Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Endocrinol., 22 May 2025

Sec. Translational and Clinical Endocrinology

Volume 16 - 2025 | https://doi.org/10.3389/fendo.2025.1606654

Bisphenols exposure and non-alcoholic fatty liver disease: from environmental trigger to molecular pathogenesis

  • 1Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
  • 2Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
  • 3Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China

Bisphenols (BPs), including bisphenol A (BPA) and its substitutes (BPS, BPF), are ubiquitous environmental contaminants with emerging links to metabolic disorders. This review synthesizes current evidence on the role of BP exposure in the pathogenesis of non-alcoholic fatty liver disease (NAFLD), a global health crisis affecting 25% of adults worldwide. Epidemiological studies reveal significant positive associations between urinary/serum BP levels and NAFLD risk, particularly in males, with maternal exposure correlating to transgenerational metabolic dysfunction. Mechanistically, BPs disrupt hepatic lipid homeostasis by activating PPAR-γ and suppressing fatty acid oxidation while concurrently inducing insulin resistance via impaired IRS-1/PI3K/Akt signaling. Oxidative stress, NLRP3 inflammasome activation, and gut-liver axis perturbations further exacerbate steatosis and inflammation. Co-exposure with phthalates or high-fat diets amplifies hepatotoxicity, highlighting synergistic environmental risks. Critically, developmental and sex-specific susceptibility underscores the need for tailored interventions. We propose preventive strategies to mitigate NAFLD progression, including BP avoidance and policy reforms. This work bridges gaps between environmental epidemiology and molecular toxicology, emphasizing BPs as modifiable drivers of metabolic liver disease.

1 Introduction

Non-alcoholic fatty liver disease (NAFLD) has emerged as a global health crisis, affecting approximately 25% of the worldwide population, with its progression to non-alcoholic steatohepatitis (NASH) and cirrhosis posing significant clinical challenges (1, 2). While genetic predisposition and metabolic syndromes like obesity are well-established risk factors (3), mounting evidence underscores the critical role of environmental endocrine-disrupting chemicals (EDCs), particularly bisphenols, in modulating NAFLD pathogenesis (46). Among these, bisphenol A (BPA) and its analogs (e.g., BPS, BPF) are ubiquitous environmental triggers, leaching from polycarbonate plastics, food packaging, and thermal paper into human ecosystems, resulting in detectable serum levels in >90% of the general population (7).

Recent epidemiological studies reveal a dose-dependent association between urinary BPA levels and NAFLD severity, independent of traditional risk factors like BMI (8). Mechanistically, BPA disrupts hepatic lipid homeostasis through two distinct mechanisms: (1) estrogen receptor (ER) antagonism, impairing lipid oxidation and promoting ectopic fat deposition (9, 10), and (2) NLRP3 inflammasome activation, driving pro-inflammatory cytokine release (e.g., TNF-α, IL-1β, and IL-6) that exacerbates hepatic insulin resistance (11, 12). Notably, emerging data suggest that BPA’s effects extend beyond direct hepatotoxicity, involving gut-liver axis dysregulation—via gut microbiota-derived metabolites like secondary bile acids and trimethylamine-N-oxide (TMAO)—that synergistically potentiates hepatic de novo lipogenesiss and oxidative stress (13, 14).

Despite these advances, critical knowledge gaps persist: (1) Temporal Dynamics: Most studies focus on acute BPA exposure, neglecting chronic low-dose effects that mimic real-world scenarios; (2) Epigenetic Modulation: BPA-induced DNA methylation changes in genes regulating lipid metabolism remain underexplored in NAFLD progression; (3) Cumulative Exposures: Synergistic interactions between bisphenols and other environmental stressors (e.g., microplastics, heavy metals) are poorly characterized but may explain geographic disparities in NAFLD prevalence.

This review bridges these critical knowledge gaps by systematically integrating epidemiological insights with mechanistic toxicological evidence to elucidate the multifaceted role of BPs in NAFLD pathogenesis. Herein, we conducted rigor methodology in literature selection (Figure 1): (1) Systematic Search Protocol Databases: We conducted searches in PubMed and MEDLINE using controlled vocabulary (e.g., MeSH terms: “Bisphenols,” “NAFLD,” “Environmental Exposure”) and free-text keywords (e.g., “endocrine disruptors,” “hepatic steatosis,” “PPAR-γ”). (2) Timeframe: Focused on 2000–2025 to capture modern exposure patterns and molecular mechanistic insights. (3) Inclusion Criteria: Human epidemiological studies (cross-sectional, cohort) and experimental models (rodent, zebrafish, cell lines); Studies reporting quantifiable BP exposure levels (urinary/serum biomarkers); Mechanistic data on lipid metabolism, insulin signaling, or oxidative stress pathways. (4) Exclusion Criteria: Reviews without original data Studies lacking control groups or exposure quantification Non-English publications (to ensure quality interpretation). We synthesize emerging data on chronic low-dose BP exposure effects, emphasizing non-monotonic dose-response relationships and developmental windows of susceptibility. A key focus is on epigenetic reprogramming mechanisms, particularly BP-induced DNA methylation changes in lipid regulatory genes and their transgenerational metabolic consequences. Furthermore, we characterize synergistic hepatotoxicity arising from co-exposures to BPs and other environmental stressors, such as phthalates and high-fat diets, which amplify oxidative stress and gut-liver axis dysfunction. By contextualizing molecular pathways within population-level exposure gradients—including occupational hazards and socioeconomic disparities in BP exposure—this work advances a unified framework linking environmental triggers to clinical NAFLD phenotypes. Finally, we propose evidence-based preventive strategies, from BP-free product alternatives to policy reforms regulating cumulative exposures, while identifying priority research directions for mitigating the global burden of metabolic liver diseases.

Figure 1
www.frontiersin.org

Figure 1. PRISMA flow diagram for study selection.

2 Environmental exposure to bisphenols

2.1 Ubiquitous sources and exposure routes

BPA and its analogs (e.g., BPS, BPF and BPAF) are extensively used in polycarbonate plastics, epoxy resins, thermal paper receipts, food packaging, and dental sealants (1517). For example, BPA-free alternatives like phenol stearic acid-based polyether resins are increasingly adopted in coatings, though their long-term safety remains understudied (18). Non-dietary sources include household dust, personal care products, and occupational settings (e.g., cashiers handling thermal paper receipts).

Humans can be exposed to BPA and its analogs by common pathways (summarized in Figure 2): (1) Dietary intake: Leaching of BPs from food containers and canned goods into foodstuffs, especially under high-temperature conditions (19); (2) Dermal absorption: direct skin contact with thermal paper (e.g., receipts) or cosmetics containing BP derivatives (20); (3) Inhalation: inhalation of BP-laden dust particles in indoor environments (21) and (4) Transplacental and lactational transfer: maternal exposure leads to fetal and infant exposure via placental circulation and breast milk (22).

Figure 2
www.frontiersin.org

Figure 2. Four common pathways humans can be exposed to BPA and its analogs.

2.2 Biomarkers and population-level burden

Biomonitoring evidence demonstrates pervasive human exposure to BPs. BPs and their phase II metabolites (e.g., BPA-glucuronide) are routinely detected in human biofluids, with detection rates exceeding 95% in urine, serum, and breast milk samples across global populations (23, 24). Occupational exposure gradients are particularly striking: urinary BP concentrations in cashiers and factory workers handling BP-containing thermal paper or epoxy resins are 3- to 5-fold higher than in the general population, reflecting direct dermal and inhalation exposure pathways.

Concerningly, three vulnerable subpopulations are facing disproportionate risks induced by BPs: Firstly, children exhibit elevated exposure per unit body weight due to developmentally driven behaviors (e.g., frequent hand-to-mouth contact) coupled with immature hepatic detoxification systems that prolong BP half-lives (25, 26); Secondly, low-income communities experience dual burdens: limited access to BP-free alternatives and dependence on processed/packaged foods (primary dietary sources of BP migration from can linings and plastic containers) (27, 28); Moreover, pregnant women represent a critical susceptibility window, as transplacental BP transfer during fetal developmental programming phases may induce epigenetic alterations with lifelong health consequences (29).

2.3 Temporal trends and regulatory gaps

The shift from BPA due to regulatory restrictions (e.g., bans on baby bottles) has led to the adoption of structurally similar analogs such as BPS and BPF (30). However, these substitutes exhibit comparable endocrine-disrupting properties, and their long-term toxicological profiles remain poorly characterized. Emerging alternatives like bisphenol AP (BPAP) and bisphenol AF (BPAF) are increasingly detected in environmental samples, yet their safety and health impacts are understudied.

A significant challenge in regulating BPs lies in the lack of harmonized standards. Current regulations focus on individual BPs (e.g., BPA) but fail to address the cumulative effects of co-exposure to multiple analogs. Additionally, biomonitoring efforts are hindered by the short half-lives of BP metabolites (e.g., less than 6 hours for BPA), complicating accurate exposure assessment in epidemiological studies (31). Another critical gap is the insufficient interdisciplinary collaboration among environmental science, toxicology, and public health, which weakens the scientific foundation for risk assessments and regulatory measures. Furthermore, public awareness of BP risks remains limited, highlighting the need for targeted health education initiatives.

Future research should prioritize comprehensive toxicological studies on emerging BP substitutes, particularly their long-term health effects. Policymakers must develop unified regulatory standards for cumulative exposures and improve biomonitoring technologies to assess population-level risks better. Addressing these gaps is essential to mitigate the health impacts of BPs and their analogs.

3 Epidemiological evidence linking BPs to NAFLD

A growing body of epidemiological studies has established significant associations between BPs exposure and NAFLD risk across diverse populations. Key findings from large-scale human studies are summarized below (Table 1).

Table 1
www.frontiersin.org

Table 1. Epidemiological evidence linking bisphenol exposure to NAFLD risk.

3.1 Dose-response relationships in adolescent and adult populations

In U.S. cohorts, previously in 2018, Sofia et al. utilized National Health and Nutrition Examination Survey (NHANES) data (2003-2010) on 12-19-year-old adolescents. They incorporated a total of 944 urinary BPA and fasting laboratory tests from a total of 7168 adolescents. They calculated that the risk of suspected NAFLD was increased in the second quartile of BPA levels (1.4-2.7 ng/mL) when compared to the first (< 1.4 ng/mL) (Odds Ratio (OR) 4.23, 95% Confidence Interval (CI) 1.44-12.41) (32). Interestingly, the association was stronger in Hispanics (n = 344) with BPA levels in the second (OR 6.12, 95% CI 1.62-23.15) quartile and when limiting the analyses to overweight/obese adolescents (n = 332), in the second (OR 5.56, 95% CI 1.28-24.06) and fourth BPA quartiles (OR 6.85, 95% CI: 1.02-46.22) compared to the first quartile. However, BPA levels were not associated with ALT elevation, indicating that laboratory indexes could not estimate BPA-induced hepatoxicity.

Apart from teenagers, in 2019, Donghee and colleagues conducted a cross-sectional analysis of data from the NHANES database (2005 - 2014) focusing on in the US adults (33). Among the initial 7605 participants (with a mean age of 47 years and 48.4% being male), a correlation was found between the prevalence of NAFLD and abnormally elevated ALT levels and urinary BPA levels (P < 0.05). Compared to the reference group with the lowest urinary BPA levels, individuals in the third and fourth quartiles had an 81% and 53% higher likelihood of developing NAFLD, as defined by the hepatic steatosis index (HIS). In a multivariate model, the ORs for NAFLD in the third and fourth quartiles were 1.69 (95% CI (36): 1.39 - 2.04) and 1.44 (95% CI: 1.19-1.76) respectively (p< 0.001).

Similarly to U.S. cohorts, Yang’s research team consistently conducted the Korean National Environmental Health Survey (KoNEHS, 2015-2017) involving 3,476 adults (1,474 men and 2,002 women) (34). In their research, abdominal ultrasonography (hyperechoic liver parenchyma and vessel blurring) was considered as the gold-standard imaging criterion (37). Their findings illustrated a linear association between urinary BPA concentrations and NAFLD risk. In a univariate analysis, the OR for NAFLD in the highest quartile of urinary BPA levels was found to be 1.47 [95% CI: 1.11-1.94] compared to the lowest quartile. After adjusting for covariates, the ORs for NAFLD in the third and fourth quartiles were determined to be 1.31 [95% CI: 1.03-1.67] and 1.32 [95% CI: 1.03-1.70], respectively.

Similarly, a cross-sectional study (n=960 U.S. adults) using the Hepatic Steatosis Index (HSI) [HSI > 36 was used to predict NAFLD (38)] identified 1.42-fold (BPA, 95% CI: 1.11–1.82) and 1.31-fold (BPS, 95% CI: 1.02–1.68) elevated NAFLD risks per unit increase in urinary BP concentrations. Mediation analyses identified insulin resistance pathways as accounting for 18.7-23.1% of the observed risk elevation (8). Similarly, Chinese cohorts showed striking serum BPS-associated NAFLD risks (OR=3.16 per unit increase, 95% CI: 2.81–3.55) (35).

3.2 Critical vulnerable subpopulations

BPs induced the development of NAFLD partially due to their sex-specific effects. Meta-analyses highlight stronger BP-NAFLD associations in males, potentially linked to androgen receptor crosstalk with estrogenic BP metabolites (8, 33, 39, 40). Concerningly, here exists socioeconomic disparities. Low-income populations face dual exposure risks due to greater dependence on processed/packaged foods (primary sources of BP migration) coupled with limited availability of BP-free alternatives (27, 28).

3.3 Methodological limitations and unresolved questions

Most studies are cross-sectional, precluding causal conclusions. Longitudinal data tracking BP exposure and liver outcomes (e.g., fibrosis, cirrhosis) over decades are urgently needed. Additionally, short half-lives of BP metabolites (e.g., BPA-glucuronide: <6 hours) complicate accurate exposure assessment, necessitating repeated biomonitoring in future studies (31). Epidemiological data on BPF, BPAF, and other substitutes are scarce despite their rising environmental prevalence (30).

Collectively, according to epidemiological research, exposure to BPs, especially BPA and BPS, is demonstrated to be related to increased risk of NAFLD for adolescents and adults. Though lots of evidence supports the suggestive relationship, unsolved limitations still exist. Most existing studies are cross-sectional, limiting causal inference between BP exposure and NAFLD progression. Thus, longitudinal data tracking BP levels and liver health outcomes over decades are critically needed, especially the risk of liver fibrosis, chronic hepatitis, and even liver cancer. Also, metabolites (e.g., BPA-glucuronide) exhibit short half-lives, complicating exposure assessment. Additionally, co-exposure to other endocrine disruptors (e.g., phthalates) may confound observed associations. While BPA and BPS have been partially characterized in the risk of NAFLD, epidemiological data on BPF, BPAF, and other substitutes remain scarce despite their increasing environmental prevalence.

4 Molecular mechanisms of BPs in NAFLD pathogenesis

In this part, we summarize the predominant roles of BPs in the pathogenesis and development of NAFLD: lipid metabolism disruption, insulin resistance and glucose dysregulation, oxidative stress, and even gut-liver axis perturbation (Figure 3, Table 2).

Figure 3
www.frontiersin.org

Figure 3. Four major mechanisms BPA in the pathogenesis and development of NAFLD: lipid metabolism disruption, insulin resistance and glucose dysregulation, oxidative stress, and gut-liver axis perturbation.

Table 2
www.frontiersin.org

Table 2. Molecular mechanisms of BPA-Induced NAFLD pathogenesis.

4.1 Disruption of lipid metabolism

Lipid metabolism disruption plays a causal role in the development and progression of NAFLD (4143). Initially, Marmugi et al. conducted a 28-day oral exposure study in male CD1 mice with BPA doses ranging from 0, 5, 50, 500, and 5,000 μg/kg/day, revealing low-dose-specific hepatic effects, particularly on lipid synthesis genes. Human daily intake estimates for BPA range from 0.01–0.1 μg/kg/day in the general population, with occupational exposures reaching up to 1–10 μg/kg/day. Notably, the No Observed Adverse Effect Level (NOAEL) for BPA in rodents is 5 mg/kg/day, far exceeding the highest experimental dose (5,000 μg/kg/day = 5 mg/kg/day).

BPA-induced nonmonotonic dose-response patterns in de novo lipogenesis genes (Acc, Fasn, Scd1) and regulatory transcription factors (LXR, SREBP-1c, ChREBP), with more substantial impacts at lower doses. Hepatic cholesterol esters and triglyceride accumulation confirmed enhanced fatty acid biosynthesis (44). Detectable BPA plasma levels have also been observed in Savastano’s research. The robust association between BPA and waist circumference, components of metabolic syndrome, along with inflammatory markers (43) (insulin resistance index, plasma monocyte chemoattractant protein 1, interleukin-6 and tumor necrosis factor-alpha, further substantiates BPA’s involvement in visceral obesity-associated low-grade chronic inflammation (45).

Then, Wang’s group employed an early-life BPA exposure model and further assessed the impacts of BPA exposure on lipid homeostasis (46). In vitro, 3T3-L1 adipocytes exposed to 30 μM BPA for 48 hours showed a 40% increase in lipid accumulation (p<0.01, oil red O quantification). In vivo, male C57BL/6 mice receiving 0.5 mg/kg/day BPA via oral gavage for 8 weeks developed hepatic steatosis, with liver triglycerides elevated by 80% and 30% (low dose and high dose, respectively) compared to controls (p<0.001, p<0.01). RNA-Seq revealed BPA-altered biological processes, including glycosphingolipid biosynthesis, adipocyte lipolysis regulation, PPAR signaling, and fatty acid metabolism in preadipocytes. Notably, adipose tissue and liver showed significant upregulation of SCD1 and APOD downregulation (p<0.01). Mice exposed to 50 or 500 μg/kg/day BPA for 8 weeks developed NAFLD features, confirming dose-dependent metabolic disruption (47). RNA-seq revealed significant expression reversals in lipid-related genes (SCD1, APOD, ANGPT4, PPARβ, LPL, G0S2) between BPA-exposed and recovery groups, particularly SCD1/APOD (p<0.01). BPA exposure markedly reduced APOD protein (p<0.01), which rebounded post-exposure. APOD overexpression suppressed TG accumulation in AML12 cells, demonstrating its critical role in mitigating BPA-induced hepatic metabolic dysfunction significantly in PPAR-related pathway. Not only by PPARβ, inhibition of PPARγ also participant in BPA (1 μg/kg/day)-induced liver lipid accumulation (48). Dysregulation of lipid-regulating factors were detected in hepatic tissue, with pharmacological inhibition of PPARγ ameliorating gestational BPA exposure-induced hepatic steatosis. Furthermore, a male-specific reduction was observed in HNF1b protein levels in offspring. These findings indicate APOD upregulation repairs BPA-mediated damage, highlighting the need to evaluate BPA exposure risks in chronic liver diseases. Furthermore, BPA (1, 2, and 4 μM for 48 h) disrupts lipid metabolism and triggers pyroptosis by upregulating O-GlcNAc transferase (OGT) in HepG2 cell line (49). NLRP3 directly interacts with OGT, with elevated OGT levels enhancing NLRP3 protein stability. Specifically, BPA enhances OGT-mediated O-GlcNAcylation to stabilize NLRP3, accelerating NAFLD progression in vitro models.

More concerningly, the perinatal and peripubertal exposure to BPA should be paid more attention. In 2017, Slitt et al. exposed pregnant mice to 25μg/kg/day BPA from gestation through lactation. Offspring showed persistent fat accumulation via lipogenic gene hypomethylation and elevated hepatic Nrf2 recruitment to Srebp-1c promoters. More epidemiological research should focus on perinatal women, babies, and children (50).

Apart from the mouse model, they also utilized zebrafish to strengthen the assumption (51). Histopathological observation and physiological and biochemical indicators revealed that BPA and TCS (200 mg/L for 90d) exposure led to hepatic fat accumulation in acute and chronic scenarios. RNA-Seq analysis showed that TCS disrupted multiple physiological processes, including drug metabolism, sucrose metabolism, fat metabolism, and bile secretion. The dysregulation of lipid metabolism-related genes indicated that liver steatosis in zebrafish exposed to TCS and BPA resulted from increased fatty acid synthetase, uptake, and suppression of β-oxidation. EDC exposure caused a decrease in global m6A levels and abnormal expression of m6A modulators in larvae.

Apart from protein-coding genes, a classic microRNA—miR-192 also participates in the process of BPA-induced NAFLD (52). Lin et al. showed that after 90-day 50μg/kg/day of BPA by oral gavage, male post-weaning C57BL/6 mice displayed a NAFLD-like phenotype. BPA-induced hepatic steatosis in mouse/HepG2 models correlated with miR-192 downregulation and SREBF1-mediated lipogenic activation. Impaired DROSHA processing reduced miR-192, which directly targeted SREBF1’s 3’UTR. Also, miR-192 overexpression reversed BPA-induced lipid dysregulation by suppressing SREBF1.

Unlike BPA, BPS induced NAFLD through other critical pathways, as Gu’s group reported. BPS activates PPARα-mediated EP300 upregulation, facilitating its nuclear-to-cytoplasmic translocation. This induces Raptor acetylation, triggering mTORC1 activation, and impairs autophagic flux and hepatic lipid metabolism. EP300 knockdown attenuated Raptor acetylation and restored autophagy, identifying EP300 as a key mediator in BPS-induced NAFLD pathogenesis. These findings reveal environmental pollutant-driven metabolic dysregulation mechanisms. Moreover, Qin et al. probed into this fascinating phenomenon through long-term BPS exposure to zebrafish (53). A 120-day BPS exposure induced hepatic steatohepatitis in zebrafish by elevating AST/ALT levels, lipid accumulation (TAG/cholesterol), and fibrosis via PERK-ATF4a UPR pathway activation. While 30-day lipid deposition reversed post-depuration, prolonged exposure upregulated lipogenic genes (srebp1, acc, fasn, and elovl6), triggered ERS-mediated autophagy (atg3, lc3) and inflammation (il1b, tnfα). Findings reveal BPS-driven metabolic disruption in NAFLD pathogenesis through ERS cascades.

Based on Fan et al.’s research, BPF also has the potential for NAFLD risk. Integrating multi-omics methods, they found that BPF exposure (2 mg/kg/day for male and 5 mg/kg/day for female) contributed to changes in hepatic transcriptome, metabolome, and chromatin-accessible regions enriched for binding sites of transcription factors in the bZIP family (54). These alterations were enriched with pathways integral to the ERS and NAFLD, which relied on bZIP family transcription factors. Intriguingly, Drp1 inhibition via Mdivi-1 or gene silencing suppressed mitochondrial fission, alleviating BPF-induced hepatic lipid deposition. Mitochondrial dynamics imbalance mediated this process, as Drp1 blockade reversed fission, restored mitochondrial metabolism, and reduced ROS overproduction - key drivers of lipid accumulation (55). This study identifies Drp1-mediated mitochondrial damage as critical in BPF-triggered NAFLD-like pathology, suggesting mitochondrial-targeted therapies as potential interventions. Liu’s group previously also described a similar phenomenon (56). Interestingly, they deciphered the glycerophospholipid metabolic pathway, which was the most pronounced in BPF-induced disturbance of lipid metabolism. This event revealed a significant mechanism and provided novel intervention strategies for BPF-induced NAFLD-like changes.

4.2 Insulin resistance and glucose dysregulation

Insulin resistance and glucose metabolism dysregulation are pivotal risk factors for NAFLD (57, 58). Interestingly, glucose can easily be disrupted by BPs, which might mediate the process of NAFLD development reported by a population-based, cross-sectional study (8). However, the underlying mechanism is complex and includes novel pathways, though BPs have been verified to contribute to insulin resistance by mimicking the effects of strong binders, such as estrogens (5961). Previously in 2012, Wang et al. conducted a community-based study to dissect the association between BPA exposure’s dose-response association with obesity and insulin resistance in 3390 Shanghai adults (≥40y, mainly middle-aged and elderly Chinese adults) (62). The highest BPA quartile participants showed increased risks of generalized obesity (OR=1.50, 95%CI=1.15-1.97), abdominal obesity (1.28;1.03-1.60), and insulin resistance (1.37;1.06-1.77). Notably, non-overweight individuals (BMI<24kg/m²) demonstrated 94% higher insulin resistance risk (1.94;1.20-3.14) in the top BPA quartile, an association absent in overweight subjects. Long et al.’s study demonstrated that hepatic metabolic analysis revealed BPA-exposed offspring developed hepatic steatosis in both sexes. Male-specific lipid accumulation, along with glucose dysregulation, was observed (48). Sex-dependent HNF1b downregulation occurred in males, suggesting gender-specific metabolic vulnerability.

Similarly, Ji et al. C57BL/6 fed mice with BPA (1-250 μg/kg) for 35 days showed disrupted hepatic fatty acid/glucose metabolism and TCA cycle via metabolomics (high-resolution MS), molecular docking, and enzyme assays. BPA activated nuclear receptor LXR, causing hypoglycemia and impaired liver metabolic functions (63). Thus, disrupted glucose metabolism in BPA-caused NAFLD generally occurs with other abnormal metabolisms.

To investigate the potential relation, Federico et al. analyzed 60 biopsy-confirmed NAFLD patients (with/without T2DM) and 60 healthy controls (64). BPA levels were significantly elevated in NAFLD patients’ plasma/urine vs controls (P<0.0001), particularly in 30 NASH vs 30 simple steatosis cases (P<0.05). A 1-month BPA-free diet reduced plasma BPA (P<0.05) without affecting urinary levels. In vitro, 0.05μM BPA exposure under high/low glucose conditions (H-/L-HepG2) for 48h increased HepG2 proliferation vs controls, with lipid peroxidation showing dose-dependent responses. Their findings suggest environmental BPA exposure may influence NAFLD progression regardless of diabetic status. Due to the sample size limitation, this conclusion still needs further research.

Intriguingly, ovariectomized (OVX) HFD-fed females exhibited obesity, glucose intolerance, insulin resistance, and moderate hepatic steatosis, linked to upregulated hepatic lipogenic (Srebf1, Scd1), β-oxidative (Cpt1a), and ER stress (Hspa5, Hyou1) genes (65). BPA exacerbated hepatic steatosis in the OVX HBPA group, elevating lipid/collagen deposition with reduced Mttp mRNA and upregulated β-oxidation (Acox1, Acadvl), mitochondrial uncoupling (Ucp2), ER stress (Hyou1, Atf6), and liver injury (Tgfb1, Casp8) genes. In OVX CBPA, BPA induced mild steatosis, increasing hepatic lipids and lipogenic/ER stress gene (Srebf1, Scd1, Hspa5, Atf6) expression. BPA amplified HFD-driven liver damage without affecting metabolic disruptions.

Not only in mouse models, Shankar’s study in Wistar rats revealed that high-dose BPA (200mg) significantly reduced hepatic insulin receptor and Akt mRNA/protein expression (66). Despite elevated serum insulin and decreased testosterone in the high-dose group, fasting glucose remained stable. Both BPA doses impaired glucose oxidation and glycogen storage, indicating defective insulin signaling disrupts hepatic glucose metabolism without altering baseline glycemia.

In Jeung’s study, BPA and octylphenol (OP) elevate insulin levels in pancreatic β-cells but impair glucose regulation, suggesting disrupted calcium homeostasis. In STZ-induced diabetes models, both chemicals promote β-cell survival yet fail to normalize blood glucose despite hyperinsulinemia (67). They downregulate calcium influx genes (cytosolic/ER Ca²+ transport) while upregulating Ca²+ efflux pathways, depleting ER calcium stores and triggering ER stress. This ER stress induces insulin resistance, evidenced by reduced GLUT4 and IRS2 expression. Mechanistically, calcium dysregulation by BPA/OP links ER stress to impaired insulin signaling, particularly exacerbating metabolic dysfunction in type 1 diabetes contexts.

Furthermore, the sheep model is also applied to probe into this phenomenon by Muraly et al. (68). Prenatal BPA exposure induced insulin resistance and adipocyte hypertrophy in female offspring. A study on 21-month-old offspring from mothers exposed to 0–5 mg/kg/day BPA revealed non-monotonic dose effects: elevated oxidative stress, hepatic/muscular lipotoxicity, and upregulated aromatase/estrogen receptors in visceral fat. These alterations correlated with tissue-level IR mechanisms, potentially explaining metabolic dysregulation observed in BPA-exposed females.

Geng et al. demonstrated that 5-day 100nM BPA exposure impaired glucose uptake and insulin signaling in HepG2 cells, triggering inflammation, oxidative stress, and JNK/p38 pathway activation. While ERK/NF-κB inhibition showed no effect, blocking JNK/p38 restored metabolic functions (69). Interestingly, curcumin counteracted BPA-induced insulin resistance, but its protective effects were reversed by JNK/p38 activator anisomycin, confirming these pathways’ critical role. However, this therapeutic method warrants further in vivo experiments.

4.3 Oxidative stress and inflammatory cascades

Oxidative stress and inflammation are involved in the BPs-related liver disease development (70). Previously, in 2016, Sahar et al. utilized 30 male Wistar albino rats (BPA in 50 mg/kg body weight/day, 8 weeks). They concluded that elevated serum hepatic enzymes, hepatic hydroxyproline, and portal collagen deposition evidenced BPA-induced liver fibrosis. It triggered inflammation (↑IL-1β, ↓IL-10), oxidative stress (↑MDA, ↓GSH, suppressed CAT), and apoptosis (↑caspase-3, ↓BCL2+ hepatocytes). BPA upregulated extracellular matrix turnover gene MMP-9 while downregulating its inhibitor TIMP-2, exacerbating fibrotic progression (71). However, they have not dissected the association between BPA exposure and NAFLD, probably due to the inadequate exposure time. Thus, the role of BPA in oxidative stress during fatty liver disease needs further in vivo experiments.

Then, Stefania et al. explored BPA and silybin co-effects on H-HepG2 cells under high glucose (72). BPA induced oxidative stress and oxidized steroid hormones into estrogenic/genotoxic metabolites. Silybin counteracted BPA effects by reducing glucose uptake/lipid peroxidation, activating vitamin D3 synthesis, and preventing steroid oxidation. Western blot revealed silybin-modulated p-ERK/ERK and Caspase-3 expression, while Mass spectrometry confirmed altered lipid/steroid profiles. Findings highlight silybin’s protective role against BPA-induced metabolic disruptions in hepatic cells, which can be a potential therapeutic method in the future.

BPA exacerbated HFD-induced hepatic metabolic dysregulation and mitochondrial dysfunction through oxidative stress elevation and antioxidant depletion while synergistically activating TLR4/NF-κB/NLRP3 axis to amplify inflammatory cytokine production and fibrogenesis (73). Accidentally, NLRP3 inflammasome can be activated in the liver of lactating dams after low-dose BPF exposure (74). BPF administration in lactating dams significantly upregulated iNOS and HO-1d expression, triggered activation of NLRP3 inflammasome components (NLRP3, PyCARD, CASP1), and enhanced secretion of proinflammatory cytokines, including IL-1β, IL-18, IFN-γ, and TNF-α. Nowadays, emerging immunological mechanisms now recognized as pivotal contributors to disease pathogenesis encompass functional impairments in innate immunity, adaptive immunity dysregulation, Toll-like receptor (TLR) signaling anomalies, and gut-liver axis homeostasis disruption. The NLRP3 inflammasome, an intracellular multiprotein complex, orchestrates caspase-1-dependent maturation of interleukin-1β (IL-1β) while propagating sterile metabolic inflammation through coordinated pathological cascades (75, 76).

Female sheep were also applied to dissect the underlying mechanism in BPA-induced NAFLD. Interestingly, accordingly to liver-specific pathways included oxidative stress/lipid synthesis. Non coding RNA (ncRNA) alterations occurred in the liver (77 lncRNAs, 14 miRNAs, 127 snoRNAs, 55 snRNAs), correlating with LCORL/MED17/ZNF41 mRNAs. Discriminant analysis identified tissue-specific gene signatures (liver: PECAM/RDH11/ABCA6/miRNAs), linking BPA to mitochondrial dysfunction, oxidative stress, and metabolic dysregulation (77). Accordingly, liver-spleen axis has been verified to involve in NAFLD development. Well-known reasons in explaining these mechanisms include the spleen involvement in immune regulation (78). Furthermore, the spleen-liver axis supports obesity-induced systemic and fatty liver inflammation via MDSC and NKT cell enrichment (79). Apart from inflammation in liver itself, BPA-induced oxidative stress in extraphepatic tissues is potentially important during NAFLD development. According to Shaibi’s study, both sexes of BPA-exposed mice exhibited elevated peripheral monocytes/lymphocytes (80). Adult spleens showed histopathological toxicity: activated germinal centers and apoptotic cells in white pulp, marked eosinophil/lymphocyte infiltration in red pulp. DNA fragmentation via electrophoresis confirmed apoptosis, while elevated malondialdehyde levels indicated oxidative lipid damage in splenic tissues versus controls. Similarly, Cai’s group demonstrated that BPS exposure triggered splenomegaly, pro-inflammatory polarization, and structural remodeling, while provoking lipidomic disruptions within white pulp immune niches, which may offer novel mechanistic insights into bisphenol-mediated multiorgan toxicity (81).

Thus, oxidative stress and inflammation contribute to the pathogenesis of BPA-caused NAFLD and related phenotypes.

4.4 Gut-liver axis perturbation

Microbial metabolites and the “gut-liver” axis are pivotal in liver diseases, including NAFLD (82). Exposomics is perceived to probe into the field of liver pathogenesis. Polyaromatic hydrocarbons, especially the BPA and its alternatives, commonly disturb the liver homeostasis via the “gut-liver” axis (83, 84). Male CD-1 mice fed 50 μg/kg/day BPA for 24 weeks developed hepatic steatosis with reduced gut microbiota diversity. BPA increased Proteobacteria while decreasing Akkermansia abundance, impairing intestinal barrier function (ZO-1/occludin decreased), and elevating endotoxin. This activated hepatic TLR4/NF-κB pathway, upregulating IL-1β, IL-18, TNF-α, and IL-6. Findings suggest BPA-induced steatosis links to gut dysbiosis-mediated endotoxemia and TLR4/NF-κB-driven inflammation (14). This initial conclusion was consistent with Feng’s study, reporting that the relative abundance of Proteobacteria and Firmicutes/Bacteroidetes ratio was increased in BPA-fed mice, and this alteration was reversed by curcumin treatment (13).

Similarly, in Wang et al.’s research, both male and female rats were exposed to BPA (300 mg/kg) by oral gavage for 60 consecutive days (85). The male-BPA group showed significant ALT, TG, TC, and LDL alterations; females exhibited GLB, IBIL, ALP, HDL, and Cr changes. BPA reduced gut microbiota diversity and decreased both sexes’ colon SCFAs (caproic, isobutyric, and isovaleric acids). Serum metabolomics revealed BPA-modulated bile acids, amino acids, hormones, and lipids. In males, immune markers IL-6, IL-23, and TGF-β increased, indicating systemic immune disruption. These findings demonstrate BPA’s sex-specific metabolic interference and gut-microbiota-mediated toxicity mechanisms. However, although the histopathological analysis partially presented hepatic impairment, these changes could not meet the criteria of liver steatosis diagnosis. Liao et al. performed a systems biology analysis evaluating liver transcriptomes, gut microbiota, and metabolic phenotypes in mouse offspring exposed to 5 μg/kg/day BPA during gestation (86). Prenatal BPA disrupted hepatic oxidative phosphorylation, PPAR signaling, and fatty acid metabolism genes while inducing sex/age-dependent microbial shifts. Key bacteria (S24-7, Lachnospiraceae) correlated with altered metabolic genes (Acadl, Dgat1) and network drivers (Malat1, Apoa2). This multi-omics integration reveals microbiota-liver interactions potentially mediating cardiometabolic risks from developmental BPA exposure.

Emerging as a prevalent substitute for BPA, BPS exposure induces gut microbiota dysbiosis while triggering obesity, hepatic steatosis, intestinal pathologies, and metabolic dysregulation (87). Notably, robust associations have been established between specific microbial compositional shifts and clinical health parameters in exposed hosts. This evidence positions particular gut microbial signatures as promising BPS exposure risk assessment diagnostic biomarkers (88).

5 Sex-specific and developmental susceptibility in mature

5.1 Sexual dimorphism in bisphenol-induced metabolic dysregulation

Herein, we summarized three significant explanations for the sexual disparity of BPs-induced hepatic metabolic dysregulation: hormonal receptor modulation, sex hormone interactions, and epigenetic and developmental drivers (Figure 4).

Figure 4
www.frontiersin.org

Figure 4. Significant reasons for the sexual disparity of BPA-induced hepatic metabolic dysregulation.

BPs exhibit sex-specific hepatotoxicity through divergent interactions with nuclear hormone receptors. Importantly, effects of EDCs like bisphenols may rely on the hormonal status of individuals. Precisely in females, the estrogeno-mimetic effect of EDCs such as BPA may trigger opposite effects as shown in a previous study (89).

In female rodents, BPA preferentially binds to estrogen receptor-α (ERα), activating hepatic lipogenic pathways (e.g., SREBP-1c, FASN) and exacerbating lipid accumulation by up to 30% compared to males (9). Conversely, male susceptibility centers on androgen receptor (AR) disruption: BPA competitively inhibits dihydrotestosterone (DHT) binding, impairing AR-mediated suppression of PPARγ and CD36 expression, thereby driving ectopic lipid deposition and insulin resistance (9092). Human studies corroborate this dimorphism—urinary BPA levels correlate more strongly with elevated ALT/AST and NAFLD prevalence than females.

BPs dysregulate steroidogenesis, amplifying metabolic risks. Prenatal BPA exposure reduces fetal testosterone synthesis in males by suppressing Cyp17a1 and HSD3β activity, leading to persistent deficits in mitochondrial β-oxidation (CPT1A↓) and hepatic steatosis in adulthood (93). BPA elevates estradiol (E2) via aromatase induction in females, activating ERα-driven SCD1 expression and triglyceride synthesis (94). Paradoxically, postmenopausal females exhibit exacerbated BP sensitivity due to the loss of endogenous E2’s protective effects on lipid homeostasis.

Sex-specific DNA methylation patterns in metabolic genes emerge after developmental BP exposures (95). Male offspring show hypermethylation of PGC-1α promoters, suppressing mitochondrial biogenesis, while females retain demethylated ERβ loci that mitigate lipid peroxidation (96). These epigenetic imprints may explain why childhood BP exposure accelerates NAFLD onset by 5–10 years in males versus females.

5.2 Developmental programming of NAFLD

Developmental exposure to BPs during critical windows (gestation, lactation, puberty) induces persistent metabolic reprogramming, predisposing offspring to NAFLD. Prenatal BPA exposure (25 μg/kg/day) in mice triggers hepatic lipogenic gene hypomethylation enhancing lipid synthesis via Nrf2-mediated transcriptional activation (50). Sex-specific susceptibility emerges, with male offspring exhibiting amplified steatosis due to estrogen receptor antagonism and impaired PPARα signaling. Epigenetic modifications in lipid oxidation genes (CPT1A, ACOX1) and gut microbiota dysbiosis persist into adulthood, creating a “second hit” for NAFLD progression (93). Transgenerational effects occur via germline epigenetic inheritance, with F2 progeny showing elevated liver triglycerides despite no direct exposure. These findings underscore developmental BP exposure as a latent driver of metabolic liver disease across lifespans.

5.3 Mechanistic insights from model organisms

Sex-divergent responses to bisphenols are rooted in hormone-signaling crosstalk. In male C57BL/6 mice, BPA activates hepatic ERα/PPARγ pathways, amplifying lipogenesis, while females exhibit compensatory estrogen-mediated suppression of SREBP-1c via GPER1. Developmental exposure models reveal transgenerational impacts: prenatal BPA (50 μg/kg/day) in CD1 mice induces hypomethylation of Lxrα promoters in F1 males, priming lipid accumulation through enhanced ChREBP recruitment (44). Zebrafish studies demonstrate AR-mediated susceptibility, where BPS upregulates PPARβ in males but not females, correlating with sex-specific steatosis. Organoid models further highlight sex-dimorphic NLRP3 activation, with testosterone potentiating BPA-induced inflammasome priming in hepatocytes (11). These findings underscore ER/AR interplay and epigenetic reprogramming as key drivers of sex- and developmental-stage-specific vulnerabilities.

6 Interactions with other environmental factors

6.1 Co-Exposure with chemical pollutants

Nowadays, EDCs are perceived as NAFLD promoters and Di-(2-Ethylhexyl) phthalate (DEHP) and BPA present significant environmental endocrine-disrupting chemical properties (97, 98). DEHP exposure is also associated with the NAFLD risk, based on NHANES database (99). However, the co-exposure of BPA and DEHP has not yet been thoroughly explored. Mechanistically, in Zhang et al.’s recent research, 36 perinatal rats were divided into DEHP (600 mg/kg/d), BPA (80 mg/kg/d), combination, and control groups (100). Screening identified 11 hepatic-damage-related chemical targets. Molecular docking showed strong interactions between 8 metabolic components and PI3K/AKT/FOXO1 pathway targets. Combined exposure induced hepatic steatosis and disrupted glucose/lipid homeostasis via PI3K/AKT/FOXO1-mediated liver dysfunction and insulin resistance.

When tested at non-cytotoxic levels, these substances and BPA-disrupted key regulatory genes govern lipid homeostasis. Specifically, cadmium, PFOA, DDE, and DEHP markedly upregulated DGAT1 (crucial for triglyceride synthesis), while butylparaben enhanced fatty acid transporter FAT/CD36 expression. BPA conversely suppressed CPT1A, a pivotal gene in fatty acid oxidation. Notably, PFOS, BPS, and dibutyl phthalate showed negligible impacts on lipid droplet formation or lipid metabolism-associated genes. Across tested EDCs, lipid accumulation demonstrated positive correlations with SREBF1, DGAT1, and CPT1A expression profiles. These results strengthen evidence linking EDC exposure to NAFLD pathogenesis while demonstrating the value of in vitro approaches for identifying environmental contaminants with steatogenic potential and metabolic disruption capabilities.

6.2 Environmental modulators of BP toxicity

Environmental co-exposures significantly amplify bisphenol-induced hepatotoxicity through synergistic or additive mechanisms. Phthalates, commonly coexisting with BPs in plastics, exacerbate hepatic lipid accumulation by competitively inhibiting PPAR-α-mediated fatty acid oxidation while amplifying ER stress via the PERK-CHOP pathway. Heavy metals like cadmium and lead act as co-modulators by depleting glutathione reserves, compounding BP-induced oxidative DNA damage in hepatocytes. Particulate matter (especially PM2.5) enhances BP bioaccumulation in liver tissue through AhR-mediated CYP450 inhibition, while microplastics serve as BP transport vectors, increasing intestinal absorption and hepatic bioavailability by 40-60% (83, 101, 102). Dietary factors critically modulate toxicity thresholds: high-fat diets upregulate hepatic CD36 expression, facilitating BP-triggered lipid uptake, whereas polyphenol-rich diets mitigate steatosis via Nrf2 activation. Crucially, circadian disruption from light pollution dysregulates hepatic clock genes (e.g., Bmal1), potentiating BPs-induced metabolic dysfunction. These interactions underscore the necessity for cumulative risk assessments in environmental health policies. Regarding dietary factors, several researches showed that environmental pollutants and high-fat diets affect common metabolic pathways, e.g., modifications in lipid homeostasis (103).

6.3 Lifestyle and dietary co-drivers

Concerningly, compared to the low-BMI population, those fatty individuals more easily suffer from NAFLD under BPA exposure. Ribeiro’s group demonstrated that BPA exposure in ovariectomized mice on high-fat diet exacerbated hepatic steatosis, increasing collagen deposition and altering lipid metabolism genes: downregulating Mttp while upregulating β-oxidation (Acox1, Acadvl), mitochondrial uncoupling (Ucp2), ER stress (Hyou1, Atf6), and liver injury markers (Tgfb1, Casp8). BPA caused mild steatosis in normal-diet mice through upregulated lipogenesis (Srebf1, Scd1) and ER stress genes (104). BPA exacerbated HFD-mediated dysregulation of critical regulators in glucose and lipid metabolism, promoted hepatic triglyceride deposition, and aggravated mitochondrial dysfunction through elevating oxidative stress while diminishing cellular antioxidant capacity (73). Perinatal exposure to BPA/experimental diets alters offspring metabolic health (not NAFLD), with maternal phenotype changes driving health trajectories, underscoring maternal diet’s role in environmental exposure assessments (105).

Similar to BPA, according to Xie’s study, BPS exposure (50/500 μg/kg/day) exacerbated HFD-induced NAFLD in mice, increasing liver/body weight ratio, serum ALT/AST levels, and hepatic lipid accumulation. It dysregulated fatty acid metabolism genes (Cd36, Pparγ, Scd-1, Fasn, Pparα) and pro-inflammatory cytokines (TNFα, IL-1β, IL-6) (106). BPS triggered hepatic ferroptosis via altered GPX4, xCT, FTH, and ACSL4 expression, accompanied by ROS overproduction, mitochondrial dysfunction, lipid peroxidation, and GSH depletion - all rescued by ferrostatin-1. Mechanistically, BPS upregulated HMGCS2 in hepatocytes, and HMGCS2 knockdown reversed ferroptosis markers, demonstrating that HMGCS2-mediated ferroptosis drives BPS-aggravated NAFLD.

Lv’s study assessed BPF effects (100 μg/kg) on glucose metabolism in mice fed normal (ND) or HFD. BPF improved glucose metabolism in HFD mice but not ND mice, enhancing insulin signaling in skeletal muscle and elevating liver metabolites linked to carbohydrate digestion and TCA cycle (107). Sustained environmentally relevant BPF exposure enhanced insulin sensitivity and glucose regulation in HFD mice. Thus, BPF also deteriorates the glucose metabolism of HFD mice, potentially contributing to the development of NAFLD.

Despite the limitation of mechanistic evidence in humans, substantially decreasing the fatty diet intake can be a promising method to avoid BPs-induced NAFLD.

6.4 Mitigating combined exposures

Addressing synergistic hepatotoxicity from bisphenol co-exposure with phthalates or dietary stressors requires integrated strategies. Regulatory frameworks should adopt cumulative risk assessment models for chemical mixtures, replacing single-pollutant paradigms. Emerging technologies like nanomaterial-based biosensors enable real-time detection of multiple contaminants in food and household products, empowering consumer choices. Public health initiatives must promote BP-free alternatives (e.g., plant-based food coatings, ceramic containers) while discouraging concurrent high-fat diets through nutritional education (108). Pharmacological interventions targeting shared pathways—such as NLRP3 inflammasome inhibitors or PPAR-γ antagonists—may counteract synergistic metabolic disruption (49, 109). Crucially, microbiome-modulating prebiotics (e.g., resistant starch, polyphenols) can mitigate gut-liver axis perturbations amplified by combined exposures (13). Policy reforms mandating eco-design principles for plastics and stricter labeling of “BPA-free” substitutes are essential to break exposure cycles.

7 Therapeutic and preventive strategies

7.1 Lifestyle intervention

Key lifestyle strategies combine BP exposure reduction, dietary modifications, and developmental-stage-specific protection: (1) Exposure minimization: avoid canned foods, plastic-packaged items, and thermal receipts; prioritize glass/ceramic containers (1, 108). (2) Antioxidant-rich diet: consume polyphenol-rich whole foods (fruits/vegetables) to counteract BP-induced oxidative stress (5). (3) Gut-liver axis support: increase resistant starch (whole grains) and prebiotic fiber to mitigate BP-disrupted gut microbiota (13). (4) Weight management: achieve 5–10% weight loss via calorie-restricted diets and combined aerobic/resistance exercise to reverse hepatic steatosis (3, 107). (5) Implement strict BP avoidance in children/adolescents through school nutrition programs and BP-free lunch packaging, targeting vulnerable metabolic programming windows (110).

7.2 Pharmacological approaches

Several researchers recently reported that green tea extract intake improves hepatic steatosis and reduces the development of NAFLD (111113). Moreover, the detrimental effects triggered by BPA in the pathogenesis of NAFLD can be ameliorated by green tea, Vitamin E, and epigallocatechin gallate (EGCG), the major catechin present in green tea (114). Underlyingly, their antioxidant and anti-inflammatory activity can influence this protective role, regulating lipid metabolism and insulin signaling pathway improvement. Furthermore, Zhang’s results demonstrated that EGCG administration effectively decreased mice’s body weight and liver-to-body weight ratio while simultaneously reducing serum triglyceride and total cholesterol concentrations (115). These metabolic improvements were mechanistically linked to transcriptional regulation, evidenced by suppressed mRNA expression of fatty acid synthesis-related Elov16 and cholesterol biosynthesis-associated CYP4A14, coupled with enhanced fatty acid oxidation-related Lss and cholesterol metabolism-regulating Cyp7a1 genes. Thus, green tea, especially its catechin-EGCG, has a promising application for preventing and treating BPs-induced liver metabolism disturbance.

It has been verified that BPA-induced lipid metabolism dysfunction and pyroptosis are driven by OGT upregulation, which critically stabilizes NLRP3 through direct interaction. Elevated OGT enhances NLRP3 protein stability via enhanced O-GlcNAcylation modification. BPA promotes OGT-mediated O-GlcNAcylation to stabilize NLRP3, accelerating NAFLD progression in vitro (49). Thus, targeting the NLRP3-OGT axis may counteract BPA-induced NAFLD pathogenesis.

Interestingly, Xu’s group first reported that oridonin alleviates bisphenol A-induced hepatotoxicity via modulating oxidative stress and metabolic pathways. In rats exposed to BPA (500 mg/kg), oridonin pretreatment (10 mg/kg) significantly reduced serum AST/ALT levels, attenuated hepatic apoptosis, and improved histopathology. UPLC-MS/MS metabolomics identified 28 differential metabolites, indicating oridonin restored BPA-disrupted purine metabolism and phenylalanine/tryptophan biosynthesis (116). Mechanistically, oridonin inhibited xanthine oxidase (XOD) activity and decreased ROS and uric acid levels while increasing hypoxanthine/xanthine content. These findings demonstrate oridonin’s hepatoprotective effects through dual regulation of oxidative stress and metabolic reprogramming.

Resveratrol, a natural polyphenolic compound belonging to the stilbene family, emerges as a promising therapeutic candidate for NAFLD prevention and treatment owing to its combined anti-inflammatory, antioxidant properties, and calorie-restriction-mimicking biological effects (117, 118). Based on Liao’s research, intriguingly, resveratrol butyrate ester (RBE) alleviates BPA-induced liver damage in male offspring rats by modulating gut microbiota (enhancing S24-7/Adlercreutzia abundance), increasing fecal short-chain fatty acids, and activating hepatic Nrf2 pathway to boost antioxidant enzymes (HO-1/SOD/CAT). RBE reinforces intestinal barrier integrity and reduces liver inflammation through gut-liver axis interactions, demonstrating its protective mechanism against developmental metabolic disruption (119).

Gut microbiota metabolites play a pivotal role in NAFLD pathogenesis and therapeutic implications; therefore, probiotic modulation emerges as a novel method for prevention and therapeutic methods (120, 121). Co-administering BPA and SLAB51 (109 CFU/g of body weight; P) restored gut integrity, enriched beneficial microbes, and reduced pathogens. In males, BPA-induced hepatic steatosis/glycogen loss was partially reversed by P, correlating with elevated anserine (neuroprotective) and reduced glutamine in the liver. Females showed no steatosis but heightened energy demand via glycogen depletion; while P reduced hepatic retinoic acid, potentially enhancing BPA detoxification.

7.3 Policy and public health measures

Importantly, enforce BPA bans in food/beverage packaging and children’s products, extending EU-style REACH regulations globally. Secondly, research funding for plant-based resins (lignin polymers) and non-estrogenic alternatives should be accelerated through public-private partnerships. National biomonitoring programs should also be established to track urinary BP metabolites and liver enzymes (ALT/AST) in high-risk groups (especially children and factory workers).

8 Future directions

8.1 Mechanistic research gaps

In Table 3, we summarized critical gaps and promising methodologies for BP-Induced NAFLD. A critical knowledge gap lies in understanding the hepatotoxic effects of BP metabolites (e.g., BPA-glucuronide) and their role in transgenerational NAFLD pathogenesis. While parental BPA exposure is linked to offspring metabolic dysfunction, the epigenetic mechanisms—such as DNA methylation, histone modifications, or non-coding RNA regulation—mediating this inheritance remain poorly characterized (122124). Future studies should delineate how BP metabolites interact with nuclear receptors (e.g., ERα/β) to reprogram hepatic lipid metabolism and oxidative stress pathways across generations.

Table 3
www.frontiersin.org

Table 3. Critical research gaps and proposed methodologies for Bisphenol-Induced NAFLD.

Current evidence lacks large-scale, long-term human data correlating BP exposure levels with NAFLD progression. Establishing multinational cohorts (e.g., integrating the NHANES database with Biobanks) is imperative to track temporal changes in gut microbiota composition, circulating BP metabolites, and liver injury biomarkers (e.g., ALT, miRNAs). Such cohorts should stratify populations by sex, age, and metabolic status to identify vulnerable subgroups and dose-response thresholds.

Most research focuses on BPA, yet substitutes like BPS and BPF exhibit similar endocrine-disrupting properties. Their individual and combined hepatotoxicity—particularly in co-exposure with microplastics (e.g., polystyrene)—requires urgent investigation. Mechanistic studies using liver organoids could clarify synergistic disruptions of lipid metabolism genes (e.g., HNF4A, CD36) and epigenetic regulators.

Sex disparities in BPA-induced hepatic steatosis (e.g., female resilience to lipid accumulation) suggest hormonal or genetic modifiers. Comparative analyses of retinoic acid signaling, hypothalamic-pituitary-liver axis interactions, and X chromosome-linked genes may reveal novel therapeutic targets.

8.2 Innovative methodologies

Lipid droplets (LDs) critically influence physiological processes, necessitating dynamic visualization in living cells (125). This study utilized QSAR theory to design organelle-targeting carbon dots (CDs), employing Log P values to predict cellular uptake and subcellular localization. Hydrophilic p-phenylenediamine-derived CDs were transformed into lipophilic PA CDs with inherent LD-targeting capability 110 by modifying precursor lipophilicity. These PA CDs successfully tracked LD dynamics and visualized bisphenol A-induced fatty liver disease progression in cellular models. The QSAR-driven strategy demonstrates the potential for developing diverse organelle-specific CDs, offering a robust design framework for subcellular imaging probes.

Human pluripotent stem cell-derived liver organoids (LOs) recapitulate sex- and age-specific responses to BP exposures (126). Co-culturing LOs with gut microbiota-derived metabolites (e.g., short-chain fatty acids) reveals cross-tissue interactions. At the same time, single-cell RNA sequencing uncovers heterogeneous cell subpopulations vulnerable to PS-BPA synergism (127). Machine learning algorithms can integrate LO-based epigenomic (HNF4A methylation), proteomic (CYP2E1/ERα), and lipidomic data to map adverse outcome pathways. Also, CRISPR-edited hepatic organoids can be considered as a drug screening platform for NAFLD (128). Human fetal hepatocyte organoids model NAFLD steatosis under three triggers: fatty acid overload, PNPLA3 I148M mutation, and APOB/MTTP mutations. Drug screening identified steatosis-resolving compounds targeting de novo lipogenesis repression (129). Delilah et al. developed FatTracer, a CRISPR platform using APOB−/−/MTTP−/− organoids, screening 35 lipid-related genes. FADS2 emerged as critical enhanced expression increases polyunsaturated fatty acids, suppressing lipogenesis. This system enables mechanistic exploration and therapeutic target discovery for hepatic steatosis.

Deep learning models trained on UPLC-MS/MS metabolomics and LD imaging data can predict structure-hepatotoxicity relationships for emerging BP substitutes. These models, validated through high-content screening in LOs, will accelerate the prioritization of safer alternatives like lignin-based polymers.

8.3 Translational priorities

Urgent efforts are needed to validate non-invasive biomarkers for early NAFLD detection in BP-exposed populations. Circulating miRNAs (e.g., miR-122, miR-34a) and epigenetic markers (e.g., HNF4A methylation) show promise for reflecting BPA-induced hepatic steatosis and ERα-mediated lipid dysregulation (127, 130). Concurrently, organoid-derived exosomes could serve as personalized platforms to identify susceptibility biomarkers linked to PS-BPA synergism. Integrating these biomarkers with multi-omics profiles (metabolomics, metagenomics) may enable risk stratification for precision interventions, such as dietary modifications or targeted detoxification therapies.

Harmonizing BP exposure assessments across regions requires establishing international consortia to standardize analytical methods (e.g., quantifying BPA-glucuronide in urine) and NAFLD diagnostic criteria (imaging vs. histology). Leveraging existing cohorts like NHANES and UK Biobank, combined with microplastic exposure mapping, could reveal geographic hotspots of coexposure risks. Priority should be given to vulnerable groups—children, pregnant women, and metabolically compromised individuals—through longitudinal monitoring of gut microbiota shifts and liver enzymes.

While plant-based resins (e.g., lignin derivatives) and non-estrogenic alternatives are emerging, their long-term hepatotoxicity and endocrine-disrupting potential remain unverified. Accelerated safety assessments using liver organoid models and QSAR-driven carbon dot probes (e.g., PA CDs) are critical to avoid regrettable substitutions. Regulatory agencies must adopt adverse outcome pathways (AOPs) for BP analogs, prioritizing compounds with minimal lipid droplet accumulation and CYP2E1 disruption in preclinical screens.

Targeted interventions—probiotics to restore BPA-disrupted gut flora or epigenetic modulators (e.g., curcumin) to counteract DNA methylation changes—should be tested in high-risk communities. Public health policies must integrate BP exposure reduction (e.g., food packaging reforms) with NAFLD prevention programs, emphasizing sex-specific vulnerabilities identified in mechanistic studies.

Author contributions

C-LL: Conceptualization, Formal analysis, Funding acquisition, Investigation, Project administration, Writing – original draft, Writing – review & editing. Z-YY: Conceptualization, Investigation, Project administration, Writing – original draft. Y-FZ: Data curation, Project administration, Writing – original draft. X-TC: Conceptualization, Data curation, Formal analysis, Writing – original draft. AS: Conceptualization, Data curation, Writing – original draft. J-YC: Formal analysis, Funding acquisition, Writing – review & editing. Z-SW: Formal analysis, Funding acquisition, Resources, Writing – original draft.

Funding

The author(s) declare that no financial support was received for the research and/or publication of this article.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Abbreviations

AR, androgen receptor; AOPs, adverse outcome pathways; BPA, bisphenol A; BPAF, bisphenol AF; BPAP, bisphenol AP; BPs, Bisphenols; CDs, carbon dots; DEHP, Di-(2-Ethylhexyl) phthalate; DHT, dihydrotestosterone; E2, estradiol; EDCs, endocrine-disrupting chemicals; EGCG, epigallocatechin gallate; ER, estrogen receptor; ERα, estrogen receptor-α; HIS, hepatic steatosis index; KoNEHS, Korean National Environmental Health Survey; LDs, Lipid droplets; Los, liver organoids; NAFLD, non-alcoholic fatty liver disease; NASH, non-alcoholic steatohepatitis; ncRNA, non coding RNA; NHANES, National Health and Nutrition Examination Survey; OGT, O-GlcNAc transferase; OR, Odds Ratio; OVX, ovariectomized; RBE, resveratrol butyrate ester; TMAO, trimethylamine-N-oxide; XOD, xanthine oxidase.

References

1. Younossi ZM. Non-alcoholic fatty liver disease - A global public health perspective. J hepatology. (2019) 70:531–44. doi: 10.1016/j.jhep.2018.10.033

PubMed Abstract | Crossref Full Text | Google Scholar

2. Powell EE, Wong VW, and Rinella M. Non-alcoholic fatty liver disease. Lancet (London England). (2021) 397:2212–24. doi: 10.1016/S0140-6736(20)32511-3

PubMed Abstract | Crossref Full Text | Google Scholar

3. Yip TC, Vilar-Gomez E, Petta S, Yilmaz Y, Wong GL, Adams LA, et al. Geographical similarity and differences in the burden and genetic predisposition of NAFLD. Hepatology (Baltimore Md.). (2023) 77:1404–27.

PubMed Abstract | Google Scholar

4. Heindel JJ, Blumberg B, Cave M, Machtinger R, Mantovani A, Mendez MA, et al. Metabolism disrupting chemicals and metabolic disorders. Reprod Toxicol (Elmsford N.Y.). (2017) 68:3–33.

Google Scholar

5. Foulds CE, Treviño LS, York B, and Walker CL. Endocrine-disrupting chemicals and fatty liver disease. Nat Rev Endocrinol. (2017) 13:445–57. doi: 10.1038/nrendo.2017.42

PubMed Abstract | Crossref Full Text | Google Scholar

6. Cano R, Pérez JL, Dávila LA, Ortega Á, Gómez Y, Valero-Cedeño NJ, et al. Role of endocrine-disrupting chemicals in the pathogenesis of non-alcoholic fatty liver disease: A comprehensive review. Int J Mol Sci 22. (2021). doi: 10.3390/ijms22094807

PubMed Abstract | Crossref Full Text | Google Scholar

7. Peters AE, Ford EA, Roman SD, Bromfield EG, Nixon B, Pringle KG, et al. Impact of Bisphenol A and its alternatives on oocyte health: a scoping review. Hum Reprod Update. (2024) 30:653–91. doi: 10.1093/humupd/dmae025

PubMed Abstract | Crossref Full Text | Google Scholar

8. Peng J, Du LL, and Ma QL. Serum glycolipids mediate the relationship of urinary bisphenols with NAFLD: analysis of a population-based, cross-sectional study. Environ health: A Global Access Sci Source. (2023) 21:124. doi: 10.1186/s12940-022-00945-w

PubMed Abstract | Crossref Full Text | Google Scholar

9. He W, Gao Z, Liu S, Tan L, Wu Y, Liu J, et al. G protein-coupled estrogen receptor activation by bisphenol-A disrupts lipid metabolism and induces ferroptosis in the liver. Environ pollution (Barking Essex. (2023) 1987) 334:122211.

PubMed Abstract | Google Scholar

10. Ilagan Y, Mamillapalli R, Goetz TG, Kayani J, and Taylor HS. Bisphenol-A exposure in utero programs a sexually dimorphic estrogenic state of hepatic metabolic gene expression. Reprod Toxicol (Elmsford N.Y.). (2017) 71:84–94.

PubMed Abstract | Google Scholar

11. Das S, Mukherjee U, Biswas S, Banerjee S, Karmakar S, and Maitra S. Unravelling bisphenol A-induced hepatotoxicity: Insights into oxidative stress, inflammation, and energy dysregulation. Environ pollution (Barking Essex. (2024) 1987) 362:124922.

PubMed Abstract | Google Scholar

12. Gao X, Liu S, Ding C, Miao Y, Gao Z, Li M, et al. Comparative effects of genistein and bisphenol A on non-alcoholic fatty liver disease in laying hens. Environ pollution (Barking Essex: 1987). (2021) 288:117795. doi: 10.1016/j.envpol.2021.117795

PubMed Abstract | Crossref Full Text | Google Scholar

13. Hong T, Jiang X, Zou J, Yang J, Zhang H, Mai H, et al. Hepatoprotective effect of curcumin against bisphenol A-induced hepatic steatosis via modulating gut microbiota dysbiosis and related gut-liver axis activation in CD-1 mice. J Nutr Biochem. (2022) 109:109103. doi: 10.1016/j.jnutbio.2022.109103

PubMed Abstract | Crossref Full Text | Google Scholar

14. Feng D, Zhang H, Jiang X, Zou J, Li Q, Mai H, et al. Bisphenol A exposure induces gut microbiota dysbiosis and consequent activation of gut-liver axis leading to hepatic steatosis in CD-1 mice. Environ pollution (Barking Essex: 1987). (2020) 265:114880. doi: 10.1016/j.envpol.2020.114880

PubMed Abstract | Crossref Full Text | Google Scholar

15. Wang J, Chan FKS, Johnson MF, Chan HK, Cui Y, Chen J, et al. Material cycles, environmental emissions, and ecological risks of bisphenol A (BPA) in China and implications for sustainable plastic management. Environ Sci Technol. (2025) 59:1631–46. doi: 10.1021/acs.est.4c09876

PubMed Abstract | Crossref Full Text | Google Scholar

16. Banaderakhshan R, Kemp P, Breul L, Steinbichl P, Hartmann C, and Fürhacker M. Bisphenol A and its alternatives in Austrian thermal paper receipts, and the migration from reusable plastic drinking bottles into water and artificial saliva using UHPLC-MS/MS. Chemosphere. (2022) 286:131842. doi: 10.1016/j.chemosphere.2021.131842

PubMed Abstract | Crossref Full Text | Google Scholar

17. Russo G, Barbato F, and Grumetto L. Monitoring of bisphenol A and bisphenol S in thermal paper receipts from the Italian market and estimated transdermal human intake: A pilot study. Sci total Environ 599-600. (2017), 68–75. doi: 10.1016/j.scitotenv.2017.04.192

PubMed Abstract | Crossref Full Text | Google Scholar

18. Franko N, Kodila A, and Sollner Dolenc M. Adverse outcomes of the newly emerging bisphenol A substitutes. Chemosphere. (2024) 364:143147. doi: 10.1016/j.chemosphere.2024.143147

PubMed Abstract | Crossref Full Text | Google Scholar

19. Lorber M, Schecter A, Paepke O, Shropshire W, Christensen K, and Birnbaum L. Exposure assessment of adult intake of bisphenol A (BPA) with emphasis on canned food dietary exposures. Environ Int. (2015) 77:55–62. doi: 10.1016/j.envint.2015.01.008

PubMed Abstract | Crossref Full Text | Google Scholar

20. Hu M, Zhang Z, Zhang Y, Zhan M, Qu W, He G, et al. Development of human dermal PBPK models for the bisphenols BPA, BPS, BPF, and BPAF with parallel-layered skin compartment: Basing on dermal administration studies in humans. Sci total Environ. (2023) 868:161639. doi: 10.1016/j.scitotenv.2023.161639

PubMed Abstract | Crossref Full Text | Google Scholar

21. Hines CJ, Christianson AL, Jackson MV, Ye X, Pretty JR, Arnold JE, et al. An evaluation of the relationship among urine, air, and hand measures of exposure to bisphenol A (BPA) in US manufacturing workers. Ann work exposures Health. (2018) 62:840–51. doi: 10.1093/annweh/wxy042

PubMed Abstract | Crossref Full Text | Google Scholar

22. Wang Y, Du X, Wang D, Wang J, and Du J. Effects of Bisphenol A Exposure during Pregnancy and lactation on Hippocampal Function in Newborn Rats. Int J Med Sci. (2020) 17:1751–62. doi: 10.7150/ijms.47300

PubMed Abstract | Crossref Full Text | Google Scholar

23. Qu J, Guo R, Liu L, Ren F, and Jin H. Occurrence of bisphenol analogues and their conjugated metabolites in foodstuff. Sci total Environ. (2024) 948:174922. doi: 10.1016/j.scitotenv.2024.174922

PubMed Abstract | Crossref Full Text | Google Scholar

24. Estévez-Danta A, Montes R, Prieto A, Santos MM, Orive G, Lertxundi U, et al. Wastewater-based epidemiology methodology to investigate human exposure to bisphenol A, bisphenol F and bisphenol S. Water Res. (2024) 261:122016. doi: 10.1016/j.watres.2024.122016

PubMed Abstract | Crossref Full Text | Google Scholar

25. Nakajima Y, Goldblum RM, and Midoro-Horiuti T. Fetal exposure to bisphenol A as a risk factor for the development of childhood asthma: an animal model study. Environ health: A Global Access Sci Source. (2012) 11:8. doi: 10.1186/1476-069X-11-8

PubMed Abstract | Crossref Full Text | Google Scholar

26. Thoene M, Rytel L, Dzika E, Włodarczyk A, Kruminis-Kaszkiel E, Konrad P, et al. Bisphenol A causes liver damage and selectively alters the neurochemical coding of intrahepatic parasympathetic nerves in juvenile porcine models under physiological conditions. Int J Mol Sci 18. (2017). doi: 10.3390/ijms18122726

PubMed Abstract | Crossref Full Text | Google Scholar

27. Quirós-Alcalá L, Hansel NN, McCormack M, Calafat AM, Ye X, Peng RD, et al. Exposure to bisphenols and asthma morbidity among low-income urban children with asthma. J Allergy Clin Immunol. (2021) 147:577–586.e7. doi: 10.1016/j.jaci.2020.05.031

PubMed Abstract | Crossref Full Text | Google Scholar

28. Gerona RR, Pan J, Zota AR, Schwartz JM, Friesen M, Taylor JA, et al. Direct measurement of Bisphenol A (BPA), BPA glucuronide and BPA sulfate in a diverse and low-income population of pregnant women reveals high exposure, with potential implications for previous exposure estimates: a cross-sectional study. Environ health: A Global Access Sci Source. (2016) 15:50. doi: 10.1186/s12940-016-0131-2

PubMed Abstract | Crossref Full Text | Google Scholar

29. Suwannarin N, Nishihama Y, Isobe T, and Nakayama SF. Urinary concentrations of environmental phenol among pregnant women in the Japan Environment and Children’s Study. Environ Int. (2024) 183:108373. doi: 10.1016/j.envint.2023.108373

PubMed Abstract | Crossref Full Text | Google Scholar

30. Oliviero F, Marmugi A, Viguié C, Gayrard V, Picard-Hagen N, and Mselli-Lakhal L. Are BPA substitutes as obesogenic as BPA? Int J Mol Sci. (2022) 23.

PubMed Abstract | Google Scholar

31. Sigvaldsen A, Frederiksen H, Højsager FD, Andersson AM, Juul A, Boye H, et al. Prenatal and childhood exposure to bisphenols and bone mineral density in 7-year-old children from the Odense Child Cohort. Int J hygiene Environ Health. (2024) 260:114408. doi: 10.1016/j.ijheh.2024.114408

PubMed Abstract | Crossref Full Text | Google Scholar

32. Verstraete SG, Wojcicki JM, Perito ER, and Rosenthal P. Bisphenol a increases risk for presumed non-alcoholic fatty liver disease in Hispanic adolescents in NHANES 2003-2010. Environ health: A Global Access Sci Source. (2018) 17:12. doi: 10.1186/s12940-018-0356-3

PubMed Abstract | Crossref Full Text | Google Scholar

33. Kim D, Yoo ER, Li AA, Cholankeril G, Tighe SP, Kim W, et al. Elevated urinary bisphenol A levels are associated with non-alcoholic fatty liver disease among adults in the United States. Liver international: Off J Int Assoc Study Liver. (2019) 39:1335–42. doi: 10.1111/liv.2019.39.issue-7

PubMed Abstract | Crossref Full Text | Google Scholar

34. An SJ, Yang EJ, Oh S, Park KJ, Kim T, Hong YP, et al. The association between urinary bisphenol A levels and nonalcoholic fatty liver disease in Korean adults: Korean National Environmental Health Survey (KoNEHS) 2015-2017. Environ Health preventive Med. (2021) 26:91. doi: 10.1186/s12199-021-01010-7

PubMed Abstract | Crossref Full Text | Google Scholar

35. Liang J, Xu C, Xu J, Yang C, Kong W, Xiao Z, et al. PPARα Senses bisphenol S to trigger EP300-mediated autophagy blockage and hepatic steatosis. Environ Sci Technol. (2023) 57:21581–92. doi: 10.1021/acs.est.3c05010

PubMed Abstract | Crossref Full Text | Google Scholar

36. Mancia G, Kreutz R, Brunström M, Burnier M, Grassi G, and Januszewicz A. 2023 ESH Guidelines for the management of arterial hypertension The Task Force for the management of arterial hypertension of the European Society of Hypertension: Endorsed by the International Society of Hypertension (ISH) and the European Renal Association (ERA): Erratum. J hypertension. (2024) 42:194. doi: 10.1097/HJH.0000000000003621

PubMed Abstract | Crossref Full Text | Google Scholar

37. Ozturk A, Olson MC, Samir AE, and Venkatesh SK. Liver fibrosis assessment: MR and US elastography. Abdominal Radiol (New York). (2022) 47:3037–50. doi: 10.1007/s00261-021-03269-4

PubMed Abstract | Crossref Full Text | Google Scholar

38. Lee JH, Kim D, Kim HJ, Lee CH, Yang JI, Kim W, et al. Hepatic steatosis index: a simple screening tool reflecting nonalcoholic fatty liver disease. Digestive liver Dis. (2010) 42:503–8. doi: 10.1016/j.dld.2009.08.002

PubMed Abstract | Crossref Full Text | Google Scholar

39. Le Magueresse-Battistoni B. Endocrine disrupting chemicals and metabolic disorders in the liver: What if we also looked at the female side? Chemosphere. (2021) 268:129212. doi: 10.1016/j.chemosphere.2020.129212

PubMed Abstract | Crossref Full Text | Google Scholar

40. Dolce A and Della Torre S. Sex, nutrition, and NAFLD: relevance of environmental pollution. Nutrients. (2023) 15. doi: 10.3390/nu15102335

PubMed Abstract | Crossref Full Text | Google Scholar

41. Xiao Z, Liu M, Yang F, Liu G, Liu J, Zhao W, et al. Programmed cell death and lipid metabolism of macrophages in NAFLD. Front Immunol. (2023) 14:1118449. doi: 10.3389/fimmu.2023.1118449

PubMed Abstract | Crossref Full Text | Google Scholar

42. Vitulo M, Gnodi E, Rosini G, Meneveri R, Giovannoni R, and Barisani D. Current therapeutical approaches targeting lipid metabolism in NAFLD. Int J Mol Sci. (2023) 24. doi: 10.3390/ijms241612748

PubMed Abstract | Crossref Full Text | Google Scholar

43. Badmus OO, Hillhouse SA, Anderson CD, Hinds TD, and Stec DE. Molecular mechanisms of metabolic associated fatty liver disease (MAFLD): functional analysis of lipid metabolism pathways. Clin Sci (London England: 1979). (2022) 136:1347–66. doi: 10.1042/CS20220572

PubMed Abstract | Crossref Full Text | Google Scholar

44. Marmugi A, Ducheix S, Lasserre F, Polizzi A, Paris A, Priymenko N, et al. Low doses of bisphenol A induce gene expression related to lipid synthesis and trigger triglyceride accumulation in adult mouse liver. Hepatology (Baltimore Md.). (2012) 55:395–407.

PubMed Abstract | Google Scholar

45. Savastano S, Tarantino G, D’Esposito V, Passaretti F, Cabaro S, Liotti A, et al. Bisphenol-A plasma levels are related to inflammatory markers, visceral obesity and insulin-resistance: a cross-sectional study on adult male population. J Trans Med. (2015) 13:169. doi: 10.1186/s12967-015-0532-y

PubMed Abstract | Crossref Full Text | Google Scholar

46. Fang R, Yang S, Gu X, Li C, Bi N, and Wang HL. Early-life exposure to bisphenol A induces dysregulation of lipid homeostasis by the upregulation of SCD1 in male mice. Environ pollution (Barking Essex: 1987). (2022) 304:119201. doi: 10.1016/j.envpol.2022.119201

PubMed Abstract | Crossref Full Text | Google Scholar

47. Li C, Shen N, Yang S, and Wang HL. Effects of BPA exposure and recovery on the expression of genes involved in the hepatic lipid metabolism in male mice. Toxics. (2023) 11. doi: 10.3390/toxics11090775

PubMed Abstract | Crossref Full Text | Google Scholar

48. Long Z, Fan J, Wu G, Liu X, Wu H, Liu J, et al. Gestational bisphenol A exposure induces fatty liver development in male offspring mice through the inhibition of HNF1b and upregulation of PPARγ. Cell Biol Toxicol. (2021) 37:65–84. doi: 10.1007/s10565-020-09535-3

PubMed Abstract | Crossref Full Text | Google Scholar

49. Zhang Y, Han S, Li T, Zhu L, and Wei F. Bisphenol A induces non-alcoholic fatty liver disease by promoting the O-GlcNAcylation of NLRP3. Arch Physiol Biochem. (2024) 130:814–22. doi: 10.1080/13813455.2023.2288533

PubMed Abstract | Crossref Full Text | Google Scholar

50. Shimpi PC, More VR, Paranjpe M, Donepudi AC, Goodrich JM, Dolinoy DC, et al. Hepatic lipid accumulation and nrf2 expression following perinatal and peripubertal exposure to bisphenol A in a mouse model of nonalcoholic liver disease. Environ Health Perspect. (2017) 125:087005. doi: 10.1289/EHP664

PubMed Abstract | Crossref Full Text | Google Scholar

51. Sun L, Ling Y, Jiang J, Wang D, Wang J, Li J, et al. Differential mechanisms regarding triclosan vs. bisphenol A and fluorene-9-bisphenol induced zebrafish lipid-metabolism disorders by RNA-Seq. Chemosphere. (2020) 251:126318. doi: 10.1016/j.chemosphere.2020.126318

PubMed Abstract | Crossref Full Text | Google Scholar

52. Lin Y, Ding D, Huang Q, Liu Q, Lu H, Lu Y, et al. Downregulation of miR-192 causes hepatic steatosis and lipid accumulation by inducing SREBF1: Novel mechanism for bisphenol A-triggered non-alcoholic fatty liver disease. Biochim Biophys Acta Mol Cell Biol Lipids. (2017) 1862:869–82. doi: 10.1016/j.bbalip.2017.05.001

PubMed Abstract | Crossref Full Text | Google Scholar

53. Qin J, Ru S, Wang W, Hao L, Ru Y, Wang J, et al. Long-term bisphenol S exposure aggravates non-alcoholic fatty liver by regulating lipid metabolism and inducing endoplasmic reticulum stress response with activation of unfolded protein response in male zebrafish. Environ pollution (Barking Essex: 1987). (2020) 263:114535. doi: 10.1016/j.envpol.2020.114535

PubMed Abstract | Crossref Full Text | Google Scholar

54. Fan Y, Li S, Yang X, Bai S, Tang M, Zhang X, et al. Multi-omics approach characterizes the role of Bisphenol F in disrupting hepatic lipid metabolism. Environ Int. (2024) 187:108690. doi: 10.1016/j.envint.2024.108690

PubMed Abstract | Crossref Full Text | Google Scholar

55. Zhang L, Xie X, Tao J, Wang S, Hu M, Wang X, et al. Mystery of bisphenol F-induced nonalcoholic fatty liver disease-like changes: Roles of Drp1-mediated abnormal mitochondrial fission in lipid droplet deposition. Sci total Environ. (2023) 904:166831. doi: 10.1016/j.scitotenv.2023.166831

PubMed Abstract | Crossref Full Text | Google Scholar

56. Wang X, Wu L, Tao J, Ye H, Wang J, Gao R, et al. A lipidomic approach to bisphenol F-induced non-alcoholic fatty liver disease-like changes: altered lipid components in a murine model. Environ Sci pollution Res Int. (2023) 30:112644–59. doi: 10.1007/s11356-023-30306-0

PubMed Abstract | Crossref Full Text | Google Scholar

57. Younossi ZM, Razavi H, Sherman M, Allen AM, Anstee QM, Cusi K, et al. Addressing the high and rising global burden of metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH): from the growing prevalence to payors’ Perspective. Alimentary Pharmacol Ther. (2025). doi: 10.1111/apt.70020

PubMed Abstract | Crossref Full Text | Google Scholar

58. Lee KC, Wu PS, and Lin HC. Pathogenesis and treatment of non-alcoholic steatohepatitis and its fibrosis. Clin Mol hepatology. (2023) 29:77–98. doi: 10.3350/cmh.2022.0237

PubMed Abstract | Crossref Full Text | Google Scholar

59. Pjanic M. The role of polycarbonate monomer bisphenol-A in insulin resistance. PeerJ. (2017) 5:e3809. doi: 10.7717/peerj.3809

PubMed Abstract | Crossref Full Text | Google Scholar

60. Le Magueresse-Battistoni B, Multigner L, Beausoleil C, and Rousselle C. Effects of bisphenol A on metabolism and evidences of a mode of action mediated through endocrine disruption. Mol Cell Endocrinol. (2018) 475:74–91. doi: 10.1016/j.mce.2018.02.009

PubMed Abstract | Crossref Full Text | Google Scholar

61. Menale C, Grandone A, Nicolucci C, Cirillo G, Crispi S, Di Sessa A, et al. Bisphenol A is associated with insulin resistance and modulates adiponectin and resistin gene expression in obese children. Pediatr obesity. (2017) 12:380–7. doi: 10.1111/ijpo.12154

PubMed Abstract | Crossref Full Text | Google Scholar

62. Wang T, Li M, Chen B, Xu M, Xu Y, Huang Y, et al. Urinary bisphenol A (BPA) concentration associates with obesity and insulin resistance. J Clin Endocrinol Metab. (2012) 97:E223–7. doi: 10.1210/jc.2011-1989

PubMed Abstract | Crossref Full Text | Google Scholar

63. Ji H, Song N, Ren J, Li W, Xu B, Li H, et al. Metabonomics reveals bisphenol A affects fatty acid and glucose metabolism through activation of LXR in the liver of male mice. Sci total Environ. (2020) 703:134681. doi: 10.1016/j.scitotenv.2019.134681

PubMed Abstract | Crossref Full Text | Google Scholar

64. Dallio M, Masarone M, Errico S, Gravina AG, Nicolucci C, Di Sarno R, et al. Role of bisphenol A as environmental factor in the promotion of non-alcoholic fatty liver disease: in vitro and clinical study. Alimentary Pharmacol Ther. (2018) 47:826–37. doi: 10.1111/apt.2018.47.issue-6

PubMed Abstract | Crossref Full Text | Google Scholar

65. Figueiredo LS, Oliveira KM, Freitas IN, Silva JA Jr., Silva JN, Favero-Santos BC, et al. Bisphenol-A exposure worsens hepatic steatosis in ovariectomized mice fed on a high-fat diet: Role of endoplasmic reticulum stress and fibrogenic pathways. Life Sci. (2020) 256:118012. doi: 10.1016/j.lfs.2020.118012

PubMed Abstract | Crossref Full Text | Google Scholar

66. Jayashree S, Indumathi D, Akilavalli N, Sathish S, Selvaraj J, and Balasubramanian K. Effect of Bisphenol-A on insulin signal transduction and glucose oxidation in liver of adult male albino rat. Environ Toxicol Pharmacol. (2013) 35:300–10. doi: 10.1016/j.etap.2012.12.016

PubMed Abstract | Crossref Full Text | Google Scholar

67. Ahn C, Kang HS, Lee JH, Hong EJ, Jung EM, Yoo YM, et al. and octylphenol exacerbate type 1 diabetes mellitus by disrupting calcium homeostasis in mouse pancreas. Toxicol Lett. (2018) 295:162–72. doi: 10.1016/j.toxlet.2018.06.1071

PubMed Abstract | Crossref Full Text | Google Scholar

68. Puttabyatappa M, Martin JD, Andriessen V, Stevenson M, Zeng L, Pennathur S, et al. Developmental programming: Changes in mediators of insulin sensitivity in prenatal bisphenol A-treated female sheep. Reprod Toxicol (Elmsford N.Y.). (2019) 85:110–22.

PubMed Abstract | Google Scholar

69. Geng S, Wang S, Zhu W, Xie C, Li X, Wu J, et al. Curcumin attenuates BPA-induced insulin resistance in HepG2 cells through suppression of JNK/p38 pathways. Toxicol Lett. (2017) 272:75–83. doi: 10.1016/j.toxlet.2017.03.011

PubMed Abstract | Crossref Full Text | Google Scholar

70. Guo X, Yin X, Liu Z, and Wang J. Non-alcoholic fatty liver disease (NAFLD) pathogenesis and natural products for prevention and treatment. Int J Mol Sci 23. (2022). doi: 10.3390/ijms232415489

PubMed Abstract | Crossref Full Text | Google Scholar

71. Elswefy SE, Abdallah FR, Atteia HH, Wahba AS, and Hasan RA. Inflammation, oxidative stress and apoptosis cascade implications in bisphenol A-induced liver fibrosis in male rats. Int J Exp Pathol. (2016) 97:369–79. doi: 10.1111/iep.2016.97.issue-5

PubMed Abstract | Crossref Full Text | Google Scholar

72. Lama S, Vanacore D, Diano N, Nicolucci C, Errico S, Dallio M, et al. Ameliorative effect of Silybin on bisphenol A induced oxidative stress, cell proliferation and steroid hormones oxidation in HepG2 cell cultures. Sci Rep. (2019) 9:3228. doi: 10.1038/s41598-019-40105-8

PubMed Abstract | Crossref Full Text | Google Scholar

73. Pirozzi C, Lama A, Annunziata C, Cavaliere G, Ruiz-Fernandez C, Monnolo A, et al. Oral bisphenol A worsens liver immune-metabolic and mitochondrial dysfunction induced by high-fat diet in adult mice: cross-talk between oxidative stress and inflammasome pathway. Antioxidants (Basel Switzerland). (2020) 9. doi: 10.3390/antiox9121201

PubMed Abstract | Crossref Full Text | Google Scholar

74. Linillos-Pradillo B, Paredes SD, Ortiz-Cabello M, Schlumpf M, Lichtensteiger W, Vara E, et al. Activation of NLRP3 inflammasome in liver of long evans lactating rats and its perinatal effects in the offspring after bisphenol F exposure. Int J Mol Sci. (2023) 24. doi: 10.3390/ijms241814129

PubMed Abstract | Crossref Full Text | Google Scholar

75. Wan X, Xu C, Yu C, and Li Y. Role of NLRP3 inflammasome in the progression of NAFLD to NASH. Can J gastroenterology hepatology. (2016) 2016:6489012. doi: 10.1155/2016/6489012

PubMed Abstract | Crossref Full Text | Google Scholar

76. de Carvalho Ribeiro M and Szabo G. Role of the inflammasome in liver disease. Annu Rev Pathol. (2022) 17:345–65. doi: 10.1146/annurev-pathmechdis-032521-102529

PubMed Abstract | Crossref Full Text | Google Scholar

77. Puttabyatappa M, Saadat N, Elangovan VR, Dou J, Bakulski K, and Padmanabhan V. Developmental programming: Impact of prenatal bisphenol-A exposure on liver and muscle transcriptome of female sheep. Toxicol Appl Pharmacol. (2022) 451:116161. doi: 10.1016/j.taap.2022.116161

PubMed Abstract | Crossref Full Text | Google Scholar

78. Tarantino G, Citro V, and Balsano C. Liver-spleen axis in nonalcoholic fatty liver disease. Expert Rev gastroenterology hepatology. (2021) 15:759–69. doi: 10.1080/17474124.2021.1914587

PubMed Abstract | Crossref Full Text | Google Scholar

79. Brummer C, Singer K, Renner K, Bruss C, Hellerbrand C, Dorn C, et al. The spleen-liver axis supports obesity-induced systemic and fatty liver inflammation via MDSC and NKT cell enrichment. Mol Cell Endocrinol. (2025) 601:112518. doi: 10.1016/j.mce.2025.112518

PubMed Abstract | Crossref Full Text | Google Scholar

80. Shaibi T, Balug HN, Ben-Othman ME, Benjama AE, Elhensheri M, Lwaleed BA, et al. Exposure to low-dose bisphenol A induces spleen damage in a murine model: Potentially through oxidative stress? Open veterinary J. (2022) 12:23–32. doi: 10.5455/OVJ.2022.v12.i1.4

PubMed Abstract | Crossref Full Text | Google Scholar

81. Zhao C, Yong T, Zhang Y, Jin Y, Xiao Y, Wang H, et al. Evaluation of the splenic injury following exposure of mice to bisphenol S: A mass spectrometry-based lipidomics and imaging analysis. Environ Int. (2020) 135:105378. doi: 10.1016/j.envint.2019.105378

PubMed Abstract | Crossref Full Text | Google Scholar

82. Albillos A, de Gottardi A, and Rescigno M. The gut-liver axis in liver disease: Pathophysiological basis for therapy. J hepatology. (2020) 72:558–77. doi: 10.1016/j.jhep.2019.10.003

PubMed Abstract | Crossref Full Text | Google Scholar

83. Barouki R, Samson M, Blanc EB, Colombo M, Zucman-Rossi J, Lazaridis KN, et al. The exposome and liver disease - how environmental factors affect liver health. J hepatology. (2023) 79:492–505. doi: 10.1016/j.jhep.2023.02.034

PubMed Abstract | Crossref Full Text | Google Scholar

84. Lai KP, Chung YT, Li R, Wan HT, and Wong CK. Bisphenol A alters gut microbiome: Comparative metagenomics analysis. Environ pollution (Barking Essex: 1987). (2016) 218:923–30. doi: 10.1016/j.envpol.2016.08.039

PubMed Abstract | Crossref Full Text | Google Scholar

85. Wang J, Su C, Qian M, Wang X, Chen C, Liu Y, et al. Subchronic toxic effects of bisphenol A on the gut-liver-hormone axis in rats via intestinal flora and metabolism. Front Endocrinol. (2024) 15:1415216. doi: 10.3389/fendo.2024.1415216

PubMed Abstract | Crossref Full Text | Google Scholar

86. Diamante G, Cely I, Zamora Z, Ding J, Blencowe M, Lang J, et al. Systems toxicogenomics of prenatal low-dose BPA exposure on liver metabolic pathways, gut microbiota, and metabolic health in mice. Environ Int. (2021) 146:106260. doi: 10.1016/j.envint.2020.106260

PubMed Abstract | Crossref Full Text | Google Scholar

87. Chi Y, Zhu L, Wang Y, Peng C, Lin Y, Ji S, et al. Long-term Bisphenol S exposure induced gut microbiota dysbiosis, obesity, hepatic lipid accumulation, intestinal lesions and dyslipidemia in mice. Toxicology. (2024) 504:153798. doi: 10.1016/j.tox.2024.153798

PubMed Abstract | Crossref Full Text | Google Scholar

88. Beausoleil C, Le Magueresse-Battistoni B, Viguié C, Babajko S, Canivenc-Lavier MC, Chevalier N, et al. Regulatory and academic studies to derive reference values for human health: The case of bisphenol S. Environ Res. (2022) 204:112233. doi: 10.1016/j.envres.2021.112233

PubMed Abstract | Crossref Full Text | Google Scholar

89. Naville D, Labaronne E, Vega N, Pinteur C, Canet-Soulas E, Vidal H, et al. Metabolic outcome of female mice exposed to a mixture of low-dose pollutants in a diet-induced obesity model. PloS One. (2015) 10:e0124015. doi: 10.1371/journal.pone.0124015

PubMed Abstract | Crossref Full Text | Google Scholar

90. Rao A, Douglas SC, and Hall JM. Endocrine disrupting chemicals, hormone receptors, and acne vulgaris: A connecting hypothesis. Cells. (2021) 10. doi: 10.3390/cells10061439

PubMed Abstract | Crossref Full Text | Google Scholar

91. Manfo FP, Jubendradass R, Nantia EA, Moundipa PF, and Mathur PP. Adverse effects of bisphenol A on male reproductive function. Rev Environ contamination Toxicol. (2014) 228:57–82.

Google Scholar

92. Chen Y, Chen X, Li X, Liu Y, Guo Y, Wang Z, et al. Effects of bisphenol AF on growth, behavior, histology and gene expression in marine medaka (Oryzias melastigma). Chemosphere. (2022) 308:136424. doi: 10.1016/j.chemosphere.2022.136424

PubMed Abstract | Crossref Full Text | Google Scholar

93. Strakovsky RS, Wang H, Engeseth NJ, Flaws JA, Helferich WG, Pan YX, et al. (BPA) exposure leads to sex-specific modification of hepatic gene expression and epigenome at birth that may exacerbate high-fat diet-induced hepatic steatosis. Toxicol Appl Pharmacol. (2015) 284:101–12. doi: 10.1016/j.taap.2015.02.021

PubMed Abstract | Crossref Full Text | Google Scholar

94. Cabaton NJ, Poupin N, Canlet C, Tremblay-Franco M, Audebert M, Cravedi JP, et al. An untargeted metabolomics approach to investigate the metabolic modulations of hepG2 cells exposed to low doses of bisphenol A and 17β-estradiol. Front Endocrinol. (2018) 9:571. doi: 10.3389/fendo.2018.00571

PubMed Abstract | Crossref Full Text | Google Scholar

95. Kundakovic M, Gudsnuk K, Franks B, Madrid J, Miller RL, Perera FP, et al. Sex-specific epigenetic disruption and behavioral changes following low-dose in utero bisphenol A exposure. Proc Natl Acad Sci United States America. (2013) 110:9956–61. doi: 10.1073/pnas.1214056110

PubMed Abstract | Crossref Full Text | Google Scholar

96. Jiang Y, Xia W, Yang J, Zhu Y, Chang H, Liu J, et al. BPA-induced DNA hypermethylation of the master mitochondrial gene PGC-1α contributes to cardiomyopathy in male rats. Toxicology. (2015) 329:21–31. doi: 10.1016/j.tox.2015.01.001

PubMed Abstract | Crossref Full Text | Google Scholar

97. Zhang Y, Mustieles V, Williams PL, Souter I, Calafat AM, Demokritou M, et al. Association of preconception mixtures of phenol and phthalate metabolites with birthweight among subfertile couples. Environ Epidemiol (Philadelphia Pa.). (2022) 6:e222. doi: 10.1097/EE9.0000000000000222

PubMed Abstract | Crossref Full Text | Google Scholar

98. Chen Y, Wang Y, Cui Z, Liu W, Liu B, Zeng Q, et al. Endocrine disrupting chemicals: A promoter of non-alcoholic fatty liver disease. Front Public Health. (2023) 11:1154837. doi: 10.3389/fpubh.2023.1154837

PubMed Abstract | Crossref Full Text | Google Scholar

99. He Y, Zou J, Hong T, and Feng D. Association between Di-2-ethylhexyl phthalate and nonalcoholic fatty liver disease among US adults: Mediation analysis of body mass index and waist circumference in the NHANES. Food Chem Toxicol. (2023) 179:113968. doi: 10.1016/j.fct.2023.113968

PubMed Abstract | Crossref Full Text | Google Scholar

100. Wang M, Wang Y, Han J, Duan Z, Yin J, Ding R, et al. Gestational and lactational co-exposure to DEHP and BPA impairs hepatic function via PI3K/AKT/FOXO1 pathway in offspring. Toxics. (2023) 11. doi: 10.3390/toxics11030216

PubMed Abstract | Crossref Full Text | Google Scholar

101. Hermano Sampaio Dias A, Yadav R, Mokkawes T, Kumar A, Skaf MS, Sastri CV, et al. Biotransformation of bisphenol by human cytochrome P450 2C9 enzymes: A density functional theory study. Inorganic Chem. (2023) 62:2244–56. doi: 10.1021/acs.inorgchem.2c03984

PubMed Abstract | Crossref Full Text | Google Scholar

102. Fang M, Webster TF, Ferguson PL, and Stapleton HM. Characterizing the peroxisome proliferator-activated receptor (PPARγ) ligand binding potential of several major flame retardants, their metabolites, and chemical mixtures in house dust. Environ Health Perspect. (2015) 123:166–72. doi: 10.1289/ehp.1408522

PubMed Abstract | Crossref Full Text | Google Scholar

103. Labaronne E, Pinteur C, Vega N, Pesenti S, Julien B, Meugnier-Fouilloux E, et al. Low-dose pollutant mixture triggers metabolic disturbances in female mice leading to common and specific features as compared to a high-fat diet. J Nutr Biochem. (2017) 45:83–93. doi: 10.1016/j.jnutbio.2017.04.001

PubMed Abstract | Crossref Full Text | Google Scholar

104. Wei J, Sun X, Chen Y, Li Y, Song L, Zhou Z, et al. Perinatal exposure to bisphenol A exacerbates nonalcoholic steatohepatitis-like phenotype in male rat offspring fed on a high-fat diet. J Endocrinol. (2014) 222:313–25. doi: 10.1530/JOE-14-0356

PubMed Abstract | Crossref Full Text | Google Scholar

105. Marchlewicz E, McCabe C, Djuric Z, Hoenerhoff M, Barks J, Tang L, et al. Gestational exposure to high fat diets and bisphenol A alters metabolic outcomes in dams and offspring, but produces hepatic steatosis only in dams. Chemosphere. (2022) 286:131645. doi: 10.1016/j.chemosphere.2021.131645

PubMed Abstract | Crossref Full Text | Google Scholar

106. Xie C, Jiang X, Yin J, Jiang R, Zhu J, and Zou S. Bisphenol S accelerates the progression of high fat diet-induced NAFLD by triggering ferroptosis via regulating HMGCS2. J hazardous materials. (2025) 487:137166. doi: 10.1016/j.jhazmat.2025.137166

PubMed Abstract | Crossref Full Text | Google Scholar

107. Lv Z, Tang Z, Huang S, Hu X, Peng C, Chen Y, et al. In vivo hypoglycemic effects of bisphenol F exposure in high-fat diet mice. Chemosphere. (2023) 311:137066. doi: 10.1016/j.chemosphere.2022.137066

PubMed Abstract | Crossref Full Text | Google Scholar

108. Eckardt M, Simat TJ, and Bisphenol A. and alternatives in thermal paper receipts - a German market analysis from 2015 to 2017. Chemosphere. (2017) 186:1016–25. doi: 10.1016/j.chemosphere.2017.08.037

PubMed Abstract | Crossref Full Text | Google Scholar

109. Charles DA and Prince SE. Deciphering the molecular mechanism of NLRP3 in BPA-mediated toxicity: Implications for targeted therapies. Heliyon. (2024) 10:e28917. doi: 10.1016/j.heliyon.2024.e28917

PubMed Abstract | Crossref Full Text | Google Scholar

110. Akash MSH, Rasheed S, Rehman K, Imran M, and Assiri MA. Toxicological evaluation of bisphenol analogues: preventive measures and therapeutic interventions. RSC Adv. (2023) 13:21613–28. doi: 10.1039/D3RA04285E

PubMed Abstract | Crossref Full Text | Google Scholar

111. Marinovic MP, Sousa-Filho CPB, Batista FAH, Avelino TM, Cogliati B, Figueira ACM, et al. Green tea extract increases adiponectin and PPAR α levels to improve hepatic steatosis. J Nutr Biochem. (2022) 103:108957. doi: 10.1016/j.jnutbio.2022.108957

PubMed Abstract | Crossref Full Text | Google Scholar

112. Xie K, He X, Chen K, Sakao K, and Hou DX. Ameliorative effects and molecular mechanisms of vine tea on western diet-induced NAFLD. Food Funct. (2020) 11:5976–91. doi: 10.1039/D0FO00795A

PubMed Abstract | Crossref Full Text | Google Scholar

113. Khoo WY, Chrisfield BJ, Sae-Tan S, and Lambert JD. Mitigation of nonalcoholic fatty liver disease in high-fat-fed mice by the combination of decaffeinated green tea extract and voluntary exercise. J Nutr Biochem. (2020) 76:108262. doi: 10.1016/j.jnutbio.2019.108262

PubMed Abstract | Crossref Full Text | Google Scholar

114. Mohsenzadeh MS, Razavi BM, Imenshahidi M, Tabatabaee Yazdi SA, Mohajeri SA, and Hosseinzadeh H. Potential role of green tea extract and epigallocatechin gallate in preventing bisphenol A-induced metabolic disorders in rats: Biochemical and molecular evidence. Phytomedicine: Int J phytotherapy phytopharmacology. (2021) 92:153754. doi: 10.1016/j.phymed.2021.153754

PubMed Abstract | Crossref Full Text | Google Scholar

115. Zhang Z, Jia Y, Zhang C, Zhang Z, Jin F, Pan D, et al. Efficacy of epigallocatechin gallate (EGCG) and its underlying mechanism in preventing bisphenol-A-induced metabolic disorders in mice. J hazardous materials. (2024) 469:134098. doi: 10.1016/j.jhazmat.2024.134098

PubMed Abstract | Crossref Full Text | Google Scholar

116. Wang X, Gao M, Wang Z, Cui W, Zhang J, Zhang W, et al. Hepatoprotective effects of oridonin against bisphenol A induced liver injury in rats via inhibiting the activity of xanthione oxidase. Sci total Environ. (2021) 770:145301. doi: 10.1016/j.scitotenv.2021.145301

PubMed Abstract | Crossref Full Text | Google Scholar

117. Tejada S, Capó X, Mascaró CM, Monserrat-Mesquida M, Quetglas-Llabrés MM, Pons A, et al. Hepatoprotective effects of resveratrol in non-alcoholic fatty live disease. Curr pharmaceutical design. (2021) 27:2558–70. doi: 10.2174/18734286MTA1dODkwx

PubMed Abstract | Crossref Full Text | Google Scholar

118. Sodum N, Kumar G, Bojja SL, Kumar N, and Rao CM. Epigenetics in NAFLD/NASH: targets and therapy. Pharmacol Res. (2021) 167:105484. doi: 10.1016/j.phrs.2021.105484

PubMed Abstract | Crossref Full Text | Google Scholar

119. Liao JX, Chen YW, Shih MK, Tain YL, Yeh YT, Chiu MH, et al. Resveratrol butyrate esters inhibit BPA-induced liver damage in male offspring rats by modulating antioxidant capacity and gut microbiota. Int J Mol Sci. (2021) 22. doi: 10.3390/ijms22105273

PubMed Abstract | Crossref Full Text | Google Scholar

120. Chen J and Vitetta L. Gut microbiota metabolites in NAFLD pathogenesis and therapeutic implications. Int J Mol Sci. (2020) 21. doi: 10.3390/ijms21155214

PubMed Abstract | Crossref Full Text | Google Scholar

121. Kuang J, Wang J, Li Y, Li M, Zhao M, Ge K, et al. Hyodeoxycholic acid alleviates non-alcoholic fatty liver disease through modulating the gut-liver axis. Cell Metab. (2023) 35:1752–1766.e8. doi: 10.1016/j.cmet.2023.07.011

PubMed Abstract | Crossref Full Text | Google Scholar

122. Cheng MD, Li CL, Pei XY, Zhang YF, Jia DD, Zuo YB, et al. Integrative analysis of DNA methylome and transcriptome reveals epigenetic regulation of bisphenols-induced cardiomyocyte hypertrophy. Ecotoxicology Environ Saf. (2023) 263:115391. doi: 10.1016/j.ecoenv.2023.115391

PubMed Abstract | Crossref Full Text | Google Scholar

123. Deng X, Liang S, Tang Y, Li Y, Xu R, Luo L, et al. Adverse effects of bisphenol A and its analogues on male fertility: An epigenetic perspective. Environ pollution (Barking Essex: 1987). (2024) 345:123393. doi: 10.1016/j.envpol.2024.123393

PubMed Abstract | Crossref Full Text | Google Scholar

124. Zhao R, Fu J, Zhu L, Chen Y, and Liu B. Designing strategies of small-molecule compounds for modulating non-coding RNAs in cancer therapy. J Hematol Oncol. (2022) 15:14. doi: 10.1186/s13045-022-01230-6

PubMed Abstract | Crossref Full Text | Google Scholar

125. Wang J, Guo Y, Geng X, Hu J, Yan M, Sun Y, et al. Quantitative structure-activity relationship enables the rational design of lipid droplet-targeting carbon dots for visualizing bisphenol A-induced nonalcoholic fatty liver disease-like changes. ACS Appl materials interfaces. (2021) 13:44086–95. doi: 10.1021/acsami.1c13157

PubMed Abstract | Crossref Full Text | Google Scholar

126. Wang Q, Gao S, Chen B, Zhao J, Li W, and Wu L. Evaluating the effects of perinatal exposures to BPSIP on hepatic cholesterol metabolism in female and male offspring ICR mice. Environ Health Perspect. (2024) 132:97011. doi: 10.1289/EHP14643

PubMed Abstract | Crossref Full Text | Google Scholar

127. Cheng W, Zhou Y, Xie Y, Li Y, Zhou R, Wang H, et al. Combined effect of polystyrene microplastics and bisphenol A on the human embryonic stem cells-derived liver organoids: The hepatotoxicity and lipid accumulation. Sci total Environ. (2023) 854:158585. doi: 10.1016/j.scitotenv.2022.158585

PubMed Abstract | Crossref Full Text | Google Scholar

128. Wang Y and Xu H. CRISPR-edited hepatic organoids as drug screening platform for non-alcoholic fatty liver disease. Hepatobiliary Surg Nutr. (2023) 12:593–4. doi: 10.21037/hbsn-23-247

PubMed Abstract | Crossref Full Text | Google Scholar

129. Hendriks D, Brouwers JF, Hamer K, Geurts MH, Luciana L, Massalini S, et al. Engineered human hepatocyte organoids enable CRISPR-based target discovery and drug screening for steatosis. Nat Biotechnol. (2023) 41:1567–81. doi: 10.1038/s41587-023-01680-4

PubMed Abstract | Crossref Full Text | Google Scholar

130. Hochreuter MY, Dall M, Treebak JT, and Barrès R. MicroRNAs in non-alcoholic fatty liver disease: Progress and perspectives. Mol Metab. (2022) 65:101581. doi: 10.1016/j.molmet.2022.101581

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: non-alcoholic fatty liver disease, bisphenols, environmental epidemiology, molecular pathogenesis, bisphenol A

Citation: Li C-L, Yao Z-Y, Zhang Y-F, Cui X-T, Sun A, Cao J-Y and Wang Z-S (2025) Bisphenols exposure and non-alcoholic fatty liver disease: from environmental trigger to molecular pathogenesis. Front. Endocrinol. 16:1606654. doi: 10.3389/fendo.2025.1606654

Received: 10 April 2025; Accepted: 29 April 2025;
Published: 22 May 2025.

Edited by:

Marcia Hiriart, Universidad Nacional Autonoma de Mexico, Mexico

Reviewed by:

Giovanni Tarantino, University of Naples Federico II, Italy
Brigitte Le Magueresse-Battistoni, INSERM U1060 Laboratoire de Recherche en Cardiovasculaire, Métabolisme, diabétologie et Nutrition, France

Copyright © 2025 Li, Yao, Zhang, Cui, Sun, Cao and Wang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Zu-Sen Wang, d2FuZ3p1c2VuQDEyNi5jb20=; Jing-Yu Cao, Y2p5NzAyN0AxNjMuY29t

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.