CAS  Google Scholar 

  7. Wang K, Zhao A, Tayier D, Tan K, Song W, Cheng Q, Li X, Chen Z, Wei Q, Yuan Y, Yang Z. Activation of AMPK ameliorates acute severe pancreatitis by suppressing pancreatic acinar cell necroptosis in obese mice models. Cell Death Discov. 2023;9:363.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Sugiura-Ogasawara M. Recurrent pregnancy loss and obesity. Best Pract Res Clin Obstet Gynaecol. 2015;29:489–97.

    Article  PubMed  Google Scholar 

  9. Boots CE, Bernardi LA, Stephenson MD. Frequency of euploid miscarriage is increased in obese women with recurrent early pregnancy loss. Fertil Steril. 2014;102:455–9.

    Article  PubMed  Google Scholar 

  10. Romanski PA, Bortoletto P, Magaoay B, Chung A, Rosenwaks Z, Spandorfer SD. Live birth outcomes in infertile patients with class III and class IV obesity following fresh embryo transfer. J Assist Reprod Genet. 2021;38:347–55.

    Article  PubMed  Google Scholar 

  11. Lee JC, Bernardi LA, Boots CE. The association of euploid miscarriage with obesity. F S Rep. 2020;1:142–8.

    PubMed  PubMed Central  Google Scholar 

  12. Tremellen K, Pearce K, Zander-Fox D. Increased miscarriage of euploid pregnancies in obese women undergoing cryopreserved embryo transfer. Reprod Biomed Online. 2017;34:90–7.

    Article  PubMed  Google Scholar 

  13. Cozzolino M, García-Velasco JA, Meseguer M, Pellicer A, Bellver J. Female obesity increases the risk of miscarriage of euploid embryos. Fertil Steril. 2021;115:1495–502.

    Article  PubMed  Google Scholar 

  14. Bellver J. BMI and miscarriage after IVF. Curr Opin Obstet Gynecol. 2022;34:114–21.

    Article  PubMed  Google Scholar 

  15. Hughes LM, McQueen DB, Jungheim ES, Merrion K, Boots CE. Maternal body mass index is not associated with increased rates of maternal embryonic aneuploidy. Fertil Steril. 2022;117:783–9.

    Article  PubMed  Google Scholar 

  16. Urian W, Siegel M, Parker PB, Garg B, Amato P, Wu D, Lee D, O’Leary T, Krieg SA, Obesity does not impact aneuploidy rates or mitochondrial dna copy number in patients undergoing preimplantation genetic testing for aneuploidy. Fertil Steril. 2020;114:e485.

    Article  Google Scholar 

  17. Leary C, Leese HJ, Sturmey RG. Human embryos from overweight and obese women display phenotypic and metabolic abnormalities. Hum Reprod. 2015;30:122–32.

    Article  PubMed  Google Scholar 

  18. Comstock IA, Kim S, Behr B, Lathi RB. Increased body mass index negatively impacts blastocyst formation rate in normal responders undergoing in vitro fertilization. J Assist Reprod Genet. 2015;32:1299–304.

    Article  PubMed  PubMed Central  Google Scholar 

  19. O’Gorman A, Wallace M, Cottell E, Gibney MJ, McAuliffe FM, Wingfield M, Brennan L. Metabolic profiling of human follicular fluid identifies potential biomarkers of oocyte developmental competence. Reproduction. 2013;146:389–95.

    Article  PubMed  Google Scholar 

  20. Fan W, Yuan Z, Li M, Zhang Y, Nan F. Decreased oocyte quality in patients with endometriosis is closely related to abnormal granulosa cells. Front Endocrinol (Lausanne). 2023;14:1226687.

    Article  PubMed  Google Scholar 

  21. Zhao YK, Gao YN, Wang LC, Wang J, Wang GJ, Wu HL. Correlation between abnormal energy metabolism of ovarian granulosa cells and in vitro fertilization-embryo transfer outcomes in patients with polycystic ovary syndrome and obesity. J Ovarian Res. 2023;16:145.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Li Q, Guo S, Yang C, Liu X, Chen X, He J, Tong C, Ding Y, Peng C, Geng Y, et al. High-fat diet-induced obesity primes fatty acid β-oxidation impairment and consequent ovarian dysfunction during early pregnancy. Ann Transl Med. 2021;9:887.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Chen R, Wu X, Qiu H, Yang B, Chen Y, Chen X, Li Y, Yuan S, Liu D, Xiao L, Yu Y. Obesity-induced inflammatory miR-133a mediates apoptosis of granulosa cells and causes abnormal folliculogenesis. Acta Biochim Biophys Sin (Shanghai). 2023;55:1234–46.

    Article  PubMed  CAS  Google Scholar 

  24. Zhang X, Xu X, Li P, Zhou F, Kong L, Qiu J, Yuan Z, Tan J. TMT Based Proteomic Analysis of Human Follicular Fluid from Overweight/Obese and normal-weight patients with polycystic ovary syndrome. Front Endocrinol (Lausanne). 2019;10:821.

    Article  PubMed  Google Scholar 

  25. Kim T, Zhao Y, Enninga E. Obese women undergoing in Vitro Fertilization have higher IL-6 and TNF-α levels in follicular fluid [27A]. Obstet Gynecol. 2020;135:15S.

  26. Valckx SD, Arias-Alvarez M, De Pauw I, Fievez V, Vlaeminck B, Fransen E, Bols PE, Leroy JL. Fatty acid composition of the follicular fluid of normal weight, overweight and obese women undergoing assisted reproductive treatment: a descriptive cross-sectional study. Reprod Biol Endocrinol. 2014;12:13.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Itami N, Shirasuna K, Kuwayama T, Iwata H. Palmitic acid induces ceramide accumulation, mitochondrial protein hyperacetylation, and mitochondrial dysfunction in porcine oocytes. Biol Reprod. 2018;98:644–53.

    Article  PubMed  Google Scholar 

  28. Turner S, Hartshorne GM. Telomere lengths in human pronuclei, oocytes and spermatozoa. Mol Hum Reprod. 2013;19:510–8.

    Article  PubMed  CAS  Google Scholar 

  29. Ge J, Li C, Sun H, Xin Y, Zhu S, Liu Y, Tang S, Han L, Huang Z, Wang Q. Telomere Dysfunction in oocytes and embryos from obese mice. Front Cell Dev Biol. 2021;9:617225.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Antunes DMF, Kalmbach KK, Wang F, Seth-Smith ML, Kramer Y, Kohlrausch FB, Keefe DL. Oocytes from women with diminished ovarian reserve and obesity have shortened telomeres. Fertil Steril. 2014;102:e331.

  31. Martens DS, Plusquin M, Gyselaers W, De Vivo I, Nawrot TS. Maternal pre-pregnancy body mass index and newborn telomere length. BMC Med. 2016;14:148.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Bortvin A, Goodheart M, Liao M, Page DC. Dppa3 / Pgc7 / stella is a maternal factor and is not required for germ cell specification in mice. BMC Dev Biol. 2004;4:2.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Li Y, Zhang Z, Chen J, Liu W, Lai W, Liu B, Li X, Liu L, Xu S, Dong Q, et al. Stella safeguards the oocyte methylome by preventing de novo methylation mediated by DNMT1. Nature. 2018;564:136–40.

    Article  PubMed  CAS  Google Scholar 

  34. Han L, Ren C, Li L, Li X, Ge J, Wang H, Miao YL, Guo X, Moley KH, Shu W, Wang Q. Embryonic defects induced by maternal obesity in mice derive from Stella insufficiency in oocytes. Nat Genet. 2018;50:432–42.

    Article  PubMed  CAS  Google Scholar 

  35. Adhikari D, Lee IW, Yuen WS, Carroll J. Oocyte mitochondria-key regulators of oocyte function and potential therapeutic targets for improving fertility. Biol Reprod. 2022;106:366–77.

    Article  PubMed  Google Scholar 

  36. Wan Y, Muhammad T, Huang T, Lv Y, Sha Q, Yang S, Lu G, Chan WY, Ma J, Liu H. IGF2 reduces meiotic defects in oocytes from obese mice and improves embryonic developmental competency. Reprod Biol Endocrinol. 2022;20:101.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Marei WFA, Smits A, Mohey-Elsaeed O, Pintelon I, Ginneberge D, Bols PEJ, Moerloose K, Leroy J. Differential effects of high fat diet-induced obesity on oocyte mitochondrial functions in inbred and outbred mice. Sci Rep. 2020;10:9806.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Chen Y, Ma G, Gai Y, Yang Q, Liu X, de Avila JM, Mao S, Zhu MJ, Du M. AMPK suppression due to obesity drives oocyte mtDNA heteroplasmy via ATF5-POLG Axis. Adv Sci (Weinh). 2024;11:e2307480.

  39. Wang L, Chen Y, Wei J, Guo F, Li L, Han Z, Wang Z, Zhu H, Zhang X, Li Z, Dai X. Administration of nicotinamide mononucleotide improves oocyte quality of obese mice. Cell Prolif. 2022;55:e13303.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Wang H, Zhu S, Wu X, Liu Y, Ge J, Wang Q, Gu L. NAMPT reduction-induced NAD(+) insufficiency contributes to the compromised oocyte quality from obese mice. Aging Cell. 2021;20:e13496.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Wen X, Han Z, Liu SJ, Hao X, Zhang XJ, Wang XY, Zhou CJ, Ma YZ, Liang CG. Phycocyanin improves Reproductive ability in obese female mice by restoring Ovary and Oocyte Quality. Front Cell Dev Biol. 2020;8:595373.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Novielli C, Anelli GM, Lisso F, Marzorati A, Parrilla B, Oneta M, Savasi VM, Cetin I, Mandò C. Effects of α-lipoic acid and myo-inositol supplementation on the oocyte environment of infertile obese women: a preliminary study. Reprod Biol. 2020;20:541–6.

    Article  PubMed  Google Scholar 

  43. Boots CE, Boudoures A, Zhang W, Drury A, Moley KH. Obesity-induced oocyte mitochondrial defects are partially prevented and rescued by supplementation with co-enzyme Q10 in a mouse model. Hum Reprod. 2016;31:2090–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Jia Z, Feng Z, Wang L, Li H, Wang H, Xu D, Zhao X, Feng D, Feng X. Resveratrol reverses the adverse effects of a diet-induced obese murine model on oocyte quality and zona pellucida softening. Food Funct. 2018;9:2623–33.

    Article  PubMed  CAS  Google Scholar 

  45. Itami N, Shirasuna K, Kuwayama T, Iwata H. Resveratrol improves the quality of pig oocytes derived from early antral follicles through sirtuin 1 activation. Theriogenology. 2015;83:1360–7.

    Article  PubMed  CAS  Google Scholar 

  46. Yang Q, Wang Y, Wang H, Li H, Zhu J, Cong L, Xu J, Chen W, Jiang Y, Sun Y. NAD(+) repletion attenuates obesity-induced oocyte mitochondrial dysfunction and offspring metabolic abnormalities via a SIRT3-dependent pathway. Clin Transl Med. 2021;11:e628.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Han L, Wang H, Li L, Li X, Ge J, Reiter RJ, Wang Q. Melatonin protects against maternal obesity-associated oxidative stress and meiotic defects in oocytes via the SIRT3-SOD2-dependent pathway. J Pineal Res. 2017;63:e12431.

  48. Yang Q, Luo X, Wang Y, Li H, Cong L, Sun Y. Cerium oxide nanoparticles protect against obesity-induced ovarian dysfunction. Appl Mater Today. 2021;22:100973.

    Article  Google Scholar 

  49. Li C, Zhang H, Wu H, Li J, Liu Q, Li Y, Pan M, Zhao X, Wei Q, Peng S, Ma B. Intermittent fasting improves the oocyte quality of obese mice through the regulation of maternal mRNA storage and translation by LSM14B. Free Radic Biol Med. 2024;217:157–72.

    Article  PubMed  CAS  Google Scholar 

  50. Neykova K, Tosto V, Giardina I, Tsibizova V, Vakrilov G. Endometrial receptivity and pregnancy outcome. J Matern Fetal Neonatal Med. 2022;35:2591–605.

    Article  PubMed  Google Scholar 

  51. Craciunas L, Pickering O, Chu J, Choudhary M, Žurauskienė J, Coomarasamy A. The transcriptomic profile of endometrial receptivity in recurrent miscarriage. Eur J Obstet Gynecol Reprod Biol. 2021;261:211–6.

    Article  PubMed  CAS  Google Scholar 

  52. Zhu RH, Dai FF, Yang DY, Liu SY, Zheng YJ, Wu ML, Deng ZM, Wang ZT, Zhang YW, Tan W, et al. The mechanism of insulin-like growth factor II mRNA-Binging protein 3 induce decidualization and maternal-fetal Interface Cross talk by TGF-β1 in recurrent spontaneous abortion. Front Cell Dev Biol. 2022;10:862180.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Salker M, Teklenburg G, Molokhia M, Lavery S, Trew G, Aojanepong T, Mardon HJ, Lokugamage AU, Rai R, Landles C, et al. Natural selection of human embryos: impaired decidualization of endometrium disables embryo-maternal interactions and causes recurrent pregnancy loss. PLoS ONE. 2010;5:e10287.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Tang L, Xu XH, Xu S, Liu Z, He Q, Li W, Sun J, Shuai W, Mao J, Zhao JY, Jin L. Dysregulated gln-Glu-α-ketoglutarate axis impairs maternal decidualization and increases the risk of recurrent spontaneous miscarriage. Cell Rep Med. 2023;4:101026.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Comstock IA, Diaz-Gimeno P, Cabanillas S, Bellver J, Sebastian-Leon P, Shah M, Schutt A, Valdes CT, Ruiz-Alonso M, Valbuena D, et al. Does an increased body mass index affect endometrial gene expression patterns in infertile patients? A functional genomics analysis. Fertil Steril. 2017;107:740–e748742.

    Article  PubMed  Google Scholar 

  56. Bellver J, Marín C, Lathi RB, Murugappan G, Labarta E, Vidal C, Giles J, Cabanillas S, Marzal A, Galliano D, et al. Obesity affects endometrial receptivity by displacing the window of implantation. Reprod Sci. 2021;28:3171–80.

    Article  PubMed  CAS  Google Scholar 

  57. Bellver J, Lathi RB, Labarta E, Vidal C, Giles J, Cabanillas S, Marzal A, Galliano D, Marin C, Ruiz-Alonso M et al. Obesity affects endometrial receptivity by inducing a displacement of the personalized WOI that after correction by personalized embryo transfer normalize clinical results. Fertil Steril. 2018;110:e69.

  58. Bergant G, Abdulkhalikova D, Šuštaršič A, Peterlin B, Vrtačnik Bokal E, Maver A, Videmšek M, Burnik Papler T. Expression of markers of endometrial receptivity in obese infertile PCOS women before and after the weight loss Program-A preliminary study. Cells. 2022;12:164.

  59. Li R, Wu J, He J, Wang Y, Liu X, Chen X, Tong C, Ding Y, Su Y, Chen W, et al. Mice endometrium receptivity in early pregnancy is impaired by maternal hyperinsulinemia. Mol Med Rep. 2017;15:2503–10.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. McCoski SR, Cockrum RR, Ealy AD. Short communication: maternal obesity alters ovine endometrial gene expression during peri-implantation development. J Anim Sci. 2022;100:skac090.

  61. Antoniotti GS, Coughlan M, Salamonsen LA, Evans J. Obesity associated advanced glycation end products within the human uterine cavity adversely impact endometrial function and embryo implantation competence. Hum Reprod. 2018;33:654–65.

    Article  PubMed  CAS  Google Scholar 

  62. Schulte MM, Stephens CS, Moley K. Obese women experience impaired in vitro decidualization due to decreased autophagy. Fertil Steril. 2016;106:e213–4.

    Article  Google Scholar 

  63. Rhee JS, Saben JL, Mayer AL, Schulte MB, Asghar Z, Stephens C, Chi MM, Moley KH. Diet-induced obesity impairs endometrial stromal cell decidualization: a potential role for impaired autophagy. Hum Reprod. 2016;31:1315–26.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Chen Z, E Y, Xiong J, Li W, Chen X, Li N, Long J, Tong C, He J, Li F, et al. Dysregulated glycolysis underpins high-fat-associated endometrial decidualization impairment during early pregnancy in mice. Biochim Biophys Acta Mol Basis Dis. 2023;1869:166659.

    Article  PubMed  CAS  Google Scholar 

  65. Mathis D. Immunological goings-on in visceral adipose tissue. Cell Metab. 2013;17:851–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Kratz M, Coats BR, Hisert KB, Hagman D, Mutskov V, Peris E, Schoenfelt KQ, Kuzma JN, Larson I, Billing PS, et al. Metabolic dysfunction drives a mechanistically distinct proinflammatory phenotype in adipose tissue macrophages. Cell Metab. 2014;20:614–25.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS, Tartaglia LA, Chen H. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest. 2003;112:1821–30.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Liang PY, Diao LH, Huang CY, Lian RC, Chen X, Li GG, Zhao J, Li YY, He XB, Zeng Y. The pro-inflammatory and anti-inflammatory cytokine profile in peripheral blood of women with recurrent implantation failure. Reprod Biomed Online. 2015;31:823–6.

    Article  PubMed  Google Scholar 

  69. Ali S, Majid S, Ali MN, Taing S, Rehman MU, Arafah A. Cytokine imbalance at materno-embryonic interface as a potential immune mechanism for recurrent pregnancy loss. Int Immunopharmacol. 2021;90:107118.

    Article  PubMed  CAS  Google Scholar 

  70. Perdu S, Castellana B, Kim Y, Chan K, DeLuca L, Beristain AG. Maternal obesity drives functional alterations in uterine NK cells. JCI Insight. 2016;1:e85560.

    Article  PubMed  PubMed Central  Google Scholar 

  71. O’Rourke RW, Gaston GD, Meyer KA, White AE, Marks DL. Adipose tissue NK cells manifest an activated phenotype in human obesity. Metabolism. 2013;62:1557–61.

    Article  PubMed  Google Scholar 

  72. Parker VJ, Solano ME, Arck PC, Douglas AJ. Diet-induced obesity may affect the uterine immune environment in early-mid pregnancy, reducing NK-cell activity and potentially compromising uterine vascularization. Int J Obes (Lond). 2014;38:766–74.

    Article  PubMed  CAS  Google Scholar 

  73. Laskewitz A, van Benthem KL, Kieffer TEC, Faas MM, Verkaik-Schakel RN, Plösch T, Scherjon SA, Prins JR. The influence of maternal obesity on macrophage subsets in the human decidua. Cell Immunol. 2019;336:75–82.

    Article  PubMed  CAS  Google Scholar 

  74. Löb S, Knabl J, Vattai A, Schmoeckel E, Kuhn C, Mittelberger J, Wöckel A, Mahner S, Jeschke U. Obesity in pregnancy is associated with macrophage influx and an upregulated GRO-alpha and IL-6 expression in the decidua. J Reprod Immunol. 2023;156:103800.

    Article  PubMed  Google Scholar 

  75. Shook LL, James KE, Roberts DJ, Powe CE, Perlis RH, Thornburg KL, O’Tierney-Ginn PF, Edlow AG. Sex-specific impact of maternal obesity on fetal placental macrophages and cord blood triglycerides. Placenta. 2023;140:100–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Dalmas E, Venteclef N, Caer C, Poitou C, Cremer I, Aron-Wisnewsky J, Lacroix-Desmazes S, Bayry J, Kaveri SV, Clément K, et al. T cell-derived IL-22 amplifies IL-1β-driven inflammation in human adipose tissue: relevance to obesity and type 2 diabetes. Diabetes. 2014;63:1966–77.

    Article  PubMed  CAS  Google Scholar 

  77. Gong P, Shi B, Wang J, Cao P, Diao Z, Wang Y, Hu Y, Li S. Association between Th1/Th2 immune imbalance and obesity in women with or without polycystic ovary syndrome. Gynecol Endocrinol. 2018;34:709–14.

    Article  PubMed  CAS  Google Scholar 

  78. Cipolletta D, Kolodin D, Benoist C, Mathis D. Tissular T(regs): a unique population of adipose-tissue-resident Foxp3 + CD4 + T cells that impacts organismal metabolism. Semin Immunol. 2011;23:431–7.

    Article  PubMed  CAS  Google Scholar 

  79. Chen Y, Tian J, Tian X, Tang X, Rui K, Tong J, Lu L, Xu H, Wang S. Adipose tissue dendritic cells enhances inflammation by prompting the generation of Th17 cells. PLoS ONE. 2014;9:e92450.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Cho KW, Zamarron BF, Muir LA, Singer K, Porsche CE, DelProposto JB, Geletka L, Meyer KA, O’Rourke RW, Lumeng CN. Adipose tissue dendritic cells are independent contributors to obesity-Induced inflammation and insulin resistance. J Immunol. 2016;197:3650–61.

    Article  PubMed  CAS  Google Scholar 

  81. Stefanovic-Racic M, Yang X, Turner MS, Mantell BS, Stolz DB, Sumpter TL, Sipula IJ, Dedousis N, Scott DK, Morel PA, et al. Dendritic cells promote macrophage infiltration and comprise a substantial proportion of obesity-associated increases in CD11c + cells in adipose tissue and liver. Diabetes. 2012;61:2330–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Frasca D, Romero M, Diaz A, Blomberg BB. Obesity accelerates age defects in B cells, and weight loss improves B cell function. Immun Ageing. 2023;20:35.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Chen X, Liu Y, Cheung WC, Zhao Y, Huang J, Chung JPW, Wang CC, Li TC. Increased expression of angiogenic cytokines in CD56 + uterine natural killer cells from women with recurrent miscarriage. Cytokine. 2018;110:272–6.

    Article  PubMed  CAS  Google Scholar 

  84. Lu H, Yang HL, Zhou WJ, Lai ZZ, Qiu XM, Fu Q, Zhao JY, Wang J, Li DJ, Li MQ. Rapamycin prevents spontaneous abortion by triggering decidual stromal cell autophagy-mediated NK cell residence. Autophagy. 2021;17:2511–27.

    Article  PubMed  CAS  Google Scholar 

  85. Ahmadi M, Ghaebi M, Abdolmohammadi-Vahid S, Abbaspour-Aghdam S, Hamdi K, Abdollahi-Fard S, Danaii S, Mosapour P, Koushaeian L, Dolati S, et al. NK cell frequency and cytotoxicity in correlation to pregnancy outcome and response to IVIG therapy among women with recurrent pregnancy loss. J Cell Physiol. 2019;234:9428–37.

    Article  PubMed  CAS  Google Scholar 

  86. Tan HX, Yang SL, Li MQ, Wang HY. Autophagy suppression of trophoblast cells induces pregnancy loss by activating decidual NK cytotoxicity and inhibiting trophoblast invasion. Cell Commun Signal. 2020;18:73.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Hou Y, Liu Q, Jin D, Li J, Huang L, Qiao C. The predictive value of NKG2C + NK cells and LILRB1 + NK cells in recurrent spontaneous abortion. Am J Reprod Immunol. 2023;89:e13627.

    Article  PubMed  CAS  Google Scholar 

  88. Yang Y, Liu H, Zhao Y, Geng C, Chao L, Hao A. Grim-19 deficiency promotes decidual macrophage autophagy in recurrent spontaneous abortion. Front Endocrinol (Lausanne). 2022;13:1023194.

    Article  PubMed  Google Scholar 

  89. Yang HL, Lai ZZ, Shi JW, Zhou WJ, Mei J, Ye JF, Zhang T, Wang J, Zhao JY, Li DJ, Li MQ. A defective lysophosphatidic acid-autophagy axis increases miscarriage risk by restricting decidual macrophage residence. Autophagy. 2022;18:2459–80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  90. Yang L, Zhang X, Gu Y, Shi Y, Wang LB, Shi JX, Zhen XX, Xin YW, Gu WW, Wang J. Section 5 is involved in M2 polarization of macrophages via the STAT6 pathway, and its dysfunction in decidual macrophages is associated with recurrent spontaneous abortion. Front Cell Dev Biol. 2022;10:891748.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Cui L, Xu F, Wang S, Li X, Lin H, Ding Y, Du M. Pharmacological activation of rev-erbα suppresses LPS-induced macrophage M1 polarization and prevents pregnancy loss. BMC Immunol. 2021;22:57.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  92. Tsao FY, Wu MY, Chang YL, Wu CT, Ho HN. M1 macrophages decrease in the deciduae from normal pregnancies but not from spontaneous abortions or unexplained recurrent spontaneous abortions. J Formos Med Assoc. 2018;117:204–11.

    Article  PubMed  CAS  Google Scholar 

  93. Zhang J, Tao Y, Cai R, Wang Y. miR-196a-5p-Rich Extracellular Vesicles from Trophoblasts Induce M1 Polarization of Macrophages in Recurrent Miscarriage. J Immunol Res. 2022;2022:6811632.

  94. Yan S, Ding J, Wang Z, Zhang F, Li J, Zhang Y, Wu S, Yang L, Pang X, Zhang Y, Yang J. CTRP6 regulates M1 macrophage polarization via the PPAR-γ/NF-κB pathway and reprogramming glycolysis in recurrent spontaneous abortion. Int Immunopharmacol. 2023;124:110840.

    Article  PubMed  CAS  Google Scholar 

  95. Zhou WJ, Yang HL, Mei J, Chang KK, Lu H, Lai ZZ, Shi JW, Wang XH, Wu K, Zhang T, et al. Fructose-1,6-bisphosphate prevents pregnancy loss by inducing decidual COX-2(+) macrophage differentiation. Sci Adv. 2022;8:eabj2488.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. Kuroda K, Nakagawa K, Horikawa T, Moriyama A, Ojiro Y, Takamizawa S, Ochiai A, Matsumura Y, Ikemoto Y, Yamaguchi K, Sugiyama R. Increasing number of implantation failures and pregnancy losses associated with elevated Th1/Th2 cell ratio. Am J Reprod Immunol. 2021;86:e13429.

    Article  PubMed  CAS  Google Scholar 

  97. He M, Jiang M, Zhou Y, Li F, Yang M, Fan Y, Xie Y, Beejadhursing R, Feng L, Deng D. Impaired Gal-9 Dysregulates the PBMC-Induced Th1/Th2 Imbalance in Abortion-Prone Matings. J Immunol Res. 2018;2018:9517842.

  98. Ahmadi M, Abdolmohammadi-Vahid S, Ghaebi M, Aghebati-Maleki L, Afkham A, Danaii S, Abdollahi-Fard S, Heidari L, Jadidi-Niaragh F, Younesi V, et al. Effect of intravenous immunoglobulin on Th1 and Th2 lymphocytes and improvement of pregnancy outcome in recurrent pregnancy loss (RPL). Biomed Pharmacother. 2017;92:1095–102.

    Article  PubMed  CAS  Google Scholar 

  99. Huo S, Wu F, Zhang J, Wang X, Li W, Cui D, Zuo Y, Hu M, Zhong F. Porcine soluble CD83 alleviates LPS-induced abortion in mice by promoting Th2 cytokine production, Treg cell generation and trophoblast invasion. Theriogenology. 2020;157:149–61.

    Article  PubMed  CAS  Google Scholar 

  100. Liu B, Wu H, Huang Q, Li M, Fu X. Phosphorylated STAT3 inhibited the proliferation and suppression of decidual Treg cells in unexplained recurrent spontaneous abortion. Int Immunopharmacol. 2020;82:106337.

    Article  PubMed  CAS  Google Scholar 

  101. Wu L, Li J, Xu HL, Xu B, Tong XH, Kwak-Kim J, Liu YS. IL-7/IL-7R signaling pathway might play a role in recurrent pregnancy losses by increasing inflammatory Th17 cells and decreasing Treg cells. Am J Reprod Immunol. 2016;76:454–64.

    Article  PubMed  CAS  Google Scholar 

  102. Shu T, Zhao D, Li B, Wang Y, Liu S, Li P, Zuo J, Bai P, Zhang R, Wu L. Prognostic evaluation of postoperative adjuvant therapy for operable cervical cancer: 10 years’ experience of National Cancer Center in China. Chin J Cancer Res. 2017;29:510–20.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. Ehrentraut S, Sauss K, Neumeister R, Luley L, Oettel A, Fettke F, Costa SD, Langwisch S, Zenclussen AC, Schumacher A. Human miscarriage is Associated with dysregulations in Peripheral blood-derived myeloid dendritic cell subsets. Front Immunol. 2019;10:2440.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Huang C, Zhang H, Chen X, Diao L, Lian R, Zhang X, Hu L, Zeng Y. Association of peripheral blood dendritic cells with recurrent pregnancy loss: a case-controlled study. Am J Reprod Immunol. 2016;76:326–32.

    Article  PubMed  CAS  Google Scholar 

  105. Lai N, Fu X, Hei G, Song W, Wei R, Zhu X, Guo Q, Zhang Z, Chu C, Xu K, Li X. The Role of Dendritic Cell Subsets in Recurrent Spontaneous Abortion and the Regulatory Effect of Baicalin on It. J Immunol Res. 2022;2022:9693064.

  106. Zhu XX, Yin XQ, Hei GZ, Wei R, Guo Q, Zhao L, Zhang Z, Chu C, Fu XX, Xu K, Li X. Increased mir-6875-5p inhibits plasmacytoid dendritic cell differentiation via the STAT3/E2-2 pathway in recurrent spontaneous abortion. Mol Hum Reprod. 2021;27:gaab044.

  107. Danaii S, Ghorbani F, Ahmadi M, Abbaszadeh H, Koushaeian L, Soltani-Zangbar MS, Mehdizadeh A, Hojjat-Farsangi M, Kafil HS, Aghebati-Maleki L, Yousefi M. IL-10-producing B cells play important role in the pathogenesis of recurrent pregnancy loss. Int Immunopharmacol. 2020;87:106806.

    Article  PubMed  CAS  Google Scholar 

  108. Kirichenko TV, Markina YV, Bogatyreva AI, Tolstik TV, Varaeva YR, Starodubova AV. The role of Adipokines in inflammatory mechanisms of obesity. Int J Mol Sci. 2022;23:14982.

  109. St-Germain LE, Castellana B, Baltayeva J, Beristain AG. Maternal obesity and the Uterine Immune Cell Landscape: the shaping role of inflammation. Int J Mol Sci. 2020;21:3776.

  110. Faas MM, de Vos P. Uterine NK cells and macrophages in pregnancy. Placenta. 2017;56:44–52.

    Article  PubMed  CAS  Google Scholar 

  111. Liu W, Liu X, Luo M, Liu X, Luo Q, Tao H, Wu D, Lu S, Jin J, Zhao Y, Zou L. dNK derived IFN-γ mediates VSMC migration and apoptosis via the induction of LncRNA MEG3: a role in uterovascular transformation. Placenta. 2017;50:32–9.

    Article  PubMed  CAS  Google Scholar 

  112. Liu S, Diao L, Huang C, Li Y, Zeng Y, Kwak-Kim JYH. The role of decidual immune cells on human pregnancy. J Reprod Immunol. 2017;124:44–53.

    Article  PubMed  CAS  Google Scholar 

  113. Inada K, Shima T, Ito M, Ushijima A, Saito S. Helios-positive functional regulatory T cells are decreased in decidua of miscarriage cases with normal fetal chromosomal content. J Reprod Immunol. 2015;107:10–9.

    Article  PubMed  CAS  Google Scholar 

  114. Saifi B, Rezaee SA, Tajik N, Ahmadpour ME, Ashrafi M, Vakili R, SoleimaniAsl S, Aflatoonian R, Mehdizadeh M. Th17 cells and related cytokines in unexplained recurrent spontaneous miscarriage at the implantation window. Reprod Biomed Online. 2014;29:481–9.

    Article  PubMed  CAS  Google Scholar 

  115. Salvany-Celades M, van der Zwan A, Benner M, Setrajcic-Dragos V, Bougleux Gomes HA, Iyer V, Norwitz ER, Strominger JL, Tilburgs T. Three Types of Functional Regulatory T Cells Control T Cell Responses at the human maternal-fetal interface. Cell Rep. 2019;27:2537–e25472535.

    Article  PubMed  CAS  Google Scholar 

  116. Liao J, Li Y, Li X, Su X, Peng J, Xiao N, Fan X, Chen H, Lu G, Lin G, et al. Blood CD4(+)CD25(+) T regulatory cells constitute a potential predictive marker of subsequent miscarriage in unexplained recurrent pregnancy loss. Int Immunopharmacol. 2022;110:108960.

    Article  PubMed  CAS  Google Scholar 

  117. Guo Z, Xu Y, Zheng Q, Liu Y, Liu X. Analysis of chromosomes and the T helper 17 and regulatory T cell balance in patients with recurrent spontaneous abortion. Exp Ther Med. 2020;19:3159–66.

    PubMed  PubMed Central  CAS  Google Scholar 

  118. Ander SE, Diamond MS, Coyne CB. Immune responses at the maternal-fetal interface. Sci Immunol. 2019;4:1135.

  119. Jena MK, Nayak N, Chen K, Nayak NR. Role of macrophages in pregnancy and related complications. Arch Immunol Ther Exp (Warsz). 2019;67:295–309.

    Article  PubMed  CAS  Google Scholar 

  120. Challier JC, Basu S, Bintein T, Minium J, Hotmire K, Catalano PM, Hauguel-de Mouzon S. Obesity in pregnancy stimulates macrophage accumulation and inflammation in the placenta. Placenta. 2008;29:274–81.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  121. Gonçalves C, Feitosa BM, Cavalcante BV, Lima A, de Souza CM, Joventino LB, Cavalcante MB. Obesity and recurrent miscarriage: the interconnections between adipose tissue and the immune system. Am J Reprod Immunol. 2023;90:e13757.

    Article  PubMed  Google Scholar 

  122. Tersigni C, Di Nicuolo F, D’Ippolito S, Veglia M, Castellucci M. Di Simone N: Adipokines: new emerging roles in fertility and reproduction. Obstet Gynecol Surv. 2011;66:47–63.

    Article  PubMed  Google Scholar 

  123. D’Ippolito S, Tersigni C, Scambia G, Di Simone N. Adipokines, an adipose tissue and placental product with biological functions during pregnancy. BioFactors. 2012;38:14–23.

    Article  PubMed  Google Scholar 

  124. Liu L, Shi Z, Ji X, Zhang W, Luan J, Zahr T, Qiang L. Adipokines, adiposity, and atherosclerosis. Cell Mol Life Sci. 2022;79:272.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  125. Shaker M, Shalabi T, Gaber KR, Amr K. Association of miRNA-27a and leptin polymorphisms with recurrent pregnancy loss in Egyptian women. Meta Gene. 2020;24:100617.

  126. Plowden TC, Zarek SM, DeVilbiss EA, Radoc JG, Kim K, Sjaarda LA, Schisterman EF, Silver RM, Kuhr DL, Omosigho UR, et al. Preconception leptin and fecundability, pregnancy, and live birth among women with a history of pregnancy loss. J Endocr Soc. 2019;3:1958–68.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Müller A, Wagner J, Hodžić A, Maver A, Škrlec I, Heffer M, Zibar L, Peterlin B. Genetic variation in leptin and leptin receptor genes is a risk factor for idiopathic recurrent spontaneous abortion. Croat Med J. 2016;57:566–71.

    Article  PubMed  PubMed Central  Google Scholar 

  128. Pheiffer C, Dias S, Jack B, Malaza N, Adam S. Adiponectin as a potential biomarker for pregnancy disorders. Int J Mol Sci. 2021;22:1326.

  129. Serazin V, Duval F, Wainer R, Ravel C, Vialard F, Molina-Gomes D, Dieudonne MN, Dos Santos E. Are leptin and adiponectin involved in recurrent pregnancy loss? J Obstet Gynaecol Res. 2018;44:1015–22.

    Article  PubMed  CAS  Google Scholar 

  130. Cheng L, Shi H, Jin Y, Li X, Pan J, Lai Y, Lin Y, Jin Y, Roy G, Zhao A, Li F. Adiponectin Deficiency leads to female subfertility and ovarian dysfunctions in mice. Endocrinology. 2016;157:4875–87.

    Article  PubMed  CAS  Google Scholar 

  131. Li W, Geng L, Liu X, Gui W, Qi H. Recombinant adiponectin alleviates abortion in mice by regulating Th17/Treg imbalance via p38MAPK-STAT5 pathway. Biol Reprod. 2019;100:1008–17.

    Article  PubMed  Google Scholar 

  132. Wang P, Zhu Q, Peng H, Du M, Dong M, Wang H. Fatty acid-binding protein 4 in endometrial epithelium is involved in embryonic implantation. Cell Physiol Biochem. 2017;41:501–9.

    Article  PubMed  CAS  Google Scholar 

  133. Pandya AD, Das MK, Sarkar A, Vilasagaram S, Basak S, Duttaroy AK. Tube formation in the first trimester placental trophoblast cells: Differential effects of angiogenic growth factors and fatty acids. Cell Biol Int. 2016;40:652–61.

    Article  PubMed  CAS  Google Scholar 

  134. Chung JY, Hong J, Kim HJ, Song Y, Yong SB, Lee J, Kim YH. White adipocyte-targeted dual gene silencing of FABP4/5 for anti-obesity, anti-inflammation and reversal of insulin resistance: efficacy and comparison of administration routes. Biomaterials. 2021;279:121209.

    Article  PubMed  CAS  Google Scholar 

  135. Dina C, Meyre D, Gallina S, Durand E, Körner A, Jacobson P, Carlsson LM, Kiess W, Vatin V, Lecoeur C, et al. Variation in FTO contributes to childhood obesity and severe adult obesity. Nat Genet. 2007;39:724–6.

    Article  PubMed  CAS  Google Scholar 

  136. An Y, Duan H. The role of m6A RNA methylation in cancer metabolism. Mol Cancer. 2022;21:14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  137. Ponce-Gonzalez JG, Martínez-Ávila Á, Velázquez-Díaz D, Perez-Bey A, Gómez-Gallego F, Marín-Galindo A, Corral-Pérez J, Casals C. Impact of the FTO Gene Variation on Appetite and Fat Oxidation in Young adults. Nutrients. 2023;15:2037.

  138. Speakman JR. The ‘Fat Mass and obesity related’ (FTO) gene: mechanisms of impact on obesity and energy balance. Curr Obes Rep. 2015;4:73–91.

    Article  PubMed  Google Scholar 

  139. Fawcett KA, Barroso I. The genetics of obesity: FTO leads the way. Trends Genet. 2010;26:266–74.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  140. Chen X, Luo Y, Jia G, Liu G, Zhao H, Huang Z. FTO promotes adipogenesis through inhibition of the Wnt/β-catenin signaling pathway in Porcine Intramuscular Preadipocytes. Anim Biotechnol. 2017;28:268–74.

    Article  PubMed  CAS  Google Scholar 

  141. Andraweera PH, Dekker GA, Jayasekara RW, Dissanayake VHW, Roberts CT. The obesity-related FTO gene variant associates with the risk of recurrent miscarriage. Acta Obstet Gynecol Scand. 2015;94:722–6.

    Article  PubMed  CAS  Google Scholar 

  142. Qiu W, Zhou Y, Wu H, Lv X, Yang L, Ren Z, Tian H, Yu Q, Li J, Lin W, et al. RNA demethylase FTO mediated RNA m(6)a modification is involved in maintaining maternal-fetal interface in spontaneous abortion. Front Cell Dev Biol. 2021;9:617172.

    Article  PubMed  PubMed Central  Google Scholar 

  143. Zhang J, Liu X, Gao Y. FTO protein regulates the TGF-β signalling pathway through RNA N6-methyladenosine modification to induce unexplained recurrent spontaneous abortion. Febs j. 2024;291:1545–59.

    Article  PubMed  CAS  Google Scholar 

  144. Busnelli A, Garolla A, Di Credico E, D’Ippolito S, Merola AM, Milardi D, Pontecorvi A, Scambia G, Di Simone N. Sperm DNA fragmentation and idiopathic recurrent pregnancy loss: results from a multicenter case-control study. Andrology. 2023;11:1673–81.

    Article  PubMed  CAS  Google Scholar 

  145. Peuranpää PL, Gissler M, Peltopuro P, Tiitinen A, Hautamäki H. The effect of paternal and maternal factors on the prognosis of live birth in couples with recurrent pregnancy loss. Acta Obstet Gynecol Scand. 2022;101:1374–85.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Haddock L, Gordon S, Lewis SEM, Larsen P, Shehata A, Shehata H. Sperm DNA fragmentation is a novel biomarker for early pregnancy loss. Reprod Biomed Online. 2021;42:175–84.

    Article  PubMed  CAS  Google Scholar 

  147. Hedegger K, Philippou-Massier J, Krebs S, Blum H, Kunzelmann S, Förstemann K, Gimpfl M, Roscher AA, Ensenauer R, Wolf E, Dahlhoff M. Sex-specific programming effects of parental obesity in pre-implantation embryonic development. Int J Obes (Lond). 2020;44:1185–90.

    Article  PubMed  CAS  Google Scholar 

  148. Deshpande SSS, Bera P, Khambata K, Balasinor NH. Paternal obesity induces epigenetic aberrations and gene expression changes in placenta and fetus. Mol Reprod Dev. 2023;90:109–26.

    Article  PubMed  CAS  Google Scholar 

  149. Keyhan S, Burke E, Schrott R, Huang Z, Grenier C, Price T, Raburn D, Corcoran DL, Soubry A, Hoyo C, Murphy SK. Male obesity impacts DNA methylation reprogramming in sperm. Clin Epigenetics. 2021;13:17.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  150. Wei S, Luo S, Zhang H, Li Y, Zhao J. Paternal high-fat diet altered SETD2 gene methylation in sperm of F0 and F1 mice. Genes Nutr. 2023;18:12.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  151. Wu Y, Li J, Li C, Lu S, Wei X, Li Y, Xia W, Qian C, Wang Z, Liu M, et al. Fat mass and obesity-associated factor (FTO)-mediated N6-methyladenosine regulates spermatogenesis in an age-dependent manner. J Biol Chem. 2023;299:104783.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  152. Pereira SC, Martins AD, Monteiro MP, Pinto S, Barros A, Oliveira PF, Alves MG. Expression of obesity-related genes in human spermatozoa affects the outcomes of reproductive treatments. F S Sci. 2021;2:164–75.

    PubMed  Google Scholar 

  153. Almutairi H, Aldhalea MS, Almaaz MA, Aljuhani SA, Aloraini RI, Alamoudi AA, Alkhalifah WF, Alrushaid LA, Alanzy HW, Alzuwayyid M et al. The effectiveness of bariatric surgery on treating infertility in Women-A systematic review and Meta-analysis. J Clin Med. 2024;13:5569.

  154. Kort JD, Winget C, Kim SH, Lathi RB. A retrospective cohort study to evaluate the impact of meaningful weight loss on fertility outcomes in an overweight population with infertility. Fertil Steril. 2014;101:1400–3.

    Article  PubMed  Google Scholar 

  155. Yang C, Yang S, Zheng W, Zu R, Ran S, Wu H, Ren B, Lv N, Kuang Y, Li M, et al. Effect of a 60-day weight reduction intervention prior to IVF/ICSI on perinatal outcomes in overweight or obese infertile women. Front Endocrinol (Lausanne). 2022;13:1062790.

    Article  PubMed  Google Scholar 

  156. Tersigni C, Neri C, D’Ippolito S, Garofalo S, Martino C, Lanzone A, Scambia G, Di Simone N. Impact of maternal obesity on the risk of preterm delivery: insights into pathogenic mechanisms. J Matern Fetal Neonatal Med. 2022;35:3216–21.

    Article  PubMed  Google Scholar 

  157. Legro RS, Hansen KR, Diamond MP, Steiner AZ, Coutifaris C, Cedars MI, Hoeger KM, Usadi R, Johnstone EB, Haisenleder DJ, et al. Effects of preconception lifestyle intervention in infertile women with obesity: the FIT-PLESE randomized controlled trial. PLoS Med. 2022;19:e1003883.

    Article  PubMed  PubMed Central  Google Scholar 

  158. Obesity. Reproduction: a committee opinion. Fertil Steril. 2021;116:1266–85.

    Article  Google Scholar 

  159. Espinós JJ, Solà I, Valli C, Polo A, Ziolkowska L, Martínez-Zapata MJ. The Effect of Lifestyle intervention on pregnancy and birth outcomes on obese Infertile women: a systematic review and Meta-analysis. Int J Fertil Steril. 2020;14:1–9.

    PubMed  PubMed Central  Google Scholar 

  160. Zhang J, Zhang R, Chi J, Li Y, Bai W. Pre-pregnancy body mass index has greater influence on newborn weight and perinatal outcome than weight control during pregnancy in obese women. Arch Public Health. 2023;81:5.

    Article  PubMed  PubMed Central  Google Scholar 

  161. Lim S, Harrison C, Callander E, Walker R, Teede H, Moran L. Addressing obesity in preconception, pregnancy, and Postpartum: a review of the literature. Curr Obes Rep. 2022;11:405–14.

    Article  PubMed  PubMed Central  Google Scholar 

  162. Caldwell AE, Gorczyca AM, Bradford AP, Nicklas JM, Montgomery RN, Smyth H, Pretzel S, Nguyen T, DeSanto K, Ernstrom C, Santoro N. Effectiveness of preconception weight loss interventions on fertility in women: a systematic review and meta-analysis. Fertil Steril. 2024;122:326–40.

    Article  PubMed  Google Scholar 

  163. Robertson N, Ladlow B. Effect of individual dietetic intervention on gestational weight gain and associated complications in obese pregnant women. Aust N Z J Obstet Gynaecol. 2018;58:274–7.

    Article  PubMed  Google Scholar 

  164. O’Brien EC, Geraghty AA, O’Sullivan EJ, Riordan JA, Horan MK, Larkin E, Donnelly J, Mehegan J, Twomey PJ, McAuliffe FM. Five-year follow up of a low glycaemic index dietary randomised controlled trial in pregnancy-no long-term maternal effects of a dietary intervention. BJOG. 2019;126:514–24.

    Article  PubMed  Google Scholar 

  165. Salas-Salvadó J, Díaz-López A, Ruiz-Canela M, Basora J, Fitó M, Corella D, Serra-Majem L, Wärnberg J, Romaguera D, Estruch R, et al. Effect of a lifestyle intervention program with Energy-Restricted Mediterranean Diet and Exercise on Weight loss and Cardiovascular Risk factors: one-year results of the PREDIMED-Plus trial. Diabetes Care. 2019;42:777–88.

    Article  PubMed  Google Scholar 

  166. Casas I, Nakaki A, Pascal R, Castro-Barquero S, Youssef L, Genero M, Benitez L, Larroya M, Boutet ML, Casu G et al. Effects of a Mediterranean Diet intervention on maternal stress, Well-Being, and Sleep Quality throughout Gestation-the IMPACT-BCN trial. Nutrients. 2023;15:2362.

  167. Flor-Alemany M, Nestares T, Alemany-Arrebola I, Marín-Jiménez N, Borges-Cosic M, Aparicio VA. Influence of Dietary habits and Mediterranean Diet adherence on Sleep Quality during pregnancy. The GESTAFIT Project. Nutrients. 2020;12:3569.

  168. Paskulin JTA, Drehmer M, Olinto MT, Hoffmann JF, Pinheiro AP, Schmidt MI, Nunes MA. Association between dietary patterns and mental disorders in pregnant women in Southern Brazil. Braz J Psychiatry. 2017;39:208–15.

    Article  PubMed  PubMed Central  Google Scholar 

  169. Yuan ZM, Li M, Ji CY, Li L, Jia L, Incecik A. Steady hydrodynamic interaction between human swimmers. J R Soc Interface. 2019;16:20180768.

    Article  PubMed  PubMed Central  Google Scholar 

  170. Ehrlich SF, Hedderson MM, Brown SD, Sternfeld B, Chasan-Taber L, Feng J, Adams J, Ching J, Crites Y, Quesenberry CP, Ferrara A. Moderate intensity sports and exercise is associated with glycaemic control in women with gestational diabetes. Diabetes Metab. 2017;43:416–23.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  171. Perales M, Valenzuela PL, Barakat R, Cordero Y, Peláez M, López C, Ruilope LM, Santos-Lozano A, Lucia A. Gestational Exercise and Maternal and Child Health: effects until Delivery and at Post-natal follow-up. J Clin Med. 2020;9:379.

  172. Poyatos-León R, García-Hermoso A, Sanabria-Martínez G, Álvarez-Bueno C, Cavero-Redondo I, Martínez-Vizcaíno V. Effects of exercise-based interventions on postpartum depression: a meta-analysis of randomized controlled trials. Birth. 2017;44:200–8.

    Article  PubMed  Google Scholar 

  173. Artal R, O’Toole M. Guidelines of the American College of obstetricians and gynecologists for exercise during pregnancy and the postpartum period. Br J Sports Med. 2003;37:6–12. discussion 12.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  174. Azward H, Ramadhany S, Pelupessy N, Usman AN, Bara FT. Prenatal yoga exercise improves sleep quality in the third trimester of pregnant women. Gac Sanit. 2021;35(Suppl 2):S258–62.

    Article  PubMed  Google Scholar 

  175. Wartena R, Matjila M. Polycystic ovary syndrome and recurrent pregnancy loss, a review of literature. Front Endocrinol (Lausanne). 2023;14:1183060.

    Article  PubMed  Google Scholar 

  176. D’Ippolito S, Capozzi A, Scambia G, Sorge R, Lello S, Simone ND. Glucose/insulin metabolism and vitamin D in women with recurrent pregnancy loss. Am J Reprod Immunol. 2022;87:e13505.

    Article  PubMed  Google Scholar 

  177. Wang M, Zhang P, Yu S, Zhou G, Lv J, Nallapothula D, Guo C, Wang Q, Singh RR. Heparin and aspirin combination therapy restores T-cell phenotype in pregnant patients with antiphospholipid syndrome-related recurrent pregnancy loss. Clin Immunol. 2019;208:108259.

    Article  PubMed  CAS  Google Scholar 

  178. Blue NR, Page JM, Silver RM. Genetic abnormalities and pregnancy loss. Semin Perinatol. 2019;43:66–73.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by cross-innovation talent project in Renmin Hospital of Wuhan University (grant number JCRCZN-2022-016); Undergraduate education quality construction comprehensive reform project (grant number 2022ZG282) and the National Natural Science Foundation of China (grant number 82071655).

Author information

Authors and Affiliations

Authors

Contributions

RQW conceived and designed the study. RQW performed literature search. ZMD and FFD drafted the manuscript and prepared the tables. DZM revised the manuscript and FFD acquired funding. LBX and GTC provided significant assistance and guidance during the revision process of the manuscript. All authors read and approved the final manuscript. Data authentication is not applicable.

Corresponding authors

Correspondence to Gan-Tao Chen, Fang-Fang Dai or Liang-Bin Xia.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

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.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, RQ., Deng, ZM., Chen, GT. et al. Obesity and recurrent spontaneous abortion: the crucial role of weight management in pregnancy. Reprod Biol Endocrinol 23, 10 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12958-024-01326-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12958-024-01326-3

Keywords