Next Article in Journal
Inverse Vaccination for Autoimmune Diseases: Insights into the Role of B Lymphocytes
Previous Article in Journal
Rapamycin-Reactivated Lipid Catabolism in Eruca sativa Mill. Exposed to Salt Stress
Previous Article in Special Issue
Placental Inflammation in Preterm Premature Rupture of Membranes and Risk of Neurodevelopmental Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Sex Difference in the Pathophysiology of Preterm Birth

1
Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
2
Division of Clinical Medicine, School of Medicine & Population Health, Faculty of Health, The University of Sheffield, Jessop Wing, Tree Root Walk, Sheffield S10 2SF, UK
*
Authors to whom correspondence should be addressed.
Cells 2025, 14(14), 1084; https://doi.org/10.3390/cells14141084
Submission received: 20 June 2025 / Revised: 12 July 2025 / Accepted: 14 July 2025 / Published: 16 July 2025
(This article belongs to the Special Issue Molecular Insight into the Pathogenesis of Spontaneous Preterm Birth)

Abstract

Preterm birth (PTB) refers to a labor before 37 gestational weeks. This is a major global contributor to neonatal morbidity and mortality. Although fetal sex is frequently treated as a confounding variable in PTB research, relatively few studies have conducted sex-stratified analyses to investigate how male and female fetuses may respond differently to various intrauterine exposures. This represents an underexplored area with important implications for understanding fetal sexual dimorphism-specific vulnerability to adverse pregnancy outcomes. Understanding the role of fetal sex differences in the pathophysiology of preterm birth (PTB) regarding processes such as inflammation, placental dysfunction, and oxidative stress is crucial. These delicate processes are tightly interrelated, but also independently contribute to pregnancy complications. Recognizing fetal sex as a biological variable for such processes is essential for improving mechanistic insight, providing refined predictive models.

1. Introduction

Preterm birth (PTB), defined by the World Health Organization as a delivery before 37 weeks of gestation [1], affects 9.9% of births worldwide and remains a leading cause of neonatal mortality and morbidity [2]. The insufficient development of the placenta contributes to PTB, and resulting newborns are at risk of short-term complications such as long-term physical and neurodevelopmental disorders, which are noted to affect male infants disproportionately [3].
Approximately two-thirds of PTBs are spontaneous PTBs (sPTB), while the remaining 30–35% of PTBs are medically indicated, sometimes associated with conditions such as preterm premature rupture of the membrane (pPROM), intrauterine infections, or inflammation-driven preterm labor [4,5,6]. These proportions may vary based on population characteristics and geographic regions. Most studies report high negative predictive values of these tests, but their positive predictive values remain low and inconsistent.
Emerging evidence suggests that PTB results from a complex interplay of multiple pathologic mechanisms, including ascending infection triggered by systemic inflammation, vaginal dysbiosis, endocrine dysregulation, and oxidative stress [7,8,9,10,11]. These pathologies are further influenced by environmental factors (e.g., particulate matter, microplastics, heavy metals, etc.), maternal behaviors (e.g., smoking, alcohol consumption, and high-fat diet), stress, socioeconomic status, and a previous PTB history [12,13,14].
Recent studies also suggest that maternal endocrine fluctuations—such as altered levels of progesterone (P4), estrogen, oxytocin, and fetal sex hormones—may contribute to sPTBs [4,15]. However, fetal sex as a biological confounding variable is still underrecognized in many observational studies, despite the accumulating evidence that male fetuses are more susceptible to adverse intrauterine conditions [16].
In this review, we aim to synthesize the current knowledge on the sex-specific mechanisms underlying PTBs, with a particular focus on how inflammation, placental dysfunction, and oxidative stress differentially affect pregnancy outcomes based on fetal sex (Figure 1).

2. Environmental Factors Influence Preterm Birth Prevalence According to Fetal Sex

The “Developmental Origins of Health and Disease (DOHaD)” hypothesis posits that prenatal exposure to environmental and physiological stressors can program long-term health outcomes [17]. Prenatal exposures, including environmental pollutants and endocrine disruptors, have been shown to influence not only fetal development but also the sex ratio at birth [18,19]. A large-scale cohort study from the Netherlands involving 1,736,615 singleton pregnancies found that a male fetus is a relevant risk factor for sPTB with intact membranes [20]. Additionally, male neonates in low-risk populations exhibit lower umbilical cord blood pH and lower APGAR scores at both 1 and 5 min than their female counterparts, indicating greater vulnerability to perinatal stress [21]. This phenomenon presents a similar tendency to neonatal adverse outcomes. Low birth weight [22], respiratory distress syndrome (RDS) [23], bronchopulmonary dysplasia (BPD) [24], and periventricular–intraventricular hemorrhage (PIVH) [25] showed higher prevalence in male infants.
Sex-based disparities in pregnancy outcomes have been consistently observed across various physiological domains (Table 1). Sex- and age-related brain differences showed that males have larger overall volumes and more white matter. In contrast, females exhibit more cortical gray matter and region-specific increases, particularly in the left anterior cingulate and superior temporal gyrus [26]. At the molecular level, prenatal exposure to air pollution is associated with the heightened activation of immune pathways in females, while males demonstrate an increased expression of genes related to synaptic signaling and mitochondrial function during the last month of gestation [27]. These findings suggest distinct biological strategies in male and female fetuses when adapting to intrauterine stress.
Despite the growing evidence, the impact of fetal sex on pregnancy complications and environmental interactions remains underexplored in many studies. The greater consideration of sex as a biological variable is essential for advancing our understanding of PTB pathogenesis, and for developing sex-specific preventive strategies.

3. Sex-Specific Modulation of Inflammation in Preterm Birth

3.1. Inflammatory Mechanism in Pregnancy

Among several physiological processes involved in pregnancy, inflammation plays a paradoxical role. While excessive or dysregulated inflammation is the primary trigger for the onset of PTB [35], controlled inflammation responses are essential for implantation and the successful maintenance of pregnancy, as well as labor in humans, mice, and opossums [36].
The cytokines, chemokines, and prostaglandins present in the seminal fluid initiate the recruitment of leukocytes and stimulate pro-inflammatory cytokines within the uterus [37,38]. During normal pregnancy, the maternal immune tolerance mechanism suppresses fetal rejection by recognizing paternal antigens expressed in fetal tissues. Maternal immune tolerance has been shown to mitigate the risk of fetal rejection and supports the remodeling of spiral arteries (SpAs) and placentation [35]. While physiological inflammation supports implantation and placentation, an imbalanced or dysregulated pathway can shift immune responses toward pathological outcomes, such as PTB.

3.2. Inflammatory Dysregulation and Preterm Birth

The onset of parturition is associated with a physiologic inflammatory cascade [39]. One of the critical components in this process is the activation of toll-like receptors (TLRs), which detect pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) originating from microbes and necrotic cells, respectively [40,41]. This TLR activation initiates inflammasome formation, which stimulates the increased expression of pro-inflammatory cytokines, chemokines (e.g., tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, IL-8, and granulocyte–macrophage colony-stimulating factor (GM-CSF)) and leukocytes (macrophages, monocytes, and neutrophils). Concurrently, anti-inflammatory cytokines (IL-10, TGF-β, etc.) are decreased in the placenta and amniotic cavity [42,43,44,45].
The microbial invasion of the amniotic cavity (MIAC) or intra-amniotic infection, often following ascending infection from the lower genital tract, can lead to sterile inflammation and pPROM, and ultimately PTB [46,47,48]. A recent study demonstrated that TLR4 signaling in terms of endothelial cells can induce IL-6 secretion in perivascular stromal cells via nuclear factor kappa-light-chain-enhancer of activated B cell (NF-κb) activation, which also involves the IL-10/STAT3 axis [49,50]. NF-κb activation induces genes associated with uterine contractility and cervical ripening, such as the prostaglandin (PG) F2α receptor, connexin-43, the oxytocin receptor, and cyclooxygenase 2 (COX-2). The accumulation of pro-inflammatory cytokines promotes PG synthesis and membrane rupture—key events in the pathophysiology of pPROM and PTB [51,52].

3.3. Fetal Sex-Based Modulation of Inflammatory Responses

3.3.1. Sex Differences in Immune Regulation

Not only does maternal inflammation signaling contribute, but the fetus itself plays a role in initiating the signal of labor. In particular, immune responses in utero are influenced by both the sex chromosome complements (XX vs. XY) and sex steroid hormones (estrogen, progesterone, and androgen) [53]. According to a comprehensive review by Klein et al., males and females exhibit distinct immunological profiles across the life stages, including fetal development [54]. These differences affect the magnitude and type of both innate and adaptive immune responses and can influence the risk of PTB (Figure 2).

3.3.2. Sex Chromosomal Contribution to Immune Expression

The Y chromosome contains the sex-determining region Y (SRY) gene, which drives testis development and testosterone synthesis. In its absence, XY– (SRY- deficient) mice develop as phenotypic females with ovaries despite having a male chromosomal complement [55]. This difference may be related to decreased Th2 cytokines (IL-4, IL-5, IL-13), and increased levels of IL-13Rα2 on macrophages (CD11b+) and dendritic cells (CD11c+) [55]. In a mouse model using SRY-deficient males, CD4+ T cells showed a significantly increased expression of the histone demethylase KDM6A, which is implicated in autoimmune disease [56,57]. Furthermore, as men age, the Y chromosome may diminish from their immune cells, which increases their risk of death from cancer [58]. The lower TLR7 expression in male cells has been associated with increased susceptibility to infections such as SARS-CoV-2 and neonatal respiratory syncytial virus [59,60]. Abdel-Hafiz et al. revealed that male patients with a Y loss mutation promote the severity and number of other mutations, increasing the dysfunction of CD8+ T cells [61].
Conversely, in the case of females, a pair of X chromosomes is inherited—one maternal and one paternal—resulting in a diverse set of gene expressions [62]. To compensate for the imbalance due to double X chromosomes, one X chromosome undergoes random inactivation during early embryogenesis, a process known as X chromosome inactivation (XCI) [63]. This results in a cellular mosaicism state, allowing a balanced gene expression irrespective of sex [64]. Escape from XCI in certain loci contributes to allelic diversity, particularly in immune genes, and influences inflammatory responses [65]. Single-cell RNA sequencing identified 24 and 49 candidate escapees from fibroblasts and lymphoblasts, respectively [66]. The TLR7, a crucial component in antiviral defense, escapes XCI and is expressed at higher levels in female cells [67]. Taken together, these sexually dimorphic immune responses contribute to altered immune expression and disease susceptibility through sex chromosome-linked genetic and epigenetic differences.

3.3.3. PTB-Related Immune Modulation by Fetal Sex Difference Manner

Evidence has suggested that both type I and II interferon signaling, as well as humoral responses, are more robust in females than in males across several species [53]. For instance, cord blood immunoglobulin (Ig) E levels—a marker for allergic sensitization—are typically higher in male neonates than in females [68]. In a clinical study, men who were treated with gonadotropin-releasing hormone (GnRH) antagonist experienced a decrease in Treg (CD4+CD25+) without an alteration in the CD4+:CD8+ ratio. Additionally, medical castration increased natural killer (NK) cell activity while reducing IFN-γ production in CD8+ T cells [69], indicating that the hormonal milieu significantly shapes immune cell profiles. Female trophoblasts show a sensitivity in response to various compounds, displaying upregulated chemokines, such as CCL3, CCL4, and CXCL8 [70]. The migration of NK cells and monocytes is regulated by the binding of CCL3 and CCL4 to decidua-expressed receptors CCR1 and CCR5, respectively. Sun et al. suggested that the Th2 response is induced by increased M2, provoked by raised CCL13 and RGS1 in female Hofbauer cells. CCRL2, LGALS13, and LGALS14, which are regarded as regulating mitochondria, immune, and pregnancy maintenance-related transcripts, were highly upregulated on chromosome 19 in male chorionic villus between 11 and 16 weeks of gestation [71]. In male trophoblast cells, HLA-C, MUC15, NOTUM, SNHG19, and SNHG25 were upregulated [70]. The higher vulnerability of male neonates to infections, such as neonatal sepsis and meningitis, further highlights inherent sex-based differences in immune competence [72].
Furthermore, 16s rRNA sequencing studies have suggested that the composition of the vaginal microbiome is associated with preterm or term birth [73,74]. A high risk of PTB might be associated with a lack of Lactobacillus spp. in the cervicovaginal fluid [74,75]. N-glycosylation, which is a major immune-modulatory feature of the cervicovaginal fluid component, is influenced by pregnancy and immune state [76]. Estrogens, which increase during pregnancy, can modulate IgG glycosylation [77]. Since glycosylation patterns affect microbial adhesion to epithelial cells [78], the estrogen-mediated accumulation of glycogen in the vaginal epithelium may indirectly regulate microbial colonization and immune responses. Together, sex-specific immune modulation—mediated by interferon signaling, hormonal regulation, and trophoblast gene expression—contributes to the differential susceptibility to PTB. Additionally, cervicovaginal microbiome composition and glycosylation profiles may further reflect fetal sex-dependent differences.

4. Placental Dysfunction in Preterm Brith

4.1. Placental Formation and Function in Pregnancy

The placenta is a critical organ that forms 5–6 days after fertilization and serves as the interface between the maternal and fetal environments throughout gestation [79]. After implantation, trophoblast cells differentiate into two distinct lineages: villous and extravillous [80]. Villous cytotrophoblasts fuse to form multinucleated syncytiotrophoblasts, which comprise the outer epithelial layer of the chorionic villi and play a key role in nutrient change and hormone secretion [81]. The SpAs undergo extensive remodeling from early pregnancy through to approximately 22 weeks of gestation to facilitate proper placental perfusion [82].
The placenta is responsible for the exchange of oxygen, nutrients, hormones, and waste along with cells, and various signaling molecules, including nucleic acid, extracellular vesicles (EVs), and exosomes [80,83]. The syncytiotrophoblasts also produce steroid hormones, glycoproteins, and cytokines, all of which are essential for fetal development and immune modulation [84]. For instance, 17β-estradiol and P4 regulate changes in the endometrium and influence the expression of adhesion molecules, growth factors, and human chorionic gonadotropin (hCG), thereby facilitating successful implantation [85]. hCG, in turn, stimulates the ovarian secretion of P4, which maintains the secretory activity of the endometrium and downregulates progesterone receptor A (PRA) expression on maternal epithelial cells [79,84].

4.2. Placenta Dysfunction and Role in Preterm Brith

Placental dysfunction—also referred to as placental insufficiency—occurs when the placenta fails to adequately provide oxygen and nutrients to a developing fetus [86]. The dysfunction is recognized as a key contributor to adverse pregnancy outcomes, including PTB, miscarriage, stillbirth, and placental abruption [3,86,87,88]. In particular, impaired placental development or the insufficient remodeling of SpAs leads to increased vascular resistance and decreased placental blood flow [89], which may trigger disorders such as preeclampsia (PE), a hypertensive condition associated with proteinuria during pregnancy [90].
Placental dysfunction is affected by not only inflammation but also oxidative stress. Interestingly, placental dysfunction also induces inflammation and/or oxidative stress. In normal gestation, placental senescence is a progressive process, with syncytiotrophoblasts showing increasing markers of oxidative stress and cellular aging [91]. Placental tissues from cesarean sections showed elevated inflammation markers (IL-1α/β, IL-6, IL-8, CCL2, and TNF-α) when they were exposed to a state of hypoxia [92]. Oxidative stress, along with the activation of pathways such as G-protein-coupled estrogen receptor 1 (GPER1), has been implicated in advancing placental senescence [91,93]. Furthermore, placental dysfunction is closely associated with pPROM and placental abruption, a major cause of sPTB [88,93,94,95].

4.3. Fetal Sex Hormone Contributing to Placental Dysfunction

4.3.1. Sex Difference Effects on Placental Structure

Sex-specific genetic and epigenetic mechanisms significantly influence placental structural development. The differential expression of genes on the X and Y chromosomes contributes to variations in placental architecture and cellular composition. For instance, male placentas exhibit a relative downregulation of the ITGβ8 gene, which promotes angiogenesis and tissue invasion, both of which are vital during early gestation [96]. This downregulation may impair placental vascularization and nutrient exchange capacity, potentially predisposing male fetuses to suboptimal intrauterine environments.
Further transcriptomic profiling has revealed that the expression of genes related to immune regulation, such as those involved in graft-versus-host disease and inflammatory responses, is elevated in male placental villi [97,98]. This higher expression of genes related to immune tolerance and pregnancy maintenance in female placentas further suggests that male fetuses may be more vulnerable to early pregnancy loss due to impaired placentation [99].
The development of sequence technologies has reinforced more detailed transcriptional and epigenetic analyses of placental proportion. Sadiqi et al. used 450K DNA methylation to investigate whether the timing of PM2.5 exposure during pregnancy differentially affects the overall placental cell composition by fetal sex [100]. In male infants, first-trimester (T1) PM2.5 exposure was associated with a decreased proportion of syncytiotrophoblasts and increased trophoblasts. In females, second- (T2) and third- (T3) trimester exposure led to a decreased proportion of nucleated red blood cells (nRBCs) [100]. Rodent studies provide further evidence to support this assertion. At embryonic day (E) 15, the placental labyrinth zone fetal and maternal blood space had reduced volume in all compartments compared with gestational age-matched males [101]. The reduced E15 female labyrinth volume was associated with an overall decrease in the expression of differentiation- and growth-related genes, including MEST, GCM1, SYNA, insulin-like growth factor (IGF)2, and IGF2r [101].
Comprehensively, sex-specific genetic, epigenetic, and hormonal influences affect structural and cellular differences in the developing placenta, and male fetuses are more susceptible to placental dysfunction and intrauterine disease due to impaired angiogenesis and immune regulation.

4.3.2. Growth Strategy According to Fetal Sex

Sexual dimorphism in fetal adaptive strategies has been extensively documented, with compelling evidence indicating that male and female fetuses exhibit divergent growth responses to intrauterine stressors. Male fetuses tend to adopt a “growth-priority” mode, maintaining somatic development even under suboptimal placental function. In contrast, female fetuses more often implement a “placental adaptation” strategy, showing growth in favor of sustaining viability in adverse conditions [102,103].
Sex chromosome complement (XX vs. XY) significantly contributes to placental development and the concentration of circulating sex hormones [53]. The distinction is partly rooted in the early expression of the SRY gene, which initiates testis development and testosterone production before significant estrogenic activity arises [104,105]. These sex-specific hormonal profiles may contribute to divergent placental responses, with male placentas displaying reduced resilience under conditions of maternal stress (Figure 3).
Epidemiological studies support this; female fetuses are more commonly associated with preterm PE (delivered < 37 weeks of gestation) and very preterm PE (delivered < 34 weeks of gestation) [106]. However, term PE (delivered > 37 week of gestation) is more often linked to male fetuses [16]. These differences may mean that pregnancies susceptible to PE due to poor placental development are more prone to result in miscarriage if the fetus is male [106]. Consequently, surviving male fetuses may potentially represent a healthier subset, explaining the female predominance in preterm PE and male predominance in term PE. Interestingly, maternal post-reproductive lifespan may also be affected by fetal sex. Several studies have suggested that bearing male offspring may reduce maternal longevity, possibly due to greater physiological demands during pregnancy [107,108].
The placental abruption accounts for approximately 5% of all PTBs [109], and a male fetus has been identified as a significant risk factor (OR 1.38; 95% CI 1.12–1.70) [110]. Furthermore, spontaneous abortion risk is approximately 30% higher for a male fetus, resulting in male-to-female ratio of 1.32 in chromosomally normal cases [111]. Research indicates that the anatomical sex ratio in spontaneous abortion is 1.25, indicating a male fetus excess that is apparent at all gestational ages and all sizes [112]. These findings reinforce the notion that the male fetus is more susceptible to placental dysfunction. This aligns with observations linking male fetal sex to other placental-related complications, including PTB.

4.3.3. Transcriptional and Epigenetic Differences

Recent advancements in high-throughput sequencing technologies have enabled the analysis of sexually dimorphic gene expression patterns in placental tissues. Transcriptomic analyses have shown that female placentas exhibit a higher expression of genes associated with immune tolerance and regulators of maternal–fetal immune interaction [99]. In contrast, women carrying male fetuses showed increased rates of chronic inflammatory placental lesions [113]. Male placentas exhibit heightened transcriptional activity in pro-inflammatory and immune rejection pathways, potentially contributing to poorer placental outcomes [97]. Ferrous iron (Fe2+) is essential for the KDM3A-mediated histone demethylation necessary for SRY during male gonadal development [114]. The disruption of iron metabolism, via Tfrc deletion or maternal iron deficiency combined with a KDM3A variant, impairs SRY expression and causes male-to-female sex reversal in mice [114]. These experiments demonstrate epigenetic programming, which can change and potentially influence placental function and the fetus’s ability to adapt to inflammatory stressors.
Moreover, hormonal signaling exhibits sexually dimorphic transcriptional modulation. Androgens are critical for the development of the male reproductive tract between gestational weeks 7 and 12 and also serve as precursors for estrogen biosynthesis in both sexes [115,116]. The androgen receptor (AR)-associated gene ARMCX3, which is upregulated in early male placental development, plays a role in mitochondrial dynamics and trophoblast function, with implications for long-term placental performance [97,117].
Additionally, the expression of pregnancy-associated plasma protein (PAPP)-A and free β-hCG, primarily secreted by syncytiotrophoblasts, is significantly higher in female pregnancies [118,119,120]. Yaron et al. reported that maternal serum levels of both PAPP-A and β-hCG were significantly increased in female fetuses compared with male fetuses [118]. This heightened expression in females may enhance endocrine support, immune tolerance, and placental adaptation by modulating trophoblast invasion and maternal vascular remodeling.
Comprehensively, these data suggest that fetal sex influences not only the structural configuration of the placenta but also its transcriptomic and hormonal milieu, thereby shaping differential susceptibilities to gestational complications such as PTB, PE, and spontaneous abortion. Male fetuses, due to a more rigid and growth-focused transcriptional program, may exhibit less plasticity in response to environmental insults, rendering them more vulnerable to placental dysfunction.

5. Oxidative Stress as a Trigger for Preterm Birth

5.1. Balanced Oxidative Stress in Normal Pregnancy

The reactive oxygen species (ROS), which include superoxide radicals (O2), hydrogen peroxide (H2O2), and hydroxyl radicals (HO˙), are the byproducts of aerobic cellular metabolism and serve as important signaling molecules in various physiological processes such as cell proliferation, autophagy, and inflammation modulation [121,122]. Maintaining a balanced level of ROS is crucial for normal pregnancy progression, including placental development and the timely initiation of labor.
As pregnancy progresses, there is an increase in mitochondrial activity in response to elevated oxygen requirements, particularly between weeks 10 and 12 of gestation. At this time, chorionic villi detach from the maternal SpAs, allowing maternal blood to flood the intervillous space and precipitating a substantial rise in O2 tension [123,124]. Notably, high levels of H2O2 are synthesized at the perivitelline space of each zygote, suggesting that H2O2 plays a vital role in forming a protective layer around the fertilized egg [121,125]. In response to ROS accumulation and to prevent oxidative damage, the placenta upregulates the antioxidant defense system. It is evidential that major placental antioxidants include heme oxygenase (HO)-1 and -2, superoxide dismutase (SOD), catalase, and GPx [126]. Collectively, these exert their antioxidant effects by neutralizing ROS, thus ensuring the maintenance of redox homeostasis, a state that is essential for the proper functioning of the placenta and the development of the fetus.

5.2. Imbalanced Oxidative Stress and Adverse Pregnancy Outcomes

When the balance between ROS production and antioxidant defenses is disrupted, oxidative stress can lead to a cascade of cellular damage that contributes to several pregnancy complications, including sPTB, PE, embryo resorption, intrauterine growth restriction (IUGR), and spontaneous abortion [11,127] (Figure 4).
Increased maternal blood flow within the placenta has been associated with the elevated production of ROS, resulting in lipid peroxidation, mitochondrial dysfunction, and DNA damage [130]. Among oxidative biomarkers, hydroxylated nucleotide 8-hydroxydeoxyguanine (8-OHdG) is considered an indicator of ROS-induced genotoxic stress [132]. Clinical studies have reported higher levels of 8-OHdG, malondialdehyde (MDA), and catalase activity in women with PTB compared with those with term deliveries [122,133,134,135]. These findings are accompanied by reduced antioxidant defense capacity, further confirming the role of oxidative imbalance in PTB pathophysiology [135].
Not every aspect of how ROS stimulates inflammation during pregnancy is clearly understood. ROS is known to activate the p38-mitogen-activated protein kinase (MAPK) pathway, ultimately triggering the labor process by generating uterotonic biomolecule signals (Figure 4). This pathway plays a role in the senescence of the placental fetal membrane and the pathogenesis of pPROM and PTB [122].
Despite the growing evidence regarding the contribution of oxidative stress to pregnancy complications, limited research has been undertaken to elucidate how these mechanisms may vary based on fetal sex. Understanding the sex-specific responses to oxidative stress could provide novel insights into differing pregnancy outcomes and fetal adaptation.

5.3. Fetal Sex Differences in Oxidative Stress Responses

The impact of oxidative stress response appears to differ by fetal sex. The mother and female fetuses showed a higher antioxidant capacity and lower oxidative stress markers than those of male fetuses. Female neonates had higher elevated antioxidant enzyme activity (catalase, GPx, and SOD) and reduced pro-inflammatory cytokines (IL-6, TNF-α), indicating a better oxidative and inflammatory balance [136]. Experimental studies in rodent models have shown that both male and ovariectomized female rats exhibit elevated levels of peroxidase and reduced mitochondrial glutathione (GSH). Interestingly, estrogen replacement restored GSH levels in ovariectomized females to those of intact females, implicating estrogen as a key mediator of antioxidant defenses [137].
It is widely accepted that the level of oxytocin in blood is higher in females compared with males [138]. The higher level of oxytocin might be the underlying factor that affects the antioxidant reaction. Furthermore, the increased total antioxidant capacity of healthy female neonates is consistent with the higher GPx activity in adult female erythrocytes compared with males [136,139]. This lower oxidative stress in females might be affected by estrogen, showing a positive relationship between estrogen and GPx [140]. Similarly, estradiol stimulates the activation of MAPK and NF-κB through the upregulation of SOD and GPx [140].
O2 availability and regulation are crucial in placental and fetal development, influencing the maintenance of trophoblast stemness, the regulation of proliferation and invasion, hormone synthesis, and transporter activity expression. Female fetuses tend to have higher oxygen O2 concentrations in the umbilical vein, which may reflect more efficient placental oxygen delivery, while a lower oxygen concentration in the umbilical artery suggests a better utilization efficiency or reduced oxygen consumption [141]. As described earlier, a high metabolic rate for the growth-priority strategy of males might generate a higher concentration of oxidative stress in males [142]. The transcriptomic analysis conducted by Lien et al. and Akram et al. supports this by showing dysregulated nutrient sensing, metabolic signaling, and catabolic processes in only the female placenta [143,144].
These differences extend to birth outcomes. Cord arterial blood in female neonates shows significantly higher levels of catecholamines in response to hypoxic stress labor, suggesting enhanced physiological adaptation mechanisms compared with males [145]. This pattern has been confirmed in animal models, where female rat pups exposed to anoxia exhibited heightened catecholamines responses, indicating sex-specific resilience under hypoxic conditions [146].
These findings collectively suggest that female fetuses may possess superior adaptive mechanisms to withstand oxidative and hypoxic stress, potentially contributing to their generally more favorable outcomes in compromised pregnancies. The higher catecholamine response in females may act as a protective mechanism to enhance survival and adaptation in utero, especially in the context of preterm labor and placental dysfunction [145].

6. Additional Fetal Sex-Specific Mechanisms in PTB

Glucocorticoids play a critical role in placental and fetal development, particularly in modulating stress responses through glucocorticoid receptor (GR) α-mediated signaling. Excessive glucocorticoids are associated with asthma, fetal growth, and metabolic processes [147,148]. The female fetal–placenta unit is more sensitive to cortisol via increased GRα activity through an interaction with GRα C and GRα D3 [147]. In contrast, male human and sheep placentas present significantly decreased levels of GRα and a high concentration of antagonistic isoform GRβ, which interferes with the activity of GRα and is related to glucocorticoid resistance (Figure 3) [149,150]. In the female rat placenta, the labyrinth zone exhibits a higher expression of GR (Nr3c1) [151]. In another mouse model, it was shown that the male placenta exhibited elevated levels of Nr3c1, vascular endothelial growth factor (Vegf) A, IGF type 1 receptor (Igf1r), and Igf2r, and slc38a1 mRNA levels during the late gestational period [151]. Conversely, female placentae demonstrated augmented levels of Igf2 and Slc2a1 mRNA expression, indicating that similar pathways are regulated by placental sex [151,152].
Elevated maternal cortisol levels stimulate the production of placental corticotropin-releasing hormone (CRH), creating a positive feedback loop that amplifies CRH and cortisol concentrations, ultimately contributing to increased estrogen production and the onset of PTB [152]. In addition to stimulating cortisol, CRH also enhances the placental production of estrogen. Oaks et al. found that higher cortisol levels in early and mid-pregnancy were linked to a shorter duration of pregnancy, while elevated cortisol levels at the onset of pregnancy increased the risk of PTB. The risk was three times higher in women carrying a male fetus, but not in those carrying a female fetus [153].
A heightened proportion of singleton male births in PTB has been observed, particularly during the period of 20–37 weeks of gestation. This excess ratio for males was found to be 7.2% and 2.8% in the singleton White and singleton Black groups, respectively (p < 0.001) [154]. Several studies have conducted genome-wide association research to identify any variants associated with PTBs or gestational age. EBF1, EEFSEC, AGTR2, ADCY5, RAP2G, and WNT4 (known to alter the binding of the estrogen receptor) loci were associated with gestational week, and the common variations in three loci (EBF1, EEFSEC, and AGTR2) were associated with PTBs in the European ancestry database [155]. However, the genome-wide association studies (GWASs) based on the Japanese population has not found significant variants [156]. The Jewish cohorts who carry a mutation of BRCA1/2 showed a significantly lower male-to-female infant ratio [157]. Despite ongoing genetic studies, the precise contribution of sexually dimorphic genetic factors to PTB remains inconclusive, highlighting the complexity of its etiology and the need for further integrative research.

7. Conclusions

Preterm birth is a multifactorial condition influenced by inflammation, oxidative stress, placental function, and fetal sex-specific physiological adaptation. The accumulating evidence shows that male fetuses are more vulnerable to adverse intrauterine conditions, contributing to their higher risk of PTB and poorer neonatal outcomes. In contrast, female fetuses exhibit an enhanced antioxidant capacity, stress resilience, and more efficient placental adaptations. These sexually dimorphic responses are especially relevant in the context of inflammation, placental dysfunction, and oxidative stress—three interconnected yet independently major pathological processes. Although this review does not comprehensively address all pregnancy mechanisms, it supports a growing consensus that fetal sex is one of the major key biological variables in placental development and pregnancy outcomes. Future research should prioritize fetal sex as a critical biological variable to refine prevention strategies and reduce disparities in perinatal health.

Author Contributions

Conceptualization, G.L. and D.O.C.A.; writing—original draft preparation, G.L.; writing—review and editing, G.M.A. and D.O.C.A.; visualization, G.L. and Y.J.K.; investigation, G.L. and G.M.A.; supervision, D.O.C.A. and Y.J.K.; project administration, D.O.C.A. and Y.J.K.; funding acquisition, Y.J.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by a grant from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health and Welfare, Republic of Korea (grant number: RS-2023-00266554, RS-2023-00262969), and by the BK21 FOUR (Fostering Outstanding Universities for Research) funded by the Ministry of Education (MOE, Korea) and National Research Foundation of Korea (NRF-5199990614253, Education Research Center for 4IR-Based Health Care).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Acknowledgments

I gratefully acknowledge Khondoker Akram, The University of Sheffield, for his valuable suggestion to pursue this work as a review manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
8-OHdG8-oxodeoxyguanosine
AGandrogen receptor
BPDbronchopulmonary dysplasia
CCLC-C motif chemokine ligand
COX-2cyclooxygenase-2
DAMPsdamage-associated molecular patterns
DOHaDdevelopmental origins of health and disease
Eembryonic day
EVsextracellular vesicles
Fe2+ferrous iron
GWASgenome-wide associated studies
GM-CSFgranulocyte–macrophage colony stimulating factor
GnRHgonadotropin-releasing hormone
GPXglutathione peroxidase
GRglucocorticoid receptor
GRER1G-protein-coupled estrogen receptor 1
GSHglutathione
H2O2hydrogen peroxide
hCGhuman chorionic gonadotropin
HIFhypoxia-inducible factor
HOheme oxygenase
HO˙hydroxyl radical
IFNinterferon
Igimmunoglobulin
IGFinsulin-like growth factor
ILinterleukin
IUGRintrauterine growth restriction
MAPKp38-mitogen-activated protein kinase
MDAmalondialdehyde
MIACmicrobial invasion of amniotic cavity
MMPmatrix metalloproteinase-9
NF-κbnuclear factor kappa-light-chain-enhancer of activated B cell
NKnatural killer
nRBCsnucleated red blood cells
O2superoxide radical
P4progesterone
PAMPspathogen-associated molecular patterns
PAPPpregnancy-associated plasma protein
PEpreeclampsia
PGprostaglandin
PlGFplacental growth factor
PIVHperiventricular–intraventricular hemorrhage
pPROMpreterm premature rupture of the membrane
PRAprogesterone receptor A
PTBpreterm birth
RDSrespiratory distress syndrome
ROSreactive oxygen species
SODsuperoxide dismutase
SpAsspiral arteries
sPTBspontaneous preterm birth
SRYsex-determining region
T1trimester 1
T2trimester 2
T3trimester 3
TLRtoll-like receptor
TNFtumor necrosis factor
Vegfvascular endothelial growth factor
XCIX chromosome inhibition

References

  1. Beck, S.; Wojdyla, D.; Say, L.; Betran, A.P.; Merialdi, M.; Requejo, J.H.; Rubens, C.; Menon, R.; Van Look, P.F. The worldwide incidence of preterm birth: A systematic review of maternal mortality and morbidity. Bull. World Health Organ. 2010, 88, 31–38. [Google Scholar] [CrossRef] [PubMed]
  2. Ohuma, E.O.; Moller, A.-B.; Bradley, E.; Chakwera, S.; Hussain-Alkhateeb, L.; Lewin, A.; Okwaraji, Y.B.; Mahanani, W.R.; Johansson, E.W.; Lavin, T.; et al. National, regional, and global estimates of preterm birth in 2020, with trends from 2010: A systematic analysis. Lancet 2023, 402, 1261–1271. [Google Scholar] [CrossRef] [PubMed]
  3. Lodefalk, M.; Chelslín, F.; Patriksson Karlsson, J.; Hansson, S.R. Placental Changes and Neuropsychological Development in Children-A Systematic Review. Cells 2023, 12, 435. [Google Scholar] [CrossRef] [PubMed]
  4. Romero, R.; Dey, S.K.; Fisher, S.J. Preterm labor: One syndrome, many causes. Science 2014, 345, 760–765. [Google Scholar] [CrossRef] [PubMed]
  5. Menon, R.; Behnia, F.; Polettini, J.; Richardson, L.S. Novel pathways of inflammation in human fetal membranes associated with preterm birth and preterm pre-labor rupture of the membranes. Semin. Immunopathol. 2020, 42, 431–450. [Google Scholar] [CrossRef] [PubMed]
  6. Agrawal, V.; Hirsch, E. Intrauterine infection and preterm labor. Semin. Fetal Neonatal Med. 2012, 17, 12–19. [Google Scholar] [CrossRef] [PubMed]
  7. Adams Waldorf, K.M.; Singh, N.; Mohan, A.R.; Young, R.C.; Ngo, L.; Das, A.; Tsai, J.; Bansal, A.; Paolella, L.; Herbert, B.R.; et al. Uterine overdistention induces preterm labor mediated by inflammation: Observations in pregnant women and nonhuman primates. Am. J. Obs. Gynecol. 2015, 213, 830.e1–830.e19. [Google Scholar] [CrossRef] [PubMed]
  8. Lamont, R.F. Advances in the prevention of infection-related preterm birth. Front. Immunol. 2015, 6, 566. [Google Scholar] [CrossRef] [PubMed]
  9. Cappelletti, M.; Della Bella, S.; Ferrazzi, E.; Mavilio, D.; Divanovic, S. Inflammation and preterm birth. J. Leucoc. Biol. 2016, 99, 67–78. [Google Scholar] [CrossRef] [PubMed]
  10. Moore, T.A.; Ahmad, I.M.; Zimmerman, M.C. Oxidative stress and preterm birth: An integrative review. Biol. Res. Nurs. 2018, 20, 497–512. [Google Scholar] [CrossRef] [PubMed]
  11. Dutta, E.H.; Behnia, F.; Boldogh, I.; Saade, G.R.; Taylor, B.D.; Kacerovský, M.; Menon, R. Oxidative stress damage-associated molecular signaling pathways differentiate spontaneous preterm birth and preterm premature rupture of the membranes. MHR Basic Sci. Reprod. Med. 2016, 22, 143–157. [Google Scholar] [CrossRef] [PubMed]
  12. Koullali, B.; Oudijk, M.; Nijman, T.; Mol, B.; Pajkrt, E. Risk assessment and management to prevent preterm birth. Semin. Fetal Neonatal Med. 2016, 21, 80–88. [Google Scholar] [CrossRef] [PubMed]
  13. Vogel, J.P.; Chawanpaiboon, S.; Moller, A.-B.; Watananirun, K.; Bonet, M.; Lumbiganon, P. The global epidemiology of preterm birth. Best Pract. Res. Clin. Obstet. Gynaecol. 2018, 52, 3–12. [Google Scholar] [CrossRef] [PubMed]
  14. Porpora, M.G.; Piacenti, I.; Scaramuzzino, S.; Masciullo, L.; Rech, F.; Benedetti Panici, P. Environmental contaminants exposure and preterm birth: A systematic review. Toxics 2019, 7, 11. [Google Scholar] [CrossRef] [PubMed]
  15. Khandre, V.; Potdar, J.; Keerti, A.; Khandre, V., Jr. Preterm birth: An overview. Cureus 2022, 14, e33006. [Google Scholar] [CrossRef] [PubMed]
  16. Broere-Brown, Z.A.; Adank, M.C.; Benschop, L.; Tielemans, M.; Muka, T.; Gonçalves, R.; Bramer, W.M.; Schoufour, J.D.; Voortman, T.; Steegers, E.A.P.; et al. Fetal sex and maternal pregnancy outcomes: A systematic review and meta-analysis. Biol. Sex. Differ. 2020, 11, 26. [Google Scholar] [CrossRef] [PubMed]
  17. Lacagnina, S. The Developmental Origins of Health and Disease (DOHaD). Am. J. Lifestyle Med. 2020, 14, 47–50. [Google Scholar] [CrossRef] [PubMed]
  18. Navara, K.J.; Nelson, R.J. Prenatal environmental influences on the production of sex-specific traits in mammals. Semin. Cell Dev. Biol. 2009, 20, 313–319. [Google Scholar] [CrossRef] [PubMed]
  19. Terrell, M.L.; Hartnett, K.P.; Marcus, M. Can environmental or occupational hazards alter the sex ratio at birth? A systematic review. Emerg. Health Threat. J. 2011, 4, 7109. [Google Scholar] [CrossRef] [PubMed]
  20. Peelen, M.J.; Kazemier, B.M.; Ravelli, A.C.; De Groot, C.J.; Van Der Post, J.A.; Mol, B.W.; Hajenius, P.J.; Kok, M. Impact of fetal gender on the risk of preterm birth, a national cohort study. Acta Obstet. Et Gynecol. Scand. 2016, 95, 1034–1041. [Google Scholar] [CrossRef] [PubMed]
  21. Galjaard, S.; Ameye, L.; Lees, C.C.; Pexsters, A.; Bourne, T.; Timmerman, D.; Devlieger, R. Sex differences in fetal growth and immediate birth outcomes in a low-risk Caucasian population. Biol. Sex Differ. 2019, 10, 48. [Google Scholar] [CrossRef] [PubMed]
  22. Vu, H.D.; Dickinson, C.; Kandasamy, Y. Sex difference in mortality for premature and low birth weight neonates: A systematic review. Am. J. Perinatol. 2018, 35, 707–715. [Google Scholar] [PubMed]
  23. Fang, K.; Yue, S.; Wang, S.; Wang, M.; Yu, X.; Ding, Y.; Lv, M.; Liu, Y.; Cao, C.; Liao, Z. The association between sex and neonatal respiratory distress syndrome. BMC Pediatr. 2024, 24, 129. [Google Scholar] [CrossRef] [PubMed]
  24. Yue, H.; Ji, X.; Ku, T.; Li, G.; Sang, N. Sex difference in bronchopulmonary dysplasia of offspring in response to maternal PM2. 5 exposure. J. Hazard. Mater. 2020, 389, 122033. [Google Scholar] [CrossRef] [PubMed]
  25. Mohamed, M.A.; Aly, H. Male gender is associated with intraventricular hemorrhage. Pediatrics 2010, 125, e333–e339. [Google Scholar] [CrossRef] [PubMed]
  26. Khan, Y.T.; Tsompanidis, A.; Radecki, M.A.; Dorfschmidt, L.; Adhya, D.; Ayeung, B.; Bamford, R.; Biron-Shental, T.; Burton, G.; Cowell, W.; et al. Sex Differences in Human Brain Structure at Birth. Biol. Sex Differ. 2024, 15, 81. [Google Scholar] [CrossRef] [PubMed]
  27. Winckelmans, E.; Vrijens, K.; Tsamou, M.; Janssen, B.G.; Saenen, N.D.; Roels, H.A.; Kleinjans, J.; Lefebvre, W.; Vanpoucke, C.; de Kok, T.M.; et al. Newborn sex-specific transcriptome signatures and gestational exposure to fine particles: Findings from the ENVIRONAGE birth cohort. Environ. Health 2017, 16, 52. [Google Scholar] [CrossRef] [PubMed]
  28. Cossi, M.; Zuta, S.; Padula, A.M.; Gould, J.B.; Stevenson, D.K.; Shaw, G.M. Role of infant sex in the association between air pollution and preterm birth. Ann. Epidemiol. 2015, 25, 874–876. [Google Scholar] [CrossRef] [PubMed]
  29. Park, S.; Kwon, E.; Lee, G.; You, Y.A.; Kim, S.M.; Hur, Y.M.; Jung, S.; Jee, Y.; Park, M.H.; Na, S.H.; et al. Effect of Particulate Matter 2.5 on Fetal Growth in Male and Preterm Infants through Oxidative Stress. Antioxidants 2023, 12, 1916. [Google Scholar] [CrossRef] [PubMed]
  30. Günther, V.; Alkatout, I.; Stein, A.; Maass, N.; Strauss, A.; Voigt, M. Impact of smoking and fetal gender on preterm delivery. J. Dev. Orig. Health Dis. 2021, 12, 632–637. [Google Scholar] [CrossRef] [PubMed]
  31. Voigt, M.; Hermanussen, M.; Wittwer-Backofen, U.; Fusch, C.; Hesse, V. Sex-specific differences in birth weight due to maternal smoking during pregnancy. Eur. J. Pediatr. 2006, 165, 757–761. [Google Scholar] [CrossRef] [PubMed]
  32. Flannigan, K.; Poole, N.; Cook, J.; Unsworth, K. Sex-related differences among individuals assessed for fetal alcohol spectrum disorder in Canada. Alcohol. Clin. Exp. Res. 2023, 47, 613–623. [Google Scholar] [CrossRef] [PubMed]
  33. Darrow, L.A.; Huang, M.; Warren, J.L.; Strickland, M.J.; Holmes, H.A.; Newman, A.J.; Chang, H.H. Preterm and early-term delivery after heat waves in 50 US metropolitan areas. JAMA Netw. Open 2024, 7, e2412055. [Google Scholar] [CrossRef] [PubMed]
  34. Yu, G.; Yang, L.; Liu, M.; Wang, C.; Shen, X.; Fan, L.; Zhang, J. Extreme Temperature Exposure and Risks of Preterm Birth Subtypes Based on a Nationwide Survey in China. Environ. Health Perspect. 2023, 131, 87009. [Google Scholar] [CrossRef] [PubMed]
  35. Green, E.S.; Arck, P.C. Pathogenesis of preterm birth: Bidirectional inflammation in mother and fetus. Semin. Immunopathol. 2020, 42, 413–429. [Google Scholar] [CrossRef] [PubMed]
  36. Griffith, O.W.; Chavan, A.R.; Protopapas, S.; Maziarz, J.; Romero, R.; Wagner, G.P. Embryo implantation evolved from an ancestral inflammatory attachment reaction. Proc. Natl. Acad. Sci. USA 2017, 114, e6566–e6575. [Google Scholar] [CrossRef] [PubMed]
  37. Bromfield, J.J. Seminal fluid and reproduction: Much more than previously thought. J. Assist. Reprod. Genet. 2014, 31, 627–636. [Google Scholar] [CrossRef] [PubMed]
  38. Kapovic, M.; Rukavina, D. Kinetics of lymphoproliferative responses of lymphocytes harvested from the uterine draining lymph nodes during pregnancy in rats. J. Reprod. Immunol. 1991, 20, 93–101. [Google Scholar] [CrossRef] [PubMed]
  39. Sivarajasingam, S.P.; Imami, N.; Johnson, M.R. Myometrial cytokines and their role in the onset of labour. J. Endocrinol. 2016, 231, R101–R119. [Google Scholar] [CrossRef] [PubMed]
  40. Kircheis, R.; Planz, O. The Role of Toll-like Receptors (TLRs) and Their Related Signaling Pathways in Viral Infection and Inflammation. Int. J. Mol. Sci. 2023, 24, 6701. [Google Scholar] [CrossRef] [PubMed]
  41. Faro, J.; Romero, R.; Schwenkel, G.; Garcia-Flores, V.; Arenas-Hernandez, M.; Leng, Y.; Xu, Y.; Miller, D.; Hassan, S.S.; Gomez-Lopez, N. Intra-amniotic inflammation induces preterm birth by activating the NLRP3 inflammasome. Biol. Reprod. 2019, 100, 1290–1305. [Google Scholar] [CrossRef] [PubMed]
  42. Presicce, P.; Park, C.W.; Senthamaraikannan, P.; Bhattacharyya, S.; Jackson, C.; Kong, F.; Rueda, C.M.; DeFranco, E.; Miller, L.A.; Hildeman, D.A.; et al. IL-1 signaling mediates intrauterine inflammation and chorio-decidua neutrophil recruitment and activation. JCI Insight 2018, 3, e98306. [Google Scholar] [CrossRef] [PubMed]
  43. Yang, X.; Tian, Y.; Zheng, L.; Luu, T.; Kwak-Kim, J. The Update Immune-Regulatory Role of Pro- and Anti-Inflammatory Cytokines in Recurrent Pregnancy Losses. Int. J. Mol. Sci. 2023, 24, 132. [Google Scholar] [CrossRef] [PubMed]
  44. Lash, G.E.; Schiessl, B.; Kirkley, M.; Innes, B.A.; Cooper, A.; Searle, R.F.; Robson, S.C.; Bulmer, J.N. Expression of angiogenic growth factors by uterine natural killer cells during early pregnancy. J. Leukoc. Biol. 2006, 80, 572–580. [Google Scholar] [CrossRef] [PubMed]
  45. Saito, S.; Kasahara, T.; Kato, Y.; Ishihara, Y.; Ichijo, M. Elevation of amniotic fluid interleukin 6 (IL-6), IL-8 and granulocyte colony stimulating factor (G-CSF) in term and preterm parturition. Cytokine 1993, 5, 81–88. [Google Scholar] [CrossRef] [PubMed]
  46. Romero, R.; Sirtori, M.; Oyarzun, E.; Avila, C.; Mazor, M.; Callahan, R.; Sabo, V.; Athanassiadis, A.P.; Hobbins, J.C. Infection and labor V. Prevalence, microbiology, and clinical significance of intraamniotic infection in women with preterm labor and intact membranes. Am. J. Obstet. Gynecol. 1989, 161, 817–824. [Google Scholar] [CrossRef] [PubMed]
  47. Kim, M.J.; Romero, R.; Gervasi, M.T.; Kim, J.-S.; Yoo, W.; Lee, D.-C.; Mittal, P.; Erez, O.; Kusanovic, J.P.; Hassan, S.S.; et al. Widespread microbial invasion of the chorioamniotic membranes is a consequence and not a cause of intra-amniotic infection. Lab. Investig. 2009, 89, 924–936. [Google Scholar] [CrossRef] [PubMed]
  48. Brown, R.G.; Marchesi, J.R.; Lee, Y.S.; Smith, A.; Lehne, B.; Kindinger, L.M.; Terzidou, V.; Holmes, E.; Nicholson, J.K.; Bennett, P.R.; et al. Vaginal dysbiosis increases risk of preterm fetal membrane rupture, neonatal sepsis and is exacerbated by erythromycin. BMC Med. 2018, 16, 9. [Google Scholar] [CrossRef] [PubMed]
  49. Deng, W.; Yuan, J.; Cha, J.; Sun, X.; Bartos, A.; Yagita, H.; Hirota, Y.; Dey, S.K. Endothelial cells in the decidual bed are potential therapeutic targets for preterm birth prevention. Cell Rep. 2019, 27, 1755–1768.e4. [Google Scholar] [CrossRef] [PubMed]
  50. Firmal, P.; Shah, V.K.; Chattopadhyay, S. Insight Into TLR4-Mediated Immunomodulation in Normal Pregnancy and Related Disorders. Front. Immunol. 2020, 11, 807. [Google Scholar] [CrossRef] [PubMed]
  51. Flis, W.; Socha, M.W. The Role of the NLRP3 Inflammasome in the Molecular and Biochemical Mechanisms of Cervical Ripening: A Comprehensive Review. Cells 2024, 13, 600. [Google Scholar] [CrossRef] [PubMed]
  52. Mendelson, C.R.; Gao, L.; Montalbano, A.P. Multifactorial Regulation of Myometrial Contractility During Pregnancy and Parturition. Front. Endocrinol. 2019, 10, 714. [Google Scholar] [CrossRef] [PubMed]
  53. Dunn, S.E.; Perry, W.A.; Klein, S.L. Mechanisms and consequences of sex differences in immune responses. Nat. Rev. Nephrol. 2024, 20, 37–55. [Google Scholar] [CrossRef] [PubMed]
  54. Klein, S.L.; Flanagan, K.L. Sex differences in immune responses. Nat. Rev. Immunol. 2016, 16, 626–638. [Google Scholar] [CrossRef] [PubMed]
  55. Smith-Bouvier, D.L.; Divekar, A.A.; Sasidhar, M.; Du, S.; Tiwari-Woodruff, S.K.; King, J.K.; Arnold, A.P.; Singh, R.R.; Voskuhl, R.R. A role for sex chromosome complement in the female bias in autoimmune disease. J. Exp. Med. 2008, 205, 1099–1108. [Google Scholar] [CrossRef] [PubMed]
  56. Itoh, Y.; Golden, L.C.; Itoh, N.; Matsukawa, M.A.; Ren, E.; Tse, V.; Arnold, A.P.; Voskuhl, R.R. The X-linked histone demethylase Kdm6a in CD4+ T lymphocytes modulates autoimmunity. J. Clin. Investig. 2019, 129, 3852–3863. [Google Scholar] [CrossRef] [PubMed]
  57. Whitacre, C.C.; Reingold, S.C.; O’Looney, P.A.; Blankenhorn, E.; Brinley, F.; Collier, E.; Duquette, P.; Fox, H.; Giesser, B.; Gilmore, W. A gender gap in autoimmunity: Task force on gender, multiple sclerosis and autoimmunity. Science 1999, 283, 1277–1278. [Google Scholar] [CrossRef] [PubMed]
  58. Forsberg, L.A.; Rasi, C.; Malmqvist, N.; Davies, H.; Pasupulati, S.; Pakalapati, G.; Sandgren, J.; de Ståhl, T.D.; Zaghlool, A.; Giedraitis, V. Mosaic loss of chromosome Y in peripheral blood is associated with shorter survival and higher risk of cancer. Nat. Genet. 2014, 46, 624–628. [Google Scholar] [CrossRef] [PubMed]
  59. Miles, M.A.; Huttmann, T.D.; Liong, S.; Liong, F.; O’Leary, J.J.; Brooks, D.A.; Selemidis, S. Exploring the Contribution of TLR7 to Sex-Based Disparities in Respiratory Syncytial Virus (RSV)-Induced Inflammation and Immunity. Viruses 2025, 17, 428. [Google Scholar] [CrossRef] [PubMed]
  60. Pradhan, A.; Olsson, P.-E. Sex differences in severity and mortality from COVID-19: Are males more vulnerable? Biol. Sex Differ. 2020, 11, 53. [Google Scholar] [CrossRef] [PubMed]
  61. Abdel-Hafiz, H.A.; Schafer, J.M.; Chen, X.; Xiao, T.; Gauntner, T.D.; Li, Z.; Theodorescu, D. Y chromosome loss in cancer drives growth by evasion of adaptive immunity. Nature 2023, 619, 624–631. [Google Scholar] [CrossRef] [PubMed]
  62. Mauvais-Jarvis, F.; Merz, N.B.; Barnes, P.J.; Brinton, R.D.; Carrero, J.-J.; DeMeo, D.L.; De Vries, G.J.; Epperson, C.N.; Govindan, R.; Klein, S.L. Sex and gender: Modifiers of health, disease, and medicine. Lancet 2020, 396, 565–582. [Google Scholar] [CrossRef] [PubMed]
  63. Libert, C.; Dejager, L.; Pinheiro, I. The X chromosome in immune functions: When a chromosome makes the difference. Nat. Rev. Immunol. 2010, 10, 594–604. [Google Scholar] [CrossRef] [PubMed]
  64. Migeon, B. Females Are Mosaics: X Inactivation and Sex Differences in Disease; Oxford University Press: Oxford, UK, 2007. [Google Scholar]
  65. Migeon, B.R. Why females are mosaics, X-chromosome inactivation, and sex differences in disease. Gend. Med. 2007, 4, 97–105. [Google Scholar] [CrossRef] [PubMed]
  66. Wainer Katsir, K.; Linial, M. Human genes escaping X-inactivation revealed by single cell expression data. BMC Genom. 2019, 20, 201. [Google Scholar] [CrossRef] [PubMed]
  67. Souyris, M.; Cenac, C.; Azar, P.; Daviaud, D.; Canivet, A.; Grunenwald, S.; Pienkowski, C.; Chaumeil, J.; Mejía, J.E.; Guéry, J.-C. TLR7 escapes X chromosome inactivation in immune cells. Sci. Immunol. 2018, 3, eaap8855. [Google Scholar] [CrossRef] [PubMed]
  68. Liu, C.-A.; Wang, C.-L.; Chuang, H.; Ou, C.-Y.; Hsu, T.-Y.; Yang, K.D. Prediction of elevated cord blood IgE levels by maternal IgE levels, and the neonate’s gender and gestational age. Chang. Gung Med. J. 2003, 26, 561–569. [Google Scholar] [PubMed]
  69. Page, S.T.; Plymate, S.R.; Bremner, W.J.; Matsumoto, A.M.; Hess, D.L.; Lin, D.W.; Amory, J.K.; Nelson, P.S.; Wu, J.D. Effect of medical castration on CD4+CD25+ T cells, CD8+ T cell IFN-γ expression, and NK cells: A physiological role for testosterone and/or its metabolites. Am. J. Physiol. -Endocrinol. Metab. 2006, 290, E856–E863. [Google Scholar] [CrossRef] [PubMed]
  70. Sun, T.; Gonzalez, T.L.; Deng, N.; DiPentino, R.; Clark, E.L.; Lee, B.; Tang, J.; Wang, Y.; Stripp, B.R.; Yao, C. Sexually dimorphic crosstalk at the maternal-fetal interface. J. Clin. Endocrinol. Metab. 2020, 105, e4831–e4847. [Google Scholar] [CrossRef] [PubMed]
  71. Braun, A.E.; Muench, K.L.; Robinson, B.G.; Wang, A.; Palmer, T.D.; Winn, V.D. Examining sex differences in the human placental transcriptome during the first fetal androgen peak. Reprod. Sci. 2021, 28, 801–818. [Google Scholar] [CrossRef] [PubMed]
  72. Hussain, A.; Malik, Q.; Nadeem, M.T.; Ullah, N.; Ikram, F.; Hussain, M. Frequency Of Meningitis In Neonatal Sepsis. J. Rawalpindi Med. Coll. (JRMC) 2023, 27, 114–118. [Google Scholar]
  73. Haahr, T.; Clausen, T.D.; Thorsen, J.; Rasmussen, M.A.; Mortensen, M.S.; Lehtimäki, J.; Shah, S.A.; Hjelmsø, M.H.; Bønnelykke, K.; Chawes, B.L.; et al. Vaginal dysbiosis in pregnancy associates with risk of emergency caesarean section: A prospective cohort study. Clin. Microbiol. Infect. 2022, 28, 588–595. [Google Scholar] [CrossRef] [PubMed]
  74. Park, S.; Oh, D.; Heo, H.; Lee, G.; Kim, S.M.; Ansari, A.; You, Y.A.; Jung, Y.J.; Kim, Y.H.; Lee, M.; et al. Prediction of preterm birth based on machine learning using bacterial risk score in cervicovaginal fluid. Am. J. Reprod. Immunol. 2021, 86, e13435. [Google Scholar] [CrossRef] [PubMed]
  75. You, Y.-A.; Park, S.; Kim, K.; Kwon, E.J.; Hur, Y.M.; Kim, S.M.; Lee, G.; Ansari, A.; Park, J.; Kim, Y.J. Transition in vaginal Lactobacillus species during pregnancy and prediction of preterm birth in Korean women. Sci. Rep. 2022, 12, 22303. [Google Scholar] [CrossRef] [PubMed]
  76. Wu, G.; Grassi, P.; MacIntyre, D.A.; Molina, B.G.; Sykes, L.; Kundu, S.; Hsiao, C.-T.; Khoo, K.-H.; Bennett, P.R.; Dell, A. N-glycosylation of cervicovaginal fluid reflects microbial community, immune activity, and pregnancy status. Sci. Rep. 2022, 12, 16948. [Google Scholar] [CrossRef] [PubMed]
  77. Ercan, A.; Kohrt, W.M.; Cui, J.; Deane, K.D.; Pezer, M.; Yu, E.W.; Hausmann, J.S.; Campbell, H.; Kaiser, U.B.; Rudd, P.M.; et al. Estrogens regulate glycosylation of IgG in women and men. JCI Insight 2017, 2, e89703. [Google Scholar] [CrossRef] [PubMed]
  78. Niemann, H.H.; Schubert, W.-D.; Heinz, D.W. Adhesins and invasins of pathogenic bacteria: A structural view. Microbes Infect. 2004, 6, 101–112. [Google Scholar] [CrossRef] [PubMed]
  79. Cindrova-Davies, T.; Sferruzzi-Perri, A.N. Human placental development and function. Semin. Cell Dev. Biol. 2022, 131, 66–77. [Google Scholar] [CrossRef] [PubMed]
  80. Burton, G.J.; Jauniaux, E. What is the placenta? Am. J. Obstet. Gynecol. 2015, 213, S6.e1–S6.e4. [Google Scholar] [CrossRef] [PubMed]
  81. Gude, N.M.; Roberts, C.T.; Kalionis, B.; King, R.G. Growth and function of the normal human placenta. Thromb. Res. 2004, 114, 397–407. [Google Scholar] [CrossRef] [PubMed]
  82. Whitley, G.S.J.; Cartwright, J.E. Trophoblast-mediated spiral artery remodelling: A role for apoptosis. J. Anat. 2009, 215, 21–26. [Google Scholar] [CrossRef] [PubMed]
  83. Farrelly, R.; Kennedy, M.G.; Spencer, R.; Forbes, K. Extracellular vesicles as markers and mediators of pregnancy complications: Gestational diabetes, pre-eclampsia, preterm birth and fetal growth restriction. J. Physiol. 2023, 601, 4973–4988. [Google Scholar] [CrossRef] [PubMed]
  84. Costa, M.A. The endocrine function of human placenta: An overview. Reprod. Biomed. Online 2016, 32, 14–43. [Google Scholar] [CrossRef] [PubMed]
  85. Massimiani, M.; Lacconi, V.; La Civita, F.; Ticconi, C.; Rago, R.; Campagnolo, L. Molecular Signaling Regulating Endometrium-Blastocyst Crosstalk. Int. J. Mol. Sci. 2019, 21, 23. [Google Scholar] [CrossRef] [PubMed]
  86. Preston, M.; Hall, M.; Shennan, A.; Story, L. The role of placental insufficiency in spontaneous preterm birth: A literature review. Eur. J. Obstet. Gynecol. Reprod. Biol. 2024, 295, 136–142. [Google Scholar] [CrossRef] [PubMed]
  87. Faye-Petersen, O. The placenta in preterm birth. J. Clin. Pathol. 2008, 61, 1261–1275. [Google Scholar] [CrossRef] [PubMed]
  88. Parry, S.; Strauss, J.F. Premature rupture of the fetal membranes. N. Engl. J. Med. 1998, 338, 663–670. [Google Scholar] [CrossRef] [PubMed]
  89. Kim, Y.M.; Bujold, E.; Chaiworapongsa, T.; Gomez, R.; Yoon, B.H.; Thaler, H.T.; Rotmensch, S.; Romero, R. Failure of physiologic transformation of the spiral arteries in patients with preterm labor and intact membranes. Am. J. Obstet. Gynecol. 2003, 189, 1063–1069. [Google Scholar] [CrossRef] [PubMed]
  90. Loftness, B.C.; Bernstein, I.; McBride, C.A.; Cheney, N.; McGinnis, E.W.; McGinnis, R.S. Preterm Preeclampsia Risk Modelling: Examining Hemodynamic, Biochemical, and Biophysical Markers Prior to Pregnancy. medRxiv 2023, 1–4. [Google Scholar] [CrossRef] [PubMed]
  91. Cindrova-Davies, T.; Fogarty, N.M.; Jones, C.J.; Kingdom, J.; Burton, G.J. Evidence of oxidative stress-induced senescence in mature, post-mature and pathological human placentas. Placenta 2018, 68, 15–22. [Google Scholar] [CrossRef] [PubMed]
  92. Baker, B.C.; Heazell, A.E.P.; Sibley, C.; Wright, R.; Bischof, H.; Beards, F.; Guevara, T.; Girard, S.; Jones, R.L. Hypoxia and oxidative stress induce sterile placental inflammation in vitro. Sci. Rep. 2021, 11, 7281. [Google Scholar] [CrossRef] [PubMed]
  93. Maiti, K.; Sultana, Z.; Aitken, R.J.; Morris, J.; Park, F.; Andrew, B.; Riley, S.C.; Smith, R. Evidence that fetal death is associated with placental aging. Am. J. Obstet. Gynecol. 2017, 217, 441.e1–441.e14. [Google Scholar] [CrossRef] [PubMed]
  94. Johns, J.; Jauniaux, E. Threatened miscarriage as a predictor of obstetric outcome. Obstet. Gynecol. 2006, 107, 845–850. [Google Scholar] [CrossRef] [PubMed]
  95. Tikkanen, M.; Metsäranta, M.; Gissler, M.; Luukkaala, T.; Hiilesmaa, V.; Ylikorkala, O.; Paavonen, J.; Andersson, S.; Nuutila, M. Male fetal sex is associated with earlier onset of placental abruption. Acta Obs. Gynecol. Scand. 2010, 89, 916–923. [Google Scholar] [CrossRef] [PubMed]
  96. Tchaicha, J.H.; Reyes, S.B.; Shin, J.; Hossain, M.G.; Lang, F.F.; McCarty, J.H. Glioblastoma angiogenesis and tumor cell invasiveness are differentially regulated by β8 integrin. Cancer Res. 2011, 71, 6371–6381. [Google Scholar] [CrossRef] [PubMed]
  97. Gonzalez, T.L.; Sun, T.; Koeppel, A.F.; Lee, B.; Wang, E.T.; Farber, C.R.; Rich, S.S.; Sundheimer, L.W.; Buttle, R.A.; Chen, Y.-D.I.; et al. Sex differences in the late first trimester human placenta transcriptome. Biol. Sex Differ. 2018, 9, 4. [Google Scholar] [CrossRef] [PubMed]
  98. Orzack, S.H.; Stubblefield, J.W.; Akmaev, V.R.; Colls, P.; Munné, S.; Scholl, T.; Steinsaltz, D.; Zuckerman, J.E. The human sex ratio from conception to birth. Proc. Natl. Acad. Sci. USA 2015, 112, E2102–E2111. [Google Scholar] [CrossRef] [PubMed]
  99. Buckberry, S.; Bianco-Miotto, T.; Bent, S.J.; Dekker, G.A.; Roberts, C.T. Integrative transcriptome meta-analysis reveals widespread sex-biased gene expression at the human fetal–maternal interface. Mol. Hum. Reprod. 2014, 20, 810–819. [Google Scholar] [CrossRef] [PubMed]
  100. Saddiki, H.; Zhang, X.; Colicino, E.; Wilson, A.; Kloog, I.; Wright, R.O.; Wright, R.J.; Lesseur, C. DNA methylation profiles reveal sex-specific associations between gestational exposure to ambient air pollution and placenta cell-type composition in the PRISM cohort study. Clin. Epigenetics 2023, 15, 188. [Google Scholar] [CrossRef] [PubMed]
  101. Kalisch-Smith, J.I.; Simmons, D.G.; Pantaleon, M.; Moritz, K.M. Sex differences in rat placental development: From pre-implantation to late gestation. Biol. Sex Differ. 2017, 8, 17. [Google Scholar] [CrossRef] [PubMed]
  102. Clifton, V.L. Review: Sex and the Human Placenta: Mediating Differential Strategies of Fetal Growth and Survival. Placenta 2010, 31, S33–S39. [Google Scholar] [CrossRef] [PubMed]
  103. Sandman, C.A.; Glynn, L.M.; Davis, E.P. Is there a viability–vulnerability tradeoff? Sex differences in fetal programming. J. Psychosom. Res. 2013, 75, 327–335. [Google Scholar] [CrossRef] [PubMed]
  104. Ao, A.; Erickson, R.P.; Winston, R.M.; Handysude, A.H. Transcription of paternal Y-linked genes in the human zygote as early as the pronucleate stage. Zygote 1994, 2, 281–287. [Google Scholar] [CrossRef] [PubMed]
  105. Burgoyne, P.S. A Y-chromosomal effect on blastocyst cell number in mice. Development 1993, 117, 341–345. [Google Scholar] [CrossRef] [PubMed]
  106. Schalekamp-Timmermans, S.; Arends, L.R.; Alsaker, E.; Chappell, L.; Hansson, S.; Harsem, N.K.; Jälmby, M.; Jeyabalan, A.; Laivuori, H.; Lawlor, D.A.; et al. Fetal sex-specific differences in gestational age at delivery in pre-eclampsia: A meta-analysis. Int. J. Epidemiol. 2017, 46, 632–642. [Google Scholar] [CrossRef] [PubMed]
  107. Helle, S.; Lummaa, V. A trade-off between having many sons and shorter maternal post-reproductive survival in pre-industrial Finland. Biol. Lett. 2013, 9, 20130034. [Google Scholar] [CrossRef] [PubMed]
  108. Helle, S.; Lummaa, V.; Jokela, J. Sons reduced maternal longevity in preindustrial humans. Science 2002, 296, 1085. [Google Scholar] [CrossRef] [PubMed]
  109. Sheiner, E.; Shoham-Vardi, I.; Hadar, A.; Hallak, M.; Hackmon, R.; Mazor, M. Incidence, obstetric risk factors and pregnancy outcome of preterm placental abruption: A retrospective analysis. J. Matern.-Fetal Neonatal Med. 2002, 11, 34–39. [Google Scholar] [CrossRef] [PubMed]
  110. Kramer, M.S.; Usher, R.H.; Pollack, R.; Boyd, M.; Usher, S. Etilogic Determinants of Abruptio Placentae. Obstet. Gynecol. 1997, 89, 221–226. [Google Scholar] [CrossRef] [PubMed]
  111. Hassold, T.; Quillen, S.; Yamane, J. Sex ratio in spontaneous abortions. Ann. Hum. Genet. 1983, 47, 39–47. [Google Scholar] [CrossRef] [PubMed]
  112. Byrne, J.; Warburton, D.; Opitz, J.M.; Reynolds, J.F. Male excess among anatomically normal fetuses in spontaneous abortions. Am. J. Med. Genet. 1987, 26, 605–611. [Google Scholar] [CrossRef] [PubMed]
  113. Ghidini, A.; Salafia, C.M. Gender differences of placental dysfunction in severe prematurity. BJOG 2005, 112, 140–144. [Google Scholar] [CrossRef] [PubMed]
  114. Okashita, N.; Maeda, R.; Kuroki, S.; Sasaki, K.; Uno, Y.; Koopman, P.; Tachibana, M. Maternal iron deficiency causes male-to-female sex reversal in mouse embryos. Nature 2025, 643, 262–270. [Google Scholar] [CrossRef] [PubMed]
  115. Dohle, G.R.; Smit, M.; Weber, R.F.A. Androgens and male fertility. World J. Urol. 2003, 21, 341–345. [Google Scholar] [CrossRef] [PubMed]
  116. Makieva, S.; Saunders, P.T.K.; Norman, J.E. Androgens in pregnancy: Roles in parturition. Hum. Reprod. Update 2014, 20, 542–559. [Google Scholar] [CrossRef] [PubMed]
  117. Cvitic, S.; Longtine, M.S.; Hackl, H.; Wagner, K.; Nelson, M.D.; Desoye, G.; Hiden, U. The human placental sexome differs between trophoblast epithelium and villous vessel endothelium. PLoS ONE 2013, 8, e79233. [Google Scholar] [CrossRef] [PubMed]
  118. Yaron, Y.; Wolman, I.; Kupferminc, M.J.; Ochshorn, Y.; Many, A.; Orr-Urtreger, A. Effect of fetal gender on first trimester markers and on Down syndrome screening. Prenat. Diagn. 2001, 21, 1027–1030. [Google Scholar] [CrossRef] [PubMed]
  119. Larsen, S.O.; Wøjdemann, K.R.; Shalmi, A.C.; Sundberg, K.; Christiansen, M.; Tabor, A. Gender impact on first trimester markers in Down syndrome screening. Prenat. Diagn. 2002, 22, 1207–1208. [Google Scholar] [CrossRef] [PubMed]
  120. Spencer, K.; Ong, C.Y.; Liao, A.W.; Papademetriou, D.; Nicolaides, K.H. The influence of fetal sex in screening for trisomy 21 by fetal nuchal translucency, maternal serum free beta-hCG and PAPP-A at 10-14 weeks of gestation. Prenat. Diagn. 2000, 20, 673–675. [Google Scholar] [CrossRef] [PubMed]
  121. Finkel, T. Signal transduction by reactive oxygen species. J. Cell Biol. 2011, 194, 7–15. [Google Scholar] [CrossRef] [PubMed]
  122. Menon, R. Oxidative Stress Damage as a Detrimental Factor in Preterm Birth Pathology. Front. Immunol. 2014, 5, 567. [Google Scholar] [CrossRef] [PubMed]
  123. Al-Gubory, K.H.; Fowler, P.A.; Garrel, C. The roles of cellular reactive oxygen species, oxidative stress and antioxidants in pregnancy outcomes. Int. J. Biochem. Cell Biol. 2010, 42, 1634–1650. [Google Scholar] [CrossRef] [PubMed]
  124. Jauniaux, E.; Watson, A.L.; Hempstock, J.; Bao, Y.P.; Skepper, J.N.; Burton, G.J. Onset of maternal arterial blood flow and placental oxidative stress. A possible factor in human early pregnancy failure. Am. J. Pathol. 2000, 157, 2111–2122. [Google Scholar] [CrossRef] [PubMed]
  125. Wong, J.L.; Créton, R.; Wessel, G.M. The Oxidative Burst at Fertilization Is Dependent upon Activation of the Dual Oxidase Udx1. Dev. Cell 2004, 7, 801–814. [Google Scholar] [CrossRef] [PubMed]
  126. Nakamura, M.; Sekizawa, A.; Purwosunu, Y.; Okazaki, S.; Farina, A.; Wibowo, N.; Shimizu, H.; Okai, T. Cellular mRNA expressions of anti-oxidant factors in the blood of preeclamptic women. Prenat. Diagn. Publ. Affil. Int. Soc. Prenat. Diagn. 2009, 29, 691–696. [Google Scholar] [CrossRef] [PubMed]
  127. Gupta, S.; Agarwal, A.; Banerjee, J.; Alvarez, J.G. The role of oxidative stress in spontaneous abortion and recurrent pregnancy loss: A systematic review. Obs. Gynecol. Surv. 2007, 62, 335–347. [Google Scholar] [CrossRef] [PubMed]
  128. Murphy, M.P. How mitochondria produce reactive oxygen species. Biochem. J. 2009, 417, 1–13. [Google Scholar] [CrossRef] [PubMed]
  129. Mirończuk-Chodakowska, I.; Witkowska, A.M.; Zujko, M.E. Endogenous non-enzymatic antioxidants in the human body. Adv. Med. Sci. 2018, 63, 68–78. [Google Scholar] [CrossRef] [PubMed]
  130. Agarwal, A.; Aponte-Mellado, A.; Premkumar, B.J.; Shaman, A.; Gupta, S. The effects of oxidative stress on female reproduction: A review. Reprod. Biol. Endocrinol. 2012, 10, 49. [Google Scholar] [CrossRef] [PubMed]
  131. Balaban, R.S.; Nemoto, S.; Finkel, T. Mitochondria, Oxidants, and Aging. Cell 2005, 120, 483–495. [Google Scholar] [CrossRef] [PubMed]
  132. Subash, P.; Gurumurthy, P.; Sarasabharathi, A.; Cherian, K. Urinary 8-OHdG: A marker of oxidative stress to DNA and total antioxidant status in essential hypertension with South Indian population. Indian J. Clin. Biochem. 2010, 25, 127–132. [Google Scholar] [CrossRef] [PubMed]
  133. Negi, R.; Pande, D.; Kumar, A.; Khanna, R.S.; Khanna, H.D. In vivo oxidative DNA damage and lipid peroxidation as a biomarker of oxidative stress in preterm low-birthweight infants. J. Trop. Pediatr. 2012, 58, 326–328. [Google Scholar] [CrossRef] [PubMed]
  134. Abdel Ghany, E.A.G.; Alsharany, W.; Ali, A.A.; Youness, E.R.; Hussein, J.S. Anti-oxidant profiles and markers of oxidative stress in preterm neonates. Paediatr. Int. Child Health 2016, 36, 134–140. [Google Scholar] [CrossRef] [PubMed]
  135. Mustafa, M.; Pathak, R.; Ahmed, T.; Ahmed, R.S.; Tripathi, A.; Guleria, K.; Banerjee, B. Association of glutathione S-transferase M1 and T1 gene polymorphisms and oxidative stress markers in preterm labor. Clin. Biochem. 2010, 43, 1124–1128. [Google Scholar] [CrossRef] [PubMed]
  136. Diaz-Castro, J.; Pulido-Moran, M.; Moreno-Fernandez, J.; Kajarabille, N.; de Paco, C.; Garrido-Sanchez, M.; Prados, S.; Ochoa, J.J. Gender specific differences in oxidative stress and inflammatory signaling in healthy term neonates and their mothers. Pediatr. Res. 2016, 80, 595–601. [Google Scholar] [CrossRef] [PubMed]
  137. Borrás, C.; Sastre, J.; García-Sala, D.; Lloret, A.; Pallardó, F.V.; Viña, J. Mitochondria from females exhibit higher antioxidant gene expression and lower oxidative damage than males. Free Radic. Biol. Med. 2003, 34, 546–552. [Google Scholar] [CrossRef] [PubMed]
  138. Marazziti, D.; Baroni, S.; Mucci, F.; Piccinni, A.; Moroni, I.; Giannaccini, G.; Carmassi, C.; Massimetti, E.; Dell’Osso, L. Sex-Related Differences in Plasma Oxytocin Levels in Humans. Clin. Pr. Epidemiol. Ment. Health 2019, 15, 58–63. [Google Scholar] [CrossRef] [PubMed]
  139. Andersen, H.R.; Nielsen, J.B.; Nielsen, F.; Grandjean, P. Antioxidative enzyme activities in human erythrocytes. Clin. Chem. 1997, 43, 562–568. [Google Scholar] [CrossRef] [PubMed]
  140. Sugioka, K.; Shimosegawa, Y.; Nakano, M. Estrogens as natural antioxidants of membrane phospholipid peroxidation. FEBS Lett. 1987, 210, 37–39. [Google Scholar] [CrossRef] [PubMed]
  141. Richardson, B.S.; Rajagopaul, A.; de Vrijer, B.; Eastabrook, G.; Regnault, T.R.H. Fetal sex impacts birth to placental weight ratio and umbilical cord oxygen values with implications for regulatory mechanisms. Biol. Sex Differ. 2022, 13, 35. [Google Scholar] [CrossRef] [PubMed]
  142. Li, Y.; Yao, B.; Men, J.; Pang, Y.; Gao, J.; Bai, Y.; Wang, H.; Zhang, J.; Zhao, L.; Xu, X.; et al. Oxidative stress and energy metabolism in male reproductive damage from single and combined high-power microwave exposure at 1.5 and 4.3 GHz. Reprod. Toxicol. 2025, 132, 108759. [Google Scholar] [CrossRef] [PubMed]
  143. Lien, Y.-C.; Zhang, Z.; Cheng, Y.; Polyak, E.; Sillers, L.; Falk, M.J.; Ischiropoulos, H.; Parry, S.; Simmons, R.A. Human placental transcriptome reveals critical alterations in inflammation and energy metabolism with fetal sex differences in spontaneous preterm birth. Int. J. Mol. Sci. 2021, 22, 7899. [Google Scholar] [CrossRef] [PubMed]
  144. Akram, K.M.; Kulkarni, N.S.; Brook, A.; Wyles, M.D.; Anumba, D.O.C. Transcriptomic analysis of the human placenta reveals trophoblast dysfunction and augmented Wnt signalling associated with spontaneous preterm birth. Front. Cell Dev. Biol. 2022, 10, 987740. [Google Scholar] [CrossRef] [PubMed]
  145. Greenough, A.; Lagercrantz, H.; Pool, J.; Dahlin, I. Plasma catecholamine levels in preterm infants: Effect of birth asphyxia and Apgar score. Acta Pædiatrica 1987, 76, 54–59. [Google Scholar] [CrossRef] [PubMed]
  146. El-Khodor, B.F.; Boksa, P. Differential vulnerability of male versus female rats to long-term effects of birth insult on brain catecholamine levels. Exp. Neurol. 2003, 182, 208–219. [Google Scholar] [CrossRef] [PubMed]
  147. Saif, Z.; Hodyl, N.; Hobbs, E.; Tuck, A.; Butler, M.; Osei-Kumah, A.; Clifton, V. The human placenta expresses multiple glucocorticoid receptor isoforms that are altered by fetal sex, growth restriction and maternal asthma. Placenta 2014, 35, 260–268. [Google Scholar] [CrossRef] [PubMed]
  148. Hodyl, N.; Stark, M.; Butler, M.; Clifton, V. Placental P-glycoprotein is unaffected by timing of antenatal glucocorticoid therapy but reduced in SGA preterm infants. Placenta 2013, 34, 325–330. [Google Scholar] [CrossRef] [PubMed]
  149. Clifton, V.L.; McDonald, M.; Morrison, J.L.; Holman, S.L.; Lock, M.C.; Saif, Z.; Meakin, A.; Wooldridge, A.L.; Gatford, K.L.; Wallace, M.J. Placental glucocorticoid receptor isoforms in a sheep model of maternal allergic asthma. Placenta 2019, 83, 33–36. [Google Scholar] [CrossRef] [PubMed]
  150. Clifton, V.; Cuffe, J.; Moritz, K.; Cole, T.; Fuller, P.; Lu, N.; Kumar, S.; Chong, S.; Saif, Z. The role of multiple placental glucocorticoid receptor isoforms in adapting to the maternal environment and regulating fetal growth. Placenta 2017, 54, 24–29. [Google Scholar] [CrossRef] [PubMed]
  151. Woods, L.; Perez-Garcia, V.; Hemberger, M. Regulation of placental development and its impact on fetal growth—New insights from mouse models. Front. Endocrinol. 2018, 9, 570. [Google Scholar] [CrossRef] [PubMed]
  152. Meakin, A.S.; Cuffe, J.S.M.; Darby, J.R.T.; Morrison, J.L.; Clifton, V.L. Let’s Talk about Placental Sex, Baby: Understanding Mechanisms That Drive Female- and Male-Specific Fetal Growth and Developmental Outcomes. Int. J. Mol. Sci. 2021, 22, 6386. [Google Scholar] [CrossRef] [PubMed]
  153. Oaks, B.M.; Adu-Afarwuah, S.; Ashorn, P.; Lartey, A.; Laugero, K.D.; Okronipa, H.; Stewart, C.P.; Dewey, K.G. Increased risk of preterm delivery with high cortisol during pregnancy is modified by fetal sex: A cohort study. BMC Pregnancy Childbirth 2022, 22, 727. [Google Scholar] [CrossRef] [PubMed]
  154. Cooperstock, M.; Campbell, J. Excess males in preterm birth: Interactions with gestational age, race, and multiple birth. Obs. Gynecol. 1996, 88, 189–193. [Google Scholar] [CrossRef] [PubMed]
  155. Zhang, G.; Feenstra, B.; Bacelis, J.; Liu, X.; Muglia, L.M.; Juodakis, J.; Miller, D.E.; Litterman, N.; Jiang, P.-P.; Russell, L. Genetic associations with gestational duration and spontaneous preterm birth. N. Engl. J. Med. 2017, 377, 1156–1167. [Google Scholar] [CrossRef] [PubMed]
  156. Hasegawa, K.; Kumasaka, N.; Nakabayashi, K.; Kamura, H.; Maehara, K.; Kasuga, Y.; Hata, K.; Tanaka, M. Genome-wide association study of preterm birth and gestational age in a Japanese population. Hum. Genome Var. 2023, 10, 19. [Google Scholar] [CrossRef] [PubMed]
  157. Moslehi, R.; Singh, R.; Lessner, L.; Friedman, J.M. Impact of BRCA mutations on female fertility and offspring sex ratio. Am. J. Hum. Biol. Off. J. Hum. Biol. Assoc. 2010, 22, 201–205. [Google Scholar] [CrossRef] [PubMed]
Figure 1. External factors affecting pathogenic preterm birth. The following scheme illustrates how related risk factors affect the pathophysiology of PTB. Several factors have been identified as contributing to placental dysfunction, including external environmental factors (e.g., particulate matter, heavy metals, and temperature), as well as infections (e.g., bacteria), smoking, malnutrition, a high-fat diet, stress, socioeconomic status, and ethnicity. These factors have been linked to being associated with increased inflammation, oxidative stress, and other related complications. It is evident that hormonal signaling, specifically estrogen, testosterone, and progesterone (P4), plays a pivotal role in the initiation of labor. Spontaneous preterm labor leading to preterm birth is a complex syndrome consisting of several diseases, each of which can be an independent initiating factor in the path to labor induction. All of these disease processes can induce inflammation and oxidative stress. The figure was created with BioRender.com, accessed on 2 June 2025.
Figure 1. External factors affecting pathogenic preterm birth. The following scheme illustrates how related risk factors affect the pathophysiology of PTB. Several factors have been identified as contributing to placental dysfunction, including external environmental factors (e.g., particulate matter, heavy metals, and temperature), as well as infections (e.g., bacteria), smoking, malnutrition, a high-fat diet, stress, socioeconomic status, and ethnicity. These factors have been linked to being associated with increased inflammation, oxidative stress, and other related complications. It is evident that hormonal signaling, specifically estrogen, testosterone, and progesterone (P4), plays a pivotal role in the initiation of labor. Spontaneous preterm labor leading to preterm birth is a complex syndrome consisting of several diseases, each of which can be an independent initiating factor in the path to labor induction. All of these disease processes can induce inflammation and oxidative stress. The figure was created with BioRender.com, accessed on 2 June 2025.
Cells 14 01084 g001
Figure 2. Modulation of inflammation-related pathway by sex steroid hormones in response to infection. This schematic illustrates the immune signaling cascade initiated by pathogen recognition and its modulation by sex steroid hormones―oestradiol (pink), androgens (blue), and progesterone (green). PAMPs (e.g., from bacteria, viruses, fungi, and parasites) are recognized by dendritic cells, leading to the activation of toll-like receptors (TLRs) and the downstream adaptor myeloid differentiation primary response 88 (MyD88). This signaling induces the expression of pro-inflammatory cytokines (e.g., interleukin (IL)-1β, IL-6, tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ)) and chemokines (e.g., C-C motif chemokine ligand (CCL2), CCL3, C-X-C motif chemokine ligand 8 (CXCL8)), which promote leukocyte recruitment. Leukocytes contribute to the production of prostaglandins (e.g., PGE2, PGF2α, PGD2) via cyclooxygenase-2 (COX-2), which in turn activates matrix metalloproteinase-9 (MMP-9). This pathway may facilitate extracellular matrix remodeling and placental membrane contraction, events implicated in the initiation of labor and preterm birth. Sex steroid hormones differentially regulate these processes: oestradiol upregulates pro-inflammatory mediators and prostaglandin synthesis; androgens and progesterone generally suppress TLR signaling, cytokine/chemokine expression, COX-2 activity, and MMP-9 activation. Abbreviations: AB, antibody; CCL, C-C motif chemokine ligand; CXCL, C-X-C motif chemokine ligand; DCs, dendritic cells; E, oestradial; IFN-γ, interferon-γ; Ig, immunoglobulin; IL, interleukin; MMP-9, matrix metalloproteinase-9; PGD, prostaglandin; PGE, prostaglandin E; PGF, prostaglandin F; TGFβ, transforming growth factor β; TLR, toll-like receptor; TNF, tumor necrosis factor.
Figure 2. Modulation of inflammation-related pathway by sex steroid hormones in response to infection. This schematic illustrates the immune signaling cascade initiated by pathogen recognition and its modulation by sex steroid hormones―oestradiol (pink), androgens (blue), and progesterone (green). PAMPs (e.g., from bacteria, viruses, fungi, and parasites) are recognized by dendritic cells, leading to the activation of toll-like receptors (TLRs) and the downstream adaptor myeloid differentiation primary response 88 (MyD88). This signaling induces the expression of pro-inflammatory cytokines (e.g., interleukin (IL)-1β, IL-6, tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ)) and chemokines (e.g., C-C motif chemokine ligand (CCL2), CCL3, C-X-C motif chemokine ligand 8 (CXCL8)), which promote leukocyte recruitment. Leukocytes contribute to the production of prostaglandins (e.g., PGE2, PGF2α, PGD2) via cyclooxygenase-2 (COX-2), which in turn activates matrix metalloproteinase-9 (MMP-9). This pathway may facilitate extracellular matrix remodeling and placental membrane contraction, events implicated in the initiation of labor and preterm birth. Sex steroid hormones differentially regulate these processes: oestradiol upregulates pro-inflammatory mediators and prostaglandin synthesis; androgens and progesterone generally suppress TLR signaling, cytokine/chemokine expression, COX-2 activity, and MMP-9 activation. Abbreviations: AB, antibody; CCL, C-C motif chemokine ligand; CXCL, C-X-C motif chemokine ligand; DCs, dendritic cells; E, oestradial; IFN-γ, interferon-γ; Ig, immunoglobulin; IL, interleukin; MMP-9, matrix metalloproteinase-9; PGD, prostaglandin; PGE, prostaglandin E; PGF, prostaglandin F; TGFβ, transforming growth factor β; TLR, toll-like receptor; TNF, tumor necrosis factor.
Cells 14 01084 g002
Figure 3. Sex-specific placental adaptation and fetal growth strategies in response to intrauterine stress. The schematic illustrates differential placental and fetal responses in pregnancies carrying male (blue) versus female (pink) fetuses. Female placentas exhibit adaptive mechanisms including increased glucocorticoid receptor (GR)-mediated signaling, reduced placental growth factor (PlGF) expression under hypoxia, and enhanced trophoblast immune modulation with reduced labyrinth development. Male fetuses exhibit a growth-prioritizing strategy characterized by decreased GR-mediated signaling, increased expression of hypoxia-inducible factor (HIF)-1α and apoptotic marker, and androgen-driven signaling. Namely, rather than diminishing the placental adaptability of male placentas under adverse intrauterine conditions, an optimal environment for fetal growth is facilitated. The figure was created with BioRender.com, accessed on 2 June 2025.
Figure 3. Sex-specific placental adaptation and fetal growth strategies in response to intrauterine stress. The schematic illustrates differential placental and fetal responses in pregnancies carrying male (blue) versus female (pink) fetuses. Female placentas exhibit adaptive mechanisms including increased glucocorticoid receptor (GR)-mediated signaling, reduced placental growth factor (PlGF) expression under hypoxia, and enhanced trophoblast immune modulation with reduced labyrinth development. Male fetuses exhibit a growth-prioritizing strategy characterized by decreased GR-mediated signaling, increased expression of hypoxia-inducible factor (HIF)-1α and apoptotic marker, and androgen-driven signaling. Namely, rather than diminishing the placental adaptability of male placentas under adverse intrauterine conditions, an optimal environment for fetal growth is facilitated. The figure was created with BioRender.com, accessed on 2 June 2025.
Cells 14 01084 g003
Figure 4. Reactive oxygen species (ROS) generation and signaling pathways. ROS are generated during cellular aerobic metabolism as a result of oxygen (O2) consumption. Superoxide radicals (O2) are produced by the single electron (e) reduction of O2, catalyzed by enzymes such as NADPH oxidase, xanthine oxidase, and as a byproduct of mitochondrial electron transport. Superoxide dismutase (SOD) catalyzes the dismutation of O2 into hydroxyl peroxide (H2O2). H2O2 can stimulate signaling pathways such as NF-κB, MAPK, and NLRP3 inflammasome, which are associated with inflammatory responses and may induce parturition through the expression of cyclooxygenase (COX)-2 and pro-inflammatory cytokines. H2O2 can also be converted to more ROS, such as hydroperoxyl (HOO˙) and hydroxyl radicals (HO˙) via the Fenton reaction. Antioxidant enzymes like catalase and glutathione peroxidase (GPx) degrade H2O2 into water (H2O), limiting oxidative stress. Excess ROS can stimulate TLR-4 signaling and cause DNA damage, such as by the oxidized derivative of deoxyguanosine, 8-oxodeoxyguanosine (8-OHdG), and lipid peroxidation [128,129,130,131].
Figure 4. Reactive oxygen species (ROS) generation and signaling pathways. ROS are generated during cellular aerobic metabolism as a result of oxygen (O2) consumption. Superoxide radicals (O2) are produced by the single electron (e) reduction of O2, catalyzed by enzymes such as NADPH oxidase, xanthine oxidase, and as a byproduct of mitochondrial electron transport. Superoxide dismutase (SOD) catalyzes the dismutation of O2 into hydroxyl peroxide (H2O2). H2O2 can stimulate signaling pathways such as NF-κB, MAPK, and NLRP3 inflammasome, which are associated with inflammatory responses and may induce parturition through the expression of cyclooxygenase (COX)-2 and pro-inflammatory cytokines. H2O2 can also be converted to more ROS, such as hydroperoxyl (HOO˙) and hydroxyl radicals (HO˙) via the Fenton reaction. Antioxidant enzymes like catalase and glutathione peroxidase (GPx) degrade H2O2 into water (H2O), limiting oxidative stress. Excess ROS can stimulate TLR-4 signaling and cause DNA damage, such as by the oxidized derivative of deoxyguanosine, 8-oxodeoxyguanosine (8-OHdG), and lipid peroxidation [128,129,130,131].
Cells 14 01084 g004
Table 1. The various environmental factors related to pregnancy complications.
Table 1. The various environmental factors related to pregnancy complications.
Environmental FactorsExposure WindowStudy (Author, Year)Cohort
(Ethnicities)
Sample SizeTrend in Fetal SexSignificanceKey Findings/Conclusion
NO2T2Cossi et al., 2015
[28]
San Joaquin Valley of California
(Hispanic)
253,704M > Fp < 0.01Exposure to NO2 during T2 was associated with a high risk of PTB (GW 20–27) in M infant.
PM2.5Entire, T1Park et al., 2023
[29]
Retrospective birth cohort
(Korean)
1880M > Fp = 0.01,
p < 0.01
The higher risk of LBW was associated with exposure to PM2.5 during T1 (OR:1.05 [95% CI: 1.01–1.10]) and
T2 (OR: 1.07 [95% CI; 1.03–1.12]).
SmokingEntire
(Survey)
Günther et al., 2020
[30]
Database of Schleswig-Holstein
(German)
220,339M > Fp < 0.001The rate of PTB subdivided into the smoking severity.
M > F for nonsmokers;
M > F for: 1–7 cigarettes/day;
M > F: 8–14 cigarettes/day;
M > F: 15–21 cigarettes/day;
M = F: ≥22 cigarettes/day.
SmokingEntire
(Survey)
Voigt et al., 2006
[31]
German birth statistics from Deutsche Perinatalerhebung
(German)
888,632M < Fp < 0.001Severe smokers (>21 cigarettes/day) have a higher risk for SGA in F (3.51-fold) and in M (3.15-fold) vs. non-smoker.
In mild smokers (1–5/day), the risk of SGA was 1.7275-fold in F, but was 1.7143-fold in M.
AlcoholNot
suggested
Flannigan et al., 2023 [32]Canada2574FASD w/wo SFF:
M = F
NDF:
M > F
EP:
M < F
p < 0.001M = F: FASD diagnostic
F: ↑EP anxiety, ↑depressive/mood disorders, ↑trauma.
M: ↑NDF impairment, ↑ADHD, ↑conduct disorder, ↑oppositional, ↑defiant disorder.
The differences were clearest in adolescents (13–17 years) and adults (≥25 years).
Heat waves4-day
(or 7-day) #
Darrow et al., 2024
[33]
National Vital Statistics System at the National Center for Health Statistics Data (##)55,748,869M < FRR (95% CI)
F: 1.011
(1.001–1.020)
M: 1.006
(0.997–1.015)
Subgroup analysis of RR per 1 °C increase.
F: PTB and early PTB > 1
M: PTB RR > 1.
Extreme temperature1 to 2 weeks before
delivery
Yu et al., 2023
[34]
Large-scale multicenter study
(Chinese)
82,221M < FOR (95% CI)
### Fifth: 1.09 (1.04, 1.13)
10th day: 1.07 (1.04, 1.12)
10th 2D: 1.13 (1.04, 1.23)
F > M: cold spells;
heat waves, ↑northern and western regions in China.
Exposure to cold spells was relevant with ↑risk of PTB, especially late.
# over the threshold during the exposure window, a continuous variable calculated as the 4-day (or 7-day) moving mean—the 97.5% threshold. ## Hispanic or non-Hispanic ethnicity and Alaska Native, American Indian, Asian, Black, Other Pacific Islander. ### 5th days, 10th days, and 10th 2D of 1 week before delivery. Abbreviations: ADHD, attention deficit hyperactivity disorder; CI, confidence interval; EP, endocrine problem; F, female; FASD, fetal alcohol spectrum disorder; GW, gestational week at; LBW, low birth weight; OR, odds ratio; p, p-value; PTB, preterm birth; SGA, small gestational age; PM2.5, particulate matter 2.5; RR, risk ratio; T1, 1st trimester; T2, 2nd trimester; M, male; NDF, neurodevelopmental functioning; NO2, nitrogen dioxide.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lee, G.; Andrade, G.M.; Kim, Y.J.; Anumba, D.O.C. The Sex Difference in the Pathophysiology of Preterm Birth. Cells 2025, 14, 1084. https://doi.org/10.3390/cells14141084

AMA Style

Lee G, Andrade GM, Kim YJ, Anumba DOC. The Sex Difference in the Pathophysiology of Preterm Birth. Cells. 2025; 14(14):1084. https://doi.org/10.3390/cells14141084

Chicago/Turabian Style

Lee, Gain, Gisela Martinez Andrade, Young Ju Kim, and Dilly O. C. Anumba. 2025. "The Sex Difference in the Pathophysiology of Preterm Birth" Cells 14, no. 14: 1084. https://doi.org/10.3390/cells14141084

APA Style

Lee, G., Andrade, G. M., Kim, Y. J., & Anumba, D. O. C. (2025). The Sex Difference in the Pathophysiology of Preterm Birth. Cells, 14(14), 1084. https://doi.org/10.3390/cells14141084

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop