Next Article in Journal
Infants Fed Breastmilk or 2′-FL Supplemented Formula Have Similar Systemic Levels of Microbiota-Derived Secondary Bile Acids
Next Article in Special Issue
Abdominal Obesity in Women with Polycystic Ovary Syndrome and Its Relationship with Diet, Physical Activity and Insulin Resistance: A Pilot Study
Previous Article in Journal
An Isocaloric High-Fat Diet Regulates Partially Genetically Determined Fatty Acid and Carbohydrate Uptake and Metabolism in Subcutaneous Adipose Tissue of Lean Adult Twins
Previous Article in Special Issue
Diet and Fertility Status: Relevance in Health and Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Sex, Nutrition, and NAFLD: Relevance of Environmental Pollution

Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(10), 2335; https://doi.org/10.3390/nu15102335
Submission received: 18 April 2023 / Revised: 12 May 2023 / Accepted: 13 May 2023 / Published: 16 May 2023
(This article belongs to the Special Issue Diet and Fertility Status: Relevance in Health and Disease)

Abstract

:
Non-alcoholic fatty liver disease (NAFLD) is the most common form of chronic liver disease and represents an increasing public health issue given the limited treatment options and its association with several other metabolic and inflammatory disorders. The epidemic, still growing prevalence of NAFLD worldwide cannot be merely explained by changes in diet and lifestyle that occurred in the last few decades, nor from their association with genetic and epigenetic risk factors. It is conceivable that environmental pollutants, which act as endocrine and metabolic disruptors, may contribute to the spreading of this pathology due to their ability to enter the food chain and be ingested through contaminated food and water. Given the strict interplay between nutrients and the regulation of hepatic metabolism and reproductive functions in females, pollutant-induced metabolic dysfunctions may be of particular relevance for the female liver, dampening sex differences in NAFLD prevalence. Dietary intake of environmental pollutants can be particularly detrimental during gestation, when endocrine-disrupting chemicals may interfere with the programming of liver metabolism, accounting for the developmental origin of NAFLD in offspring. This review summarizes cause–effect evidence between environmental pollutants and increased incidence of NAFLD and emphasizes the need for further studies in this field.

1. Introduction

Non-alcoholic fatty liver disease (NAFLD) is the most common form of chronic liver disease with a 30% prevalence in the general population [1] and represents an increasing public health issue being an independent risk factor for several comorbidities, such as type 2 diabetes (T2D), hypertension, dyslipidemia, atherosclerosis, and cardiovascular diseases (CVDs) among others [2,3,4]. In NAFLD patients, enhanced lipid uptake and de novo lipogenesis (DNL), insufficient fatty acid oxidation (FAO), and reduced lipid secretion lead to excessive accumulation of triglycerides (TG) within hepatocytes [5]. The increased lipid content together with the impaired FAO promotes the generation of reactive oxidative species (ROS) and lipotoxic lipid intermediates within the hepatocytes, fostering oxidative stress and endoplasmic reticulum (ER) stress [6,7,8]. Chronic oxidative stress, in turn, triggers a pro-inflammatory response, mainly mediated by JNK (c-Jun N-terminal kinase) and NF-κB (nuclear factor-κB) signaling pathways, that results in the increased production of pro-inflammatory cytokines (i.e., IL-6, interleukin 6; TNFα, tumor necrosis factor α) by hepatocytes and non-parenchymal cells [7,9,10,11]. The sustained activation of the pro-inflammatory response bolsters a chronic inflammatory state that triggers the recruitment of other immune cells and activates apoptosis and other cell death mechanisms, boosting the progression of NAFLD toward non-alcoholic steatohepatitis (NASH), fibrosis, and hepatocellular carcinoma (HCC) [9,10,11].
Despite the growing public health impact of NAFLD, treatment options remain limited, likely a consequence of the poor understanding of the biological drivers responsible for NAFLD pathogenesis and progression [5,12,13].

2. Nutrition, Sex Differences, and NAFLD

In addition to genetic and epigenetic factors [14,15], obesity, dietary habits, lifestyle, and gut microbiota dysbiosis have a major role in NAFLD incidence, development, and progression [5,16,17,18,19,20]. In particular, overnutrition and poor dietary habits trigger insulin resistance (IR) and increase adiposity, favoring lipid deposition in the liver [5,21]. Beyond positive energy intake, consumption of specific nutrients such as saturated and trans fatty acids or fructose can be particularly detrimental to liver health, facilitating NAFLD [16,17,22]. Overnutrition and unbalanced diets may induce gut dysbiosis and increase gut permeability, alter the gut–liver axis, and expose the liver to microbiota-derived harmful substances, such as lipopolysaccharide (LPS), ROS, pro-inflammatory cytokines, thereby increasing susceptibility to NAFLD [20].
Compared to men, women show a lower susceptibility to NAFLD, at least until menopause, after which NAFLD prevalence becomes similar for the two sexes [23,24], pointing to the protective effect of estrogens. Accordingly, the incidence of NAFLD is greater in women with reproductive dysfunctions characterized by altered estrogen levels (i.e., polycystic ovary syndrome, PCOS) with respect to fertile women [23,24]. Notably, circulating levels of 17β-estradiol are lower in pre-menopausal, post-menopausal, and PCOS women with NAFLD compared to their control counterparts, while estrogen-based hormone replacement therapy (HRT) reduces the risk of developing NAFLD in post-menopausal women [23,24]. In the liver of fertile female mice, estrogens act mainly through ERα (estrogen receptor alpha), which activity changes according to circulating estrogens, thus modulating the hepatic metabolism to the energy requirements characterizing each reproductive stage [25,26]. In the absence of such an oscillatory activation (e.g., after ovariectomy, OVX), liver metabolic homeostasis is altered, leading to hepatic lipid deposition and inflammation [25,27], a condition resembling the increased incidence of NAFLD observed in post-menopausal women [23,24].
By virtue of its role in the female liver, hepatic estrogen signaling strongly contributes to sex differences in the regulation of hepatic metabolism [28] and in the sex-specific susceptibility to NAFLD [23,24,29]. Hepatic ERα signaling confers to females but not to males the ability to adapt a metabolic response to the excess of dietary lipids, thus limiting liver lipid deposition in a diet-induced mouse model of NAFLD [29]. Notably, such a female-specific and ERα-dependent feature is associated with the ability to preserve the hepatic homeostasis of amino acids (AA), especially of branched-chain amino acids (BCAA) [29], in agreement with studies reporting an impaired BCAA metabolism [30,31] and sex-dimorphic changes in BCAA levels in patients with NAFLD/NASH [32]. Accordingly, BCAA shows the potential to alleviate hepatic steatosis and liver injury in a NASH mouse model [33]. Notably, a dietary formula enriched in essential AA and, especially, in BCAA has been shown to rescue the hepatic transcriptomic profile and limit liver lipid deposition in a mouse model of menopause, an effect that strongly relies on hepatic ERα [27].
This ERα-dependent, female-specific ability to modulate hepatic metabolism according to dietary AA may depend on the strict interplay between metabolism and reproduction gained during evolution by the female liver [18]. In addition to estrogens, indeed, dietary AA activates ERα in the liver of females to promote the progression of the reproductive cycle [17]. By virtue of this mechanism and of the sex-dimorphic expression of hepatic ERα, the regulation of hepatic AA metabolism is significantly different in the liver of the two sexes, and it is strongly affected by the nutritional status [28,29]. Under short-term fasting, liver ERα promotes in females, but not in males, the catabolism of the hepatic AA to preserve lipid synthesis, ensuring the progression of the reproductive cycle [28].
In this view, hepatic ERα may act as a sensor of hormonal and nutritional status and differently accounts for the regulation of hepatic metabolism in the two sexes, thus contributing to sex differences in NAFLD susceptibility [34,35]. Furthermore, ERα might have a role in counteracting NAFLD progression to NASH, as suggested by studies reporting a low expression of ERα in the liver of NASH compared to NAFLD patients [36].

3. Nutrition, Environmental Pollutants, and NAFLD

Increasing evidence indicates that dietary intake of several environmental pollutants, including persistent organic pollutants (POPs), endocrine disrupting chemicals (EDCs), heavy metals, and micro- and nanoplastics promotes NAFLD development and progression (Figure 1). In addition to ingestion, exposure to pollutants through inhalation (i.e., particulate matter) can be another risk factor for developing NAFLD, especially in the context of high urbanization due to the combination with other risk factors, including obesogenic food environment, circadian disruption by light at night, and reduced physical activity [16,18,37,38].

3.1. Persistent Endocrine Disrupting Chemicals

POPs are long-lived carbon-based organic chemicals that have become a concern for human health, given their large use in agricultural and industrial sectors and their potential to enter the food chain [39]. To this class belong pesticides, including organochlorine pesticides (OCPs) such as DDT (dichlorodiphenyltrichloroethane) and its metabolites; industrial chemicals, including long-chain perfluorinated chemicals such as PFOA (perfluorooctanoic acid) and PFOS (perfluorooctanesulfonic acid), PCBs (polychlorinated biphenyls), and PBDEs (polybrominated diphenyl ethers); by-products of industrial processes, including PCDDs (polychlorinated dibenzo-p-dioxins), PCDFs (polychlorinated dibenzofurans), and PAHs (polyaromatic hydrocarbons) [39]. Given their widespread distribution in the environment, their employment in food production (i.e., pesticides), and in food contact materials (i.e., phthalates) from which they can be released, POPs can enter the food chain. It has been estimated that over 90% of human exposure to POPs released into the environment occurs through the consumption of contaminated food [39]. Given their lipophilic nature, POPs can be accumulated especially in the adipose tissue [40,41,42,43], and can be sequestrated in the liver [44], thus leading to metabolic dysfunctions.
Several POPs, including industrial chemicals such as PCBs, plasticizers including phthalates and bisphenol A (BPA), and agricultural pesticides act as EDCs by interfering with hormone signaling and endocrine functions [45,46]. EDCs can disrupt endocrine function in a variety of ways, by interfering with the synthesis, transport, metabolism, and/or receptor binding of hormones, resulting in reproductive, developmental, and metabolic dysfunctions [47,48]. Accumulating evidence suggests that exposure to EDCs may increase NAFLD risk [45,46,49]. The steatosis-inducing effect of EDCs is mostly mediated by the binding with different nuclear receptors (NRs), especially ERα and PPARs (peroxisome proliferator-activated receptors) (Figure 2a). EDC-induced disruption of NR signaling promotes lipid accumulation within hepatocytes through several metabolic alterations, including the increase in lipid uptake, enhanced DNL, inhibition of FAO, and reduction of lipid secretion in the form of VLDL (very-low-density lipoproteins) particles and bile acids [46] (Figure 2b). Several EDCs can contribute to the progression of NAFLD and hepatic inflammation by inducing the production of cytokines and the polarization of Kupffer cells towards a pro-inflammatory phenotype, by increasing hepatocyte proliferation and immune cell infiltration, by promoting the transformation of hepatic stellate cells (HSCs) into myofibroblast-like cells thus favoring apoptosis, liver damage, and fibrosis development [46,50] (Figure 2b).
BPA is an anti-androgen and estrogen-like synthetic chemical, mostly employed in epoxy resins, polymer materials, and plastics; it can be released from food containers and water bottles [51]. Although its use has been banned by several countries, the study of the effects of exposure to BPA is still relevant and representative of other chemical compounds with potential estrogenic activity. In addition to cancer, hormonal disruption, immunosuppression, and infertility, BPA is linked to obesity and several metabolic disorders, including NAFLD [46,51,52,53]. A positive association has been found between high urinary levels of BPA and increased incidence of NAFLD [54,55]. In vitro and animal-based studies have demonstrated that BPA stimulates TG accumulation in liver cells by upregulating the expression of the genes involved in DNL and IR, and by disrupting lipid homeostasis [46,56,57,58,59]. Through the generation of oxidative stress [60] and of a pro-inflammatory status that involves the polarization of Kupffer cells toward an M1 phenotype [50], and through the cross-talk among these two signaling pathways [61], BPA further promotes the progression of NAFLD and liver injury [46,59]. When coupled with a high-fat diet (HFD), BPA exposure worsens hepatic steatosis by stimulating ROS-induced overexpression of Cd36 (fatty acid translocase) in the mouse liver, thus facilitating lipid uptake [62]. In rodents, BPA may lead to hepatic steatosis and hepatotoxicity also by inducing gut microbiota dysbiosis and activating the gut-liver axis [63,64]. In OVX mice fed with HFD, BPA exposure aggravates hepatic steatosis by upregulating genes involved in DNL, fatty acid β-oxidation, and mitochondrial uncoupling, lowering lipid export with decreased expression of Mttp (microsomal triglyceride transport protein), and inducing ER stress, resulting in hepatic lipotoxicity, collagen deposition, and fibrosis [65].
Perfluoroalkyl acids (PFAAs, i.e., PFOA; perfluorononanoic acid, PFNA) are persistent, active chemicals in the environment, that can be ingested also through contaminated food and water [66], and, due to their exceptional stability to degradation, are slowly eliminated by the human body [67]. Environmental exposure to high levels of PFAAs increases the risk of developing hepatic steatosis and NAFLD in adults and children [68,69] and promotes liver injury [70]. PFAAs administration induces hepatic steatosis in rodents [71,72,73] and increases lipogenic gene expression signatures in cultured hepatocytes [74]. In male mice, PFOS induces fatty liver in a dose and time-dependent manner, by upregulating Cd36 and Lpl (lipoprotein lipase), inhibiting mitochondrial β-oxidation, and causing a shift of the hepatic proteome [71,72]. In HFD-fed mice, PFOS facilitates liver inflammation and steatosis through the activation of NLRP3 (NLR family pyrin domain containing 3) inflammasome that mediates hepatocyte pyroptosis [73]. PFOA exacerbates HFD-induced hepatotoxicity and lipid accumulation in the liver through the activation of peroxisome proliferator-activated receptor α (PPARα) [75]. Other studies have demonstrated that the co-administration of PFOS and PFNA with HFD reduces hepatic lipid deposition, a paradoxical effect likely due to the reduced expression of hepatic PFAAs uptake transporters and organic anion transporter proteins in the presence of HFD [76]. In rodents, PFAAs-induced liver steatosis and toxicity have been mainly attributed to PPARα, which activation leads to increased expression of genes involved in FAO, lipogenesis (Srebf1, sterol regulatory element-binding transcription factor 1; Fasn, fatty acid synthase), and lipid uptake (Cd36) [77]. However, studies performed in PPARα KO mice have demonstrated that other nuclear receptors and transcription factors, including ERα, peroxisome proliferator-activated receptor γ (PPARγ), constitutive activated receptor (CAR), liver X receptor (LXRα), pregnane X receptor (PXR), farnesoid X receptor (FXR), and hepatocyte nuclear factor 4α (HNF4α), may account for PFAAs effects in the liver (Figure 2a), resulting in a metabolic shift from carbohydrate metabolism to fatty acid oxidation and in hepatic TG accumulation [78,79]. Likely a consequence of sex differences in their pharmacokinetics and tissue distribution [80,81], PFAAs differently account for steatosis and toxicity in the liver of the two sexes [82,83,84], with females showing a positive association with NAFLD and increased hepatic inflammation and injury compared to males [83,84].
Several studies have reported a correlation between exposure to pesticides such as DDT and its metabolites and metabolic disorders, including NAFLD [85,86,87,88], likely because the liver is the main target for these toxicants. Retrospective analysis of the U.S. National Health and Nutrition Examination Survey 2003–2004 has indicated that exposure to several OCPs, especially to oxychlordane, is strongly associated with hepatic steatosis and altered levels of hepatic enzymes [89]. Although the underlying mechanisms have not yet been fully clarified, multi-omic analysis has suggested that OCPs favor NAFLD by altering the hepatic expression of genes involved in liver steatosis, PPAR signaling and fatty acid metabolism, inflammation, and necrosis [90]. Notably, some altered signaling pathways are common among different OCPs or a mixture of OCPs, while others are typical for a specific chemical [90]. For example, DDT leads to an over-expression of genes and transcription factors known to be linked to IR, cell death, and necrosis; in addition, DDT increases the hepatic levels of arginine, proline, and putrescine, and lowers the levels of urea and ornithine, impacting on liver regeneration process and boosting NAFLD progression [90]. In addition to these direct effects on the liver, OCPs can trigger NAFLD also by causing gut microbiota dysbiosis [91] and inhibiting thermogenesis in brown adipose tissue (BAT) [92]. In treated mice, insecticides such as fipronil (a phenylpyrazole commonly used in agricultural and veterinary fields) and thiamethoxam (a major compound of neonicotinoids) alter lipid metabolism by increasing Pparγ and Fasn expression and promote the generation of oxidative stress and inflammation by decreasing Pparα and Gnmt (glycine n-methyltransferase), ultimately contributing to NAFLD and liver injury [93,94]. Fungicides such as myclobutanil and mancozeb contribute to hepatic lipid deposition, cellular damage, and NAFLD development and progression [95,96,97]. In mouse liver, the effects of Myclobutanil can be differently mediated by the signaling of FXR depending on nutritional conditions [95]. In HepG2 cells, myclobutanil, mancozeb, and other fungicides such as tributyltin (TBT) induce steatosis also via retinoid X receptor alpha (RXRα), and hepatotoxicity by decreasing the expression of anti-apoptotic markers [96,97,98].
Dioxins consist of a group of organochlorines that include PCDDs, PCDFs, PCBs, and other related compounds that persist in the environment and accumulate in the food chain [99,100]. Dioxins can be produced from natural sources (i.e., volcanoes, forest fires), but are mostly derived from human industrial activities, especially as by-products of organochloride and chlorine-containing substances such as polyvinyl chloride (PVC). Dioxins are neither readily metabolized nor excreted, with a mean half-life of several years in humans, and bioaccumulate in fat tissues due to their lipophilic nature [101]. Several epidemiological studies have reported an increased risk of developing cancers, nervous system degeneration, immune damage, thyroid disease, metabolic disorders, and reproductive and sexual development disorders in the general population as well as in subgroups such as Vietnam War veterans heavily exposed to dioxins [100,102]. With respect to liver health, TCDD (2, 3, 7, 8-tetrachlorodibenzo-p-dioxin), the most toxic dioxin, has been shown to alter blood and hepatic lipid levels, disrupt bile synthesis, impair the microbiome, and strongly contribute to NAFLD development and progression through the activation of aryl hydrocarbon receptor (AhR) [103,104,105,106,107]. Several other dioxins, including PCBs, have become a major concern for liver health, given their strong association with IR and NAFLD/NASH, especially under diet-inducing obesogenic conditions [86,108,109,110,111]. Mice show significant differences in hepatic transcriptomic response to TCDD, with greater changes in the liver of males than females, likely due to sex differences in hepatic detoxification and to a complex interaction between the AhR and sex hormone receptors [112]. Notably, TCDD-induced activation of AhR leads to the disruption of hepatic sexual dimorphism in mice [113] and to a sex-specific induction of genes such as Car and proteins such as FMO3 (flavin-containing dimethylaniline monooxygenase 3) only in the livers of male mice [112,114]. Conversely, prolonged exposure to low doses of dioxin impairs metabolic adaptability to HFD only in female mice, which show hyperglycemia and impaired glucose-induced plasma insulin [115]. Furthermore, hepatic metabolic alterations could be indirect and secondary to sex-dependent glucocorticoid signaling disturbances and clock-related gene expression modifications caused by dioxins in adipose tissue [116].
PAHs are chemical compounds comprised of carbon and hydrogen molecules in a cyclic arrangement, that can be generated from the incomplete combustion of coal, wood, oil, and gas, and through volcanic activity. Humans are exposed to PAHs mainly through inhalation, but also through the ingestion of food contaminated by environmental pollutants or during preparation techniques involving high temperatures (e.g., grilled, smoked, toasted, roasted, and fried foods) [117]. Owing to their lipophilicity, PAHs have a low clearance in the liver, where they are metabolized to epoxides, dihydrodiols, quinones, or phenols, thus producing ROS that cause hepatotoxicity [118]. Animal-based studies have reported that exposure to PAHs such as benzo[a]pyrene (BaP) promotes lipid uptake from blood and lipid biosynthesis in the liver, contributing to NAFLD, oxidative stress, hepatic inflammation, and injury [119,120]. By activating the AhR pathway, BaP causes an overexpression of the estrogen-metabolizing enzyme cytochrome P450 1A1 (CYP1A1), which affects the estrogen signaling pathway, leading to the suppression of FAO and TG export and to the increase in peripheral fat mobilization, resulting in hepatic lipid deposition [120]. Such a mechanism of action of BaP suggests that exposure to this chemical has the potential to affect especially women, who may thus lose the protective effect of estrogen signaling on hepatic steatosis [23,24].

3.2. Heavy Metals

“Heavy metals” refer to metals and metalloids with relatively high densities (more than 5 g/cm3), that can accumulate along the food chain, and may lead to high toxicity in living organisms [121,122,123]. The group of potentially toxic elements comprises cadmium (Cd), lead (Pb), nickel (Ni), chromium (Cr), mercury (Hg), and metalloids, such as arsenic (As), from both natural sources and industrial activities [123]. It is well known that exposure to xenobiotic metals can cause gastrointestinal, respiratory, cardiovascular, reproductive, renal, hemopoietic, and neurological disorders [123]. The oxidative stress caused by these metals, by rising levels of oxidative damage in a cell, destroys lipids, proteins, and DNA molecules, and supports carcinogenesis. Exposure to environmental heavy metals, especially through ingestion and inhalation, plays a role in the development of NAFLD even in lean men [124,125,126,127,128,129,130,131,132,133].
Industrial emissions, smoking, and consumption of contaminated food and water represent the main sources of Cd, another toxic heavy metal widely disseminated in the environment [121,134]. In the body, Cd accumulates mainly in the liver, where it causes hepatotoxicity [126,134]. Several population-based studies indicate that environmental Cd is a tangible risk factor for NAFLD [125,127,128]. Elevated urinary Cd levels have been found to correlate with markers of NAFLD/NASH, hepatic necroinflammation, and cytokine levels [129,135]. In rodent models, Cd exposure leads to fatty liver and non-specific chronic inflammation, apoptosis, and liver cell regeneration, facilitating NASH development [136,137,138]. In other mouse models of NAFLD, chronic exposure to low doses of Cd alters HFD-associated adverse health effects, by limiting or exacerbating lipid deposition and liver injury dependent on Cd concentration [139]. Once taken up from the bloodstream, Cd interacts with the liver through heavy metal binding proteins and leads to impaired regulation of lipid metabolism, marked hepatic mitochondrial dysfunctions along with significant suppression of sirtuin 1 (SIRT1), inhibition of mitochondrial FAO and autophagy [140] (Figure 3).
The main sources of Pb include natural soil enrichment, paint, industrial legacy, batteries, contemporary mining emissions, contaminated food and water, and differ depending on different regional zones [141]. The accumulation through the food chain is the main way of Pb exposure, followed by direct inhalation and skin contact [122]. In a cohort of 41,723 individuals, exposure to Pb resulted to be an independent risk factor for MAFLD (metabolic dysfunction-associated fatty liver disease) [130], a renewed name for NAFLD [142]. In a population-based study of 4582 subjects, blood Pb levels positively correlated with the suspected NAFLD [143]. Chronic Pb exposure during early childhood is associated with hepatic steatosis and hepatocellular injury in young adulthood [144]. Although the mechanism(s) are not fully uncovered, Pb exposure seems to promote NAFLD development and progression by enhancing the production of ROS and oxidative stress, resulting in changes in lipid peroxidation and reduced antioxidant activity in hepatic cells [131]. An interesting study by Daniel et al. showed that Pb exposure may favor NAFLD by lowering the levels of sorcin, a protein that confiscates ChREBP (carbohydrate-responsive element binding protein) within the cytoplasm, thus enhancing the nuclear shuttling and transactivation of ChREBP, that results in increased hepatic DNL [145] (Figure 3). Hepatic Pb-induced liver toxicity may be boosted by the activation of a pro-inflammatory response due to variations in the intercellular signaling between Kupffer cells and hepatic cells [146], or to variations in gut microbiota [147].
Positive correlations have been found between soil heavy metal mixture containing As and the risk of developing NAFLD [86,124,132] as well as between urinary As levels and fatty liver/NAFLD [133]. Exposure to As impairs the normal metabolic features, increases the risk of NAFLD, and induces liver damage and inflammation in mice fed with HFD [148]. As induces the mitochondrial production of ROS, which upregulates the level of mitophagy and oxidizes mitochondrial DNA [149]. In rodents, As promotes NASH due to increased lipid accumulation, autophagy and NLRP3 inflammasome activation, dysregulated expression of lipid-related genes, and alternative cell death processes such as ferroptosis, a type of programmed cell death dependent on iron and characterized by the accumulation of lipid peroxides [150] (Figure 3). As-induced activation of NLRP3 inflammasome promotes the maturation and secretion of pro-inflammatory cytokines such as IL-1β and IL-18 (interleukin 1β and 18), that, in turn, boost hepatic IR and favor NAFLD development [149]. In rat liver, As induces ferroptosis-mediated NASH by affecting the interaction between Mitofusin 2 (a physical tether between the endoplasmic reticulum and mitochondria) and inositol-requiring enzyme 1 alpha (IRE1α) [151].
The combined exposure to several heavy metals can further increase the risk of developing NAFLD and the associated comorbidities [86,131,132,152], further pointing to cumulative effects derived by multiple exposures.

3.3. Microplastics and Nanoplastics

In the so-called era of Plasticene, plastics and their derived micro- and nano-particles represent a concern for human health [153]. Microplastics (MPs) are defined as “synthetic solid particles or polymeric matrices, with regular or irregular shape and with size ranging from 1 µm to 5 mm, of either primary or secondary manufacturing origin, which are insoluble in water” [154], while nanoplastics (NPs) are particles smaller than 1 µm [155]. MPs and NPs can be of primary manufacturing origin when deliberately created for consumer and industrial uses, such as occurs for exfoliants in cleansers, cosmetics, drug delivery particles in medicines, and industrial air blasting [155,156]. Otherwise, MPs and NPs can be of secondary origin when derived from the degradation of macroplastics through enzyme-based biodegradation or non-biodegradation processes, such as thermal degradation, physical degradation, photodegradation, thermo-oxidative degradation, and hydrolysis [155,156].
MPs and NPs can occur in both aquatic and terrestrial environments, and eventually enter the human body through the ingestion of contaminated food and water, the inhalation of airborne plastic particles that originate from synthetic textiles and urban dust, and, in a minimal amount, through weakened skin barrier and skin wounds [155,157,158,159]. Among these three routes, the ingestion of plastic-containing food and drinks represents the main route for MPs and NPs to enter the human body, affecting mainly the digestive and secretory systems [155,160]. MPs fragments have been detected in several types of food, including honey, beer, salt, sugar, fish, shrimps, and bivalves, and have been found in 81% of tap and 93% of bottled water, respectively [155]. It has been estimated that the average human is consuming around 39,000 to 52,000 MPs particles per year; people who drink only bottled water can ingest an extra 90,000 particles, further pointing to the food chain as a major source of microplastic consumption by humans [155]. Notably, MPs have been found in stool samples and cirrhotic liver tissues, confirming the exposure of the human digestive tract to MPs [161,162]. Although analytical tools to detect the presence of NPs in food are not yet available, it is conceivable that NPs can occur in the food chain due to the degradation of MPs [155].
Although their widespread distribution, particles in the seafood and the aquatic environment represent the greatest risk of absolute exposure to MPs and NPs and a concern for human health, especially due to the long-term weathering of polymers and the leaching of polymer chemical additives [153,155]. Several chemical additives, including inert or reinforcing fillers, plasticizers, antioxidants, UV stabilizers, lubricants, heavy metals, dyes, and flame-retardants, are added during production to modify plastic qualities such as color and transparency, to enhance the performance of plastic products, and to improve thermal, electrical, and mechanical resistance [153]. Other chemicals can be absorbed by plastic particles from the surrounding environment. In almost all cases, added and adsorbed additives are not chemically bound to the plastic polymer, and, thus, can leach into the air, water, food, and, potentially, the human body [153].
After intestinal absorption or epidermal infiltration, small MPs and NPs enter the bloodstream and reach the liver, where they can be easily internalized through endocytosis or, in an energy-independent way, through passive diffusion in a dose–response manner [157]. Small MPs (25 nm~90 μm) and NPs can accumulate in the livers of marine fish and mammals, leading to hepatic morphological changes, inflammation (necrosis, infiltration), and accumulation of lipid droplets, thereby affecting the normal function of the liver and resulting in hepatotoxicity [155,157].
Increasing evidence suggests that ingested or inhaled MPs and NPs damage liver health [155,157] and facilitate NAFLD development and progression [163,164,165] (Figure 4). Oral exposure to polystyrene MPs induces metabolic disturbances, such as diabetes and NAFLD, especially in mice fed with HFD, mainly due to inflammation of the intestinal mucosa that impairs nutrient absorption [164]. In mice fed with HFD, polystyrene NPs potentiate liver damage and trigger the development of hepatic fibrosis, by interfering with liver lipid metabolism, lowering superoxide dismutase (SOD) activity, inducing oxidative stress, inflammation, and collagen fiber deposition [165]. Excessive production of ROS activates the PI3K/Akt (phosphoinositide-3-kinase/protein kinase B) signaling pathways, blocking insulin signal transduction, and leading to IR, which may account for the increased plasma glucose levels and liver lipid droplets in mice following long-term and low-dose oral administration of polystyrene NPs [166]. In human hepatic cells, the uptake, intracellular localization, and cytotoxic effects of polystyrene NPs depend on particle concentration and surface functionalization and induce profound metabolic changes, especially in mitochondrial-related processes, accounting for mitochondrial damage [167,168,169]. In human liver organoids, 1 μm polystyrene MPs microbeads have been found to disrupt lipid metabolism by increasing the expression of HNF4A, alter ATP production, promote ROS generation, oxidative stress by inducing CYP2E1 (cytochrome P450 family 2 subfamily E member 1), inflammation, lipotoxicity, and hepatotoxicity, thus providing evidence for the implication of these particles in human NAFLD [170].
From a mechanistic point of view, MPs/NPs promote liver lipid deposition by altering the hepatic expression of genes involved in lipid metabolism, such as PPARα and PPARγ and their target genes, leading to enhanced DNL (i.e., higher mRNA levels of Fasn, Srebp1) and lipid transport (i.e., higher mRNA levels of Cd36) and to a reduction of lipid catabolism (i.e., lower mRNA levels of Cpt1α, carnitine palmitoyltransferase 1A) and PPARα-mediated lipolysis [157,165,171]. MPs/NPs-associated chemical additives may prompt the excessive production of ROS, leading to lipid peroxidation and oxidative damage [157,163,170,172]. Compared to MPs, NPs easily aggregate in living organisms, favoring a stronger oxidative stress response and liver damage [157]. Exposure to MPs/NPs increases the expression and activities of inflammatory factors such as IL-1β, TNFα, and NF-κB, the infiltration in the liver, and polarization of macrophages toward an M1 phenotype, further aggravating liver injury [157]. Excessive oxidative stress and inflammation prompt ER stress and programmed cell death through apoptosis, pyroptosis, and ferroptosis [157,173,174,175].
Transcriptomic and metabolomic analysis has revealed that several biological processes related to energy metabolism, including glycolysis/gluconeogenesis, glucose transport, pentose phosphate pathway (PPP), fatty acid synthesis, and oxidation, are inhibited in the livers of mice and fish exposed to MPs [157,163,169]. Inhibition of the PPP pathway reduces the levels of the bio-reductant NADPH [176], further boosting oxidative-dependent cell damage. In fish, MPs more than NPs affect the energy metabolism, causing changes in feeding activity, depletion of the energy reserves, disturbances in the ability to mobilize energy reserves, defects in the mitochondrial membrane respiratory chain, and low levels of ATP/ADP/AMP in the liver [157,169,170].
Exposure to MPs and NPs may also enhance the expression and activity of cytochrome P450 oxidases (CYP450s, i.e., CYP1A1, CYP11A1, CYP19A1, CYP2E1) mainly involved in phase I reactions of oxidation, reduction, and hydrolysis, and may increase the activities of alkaline phosphatase (ALP), aspartate aminotransferase (AST), alanine aminotransferase (ALT), that are released into the blood during liver injury [157].
MPs and NPs damage intestinal function favoring gut dysbiosis and gut permeability and impair the balance of gut microbes leading to changes in the production of short-chain fatty acids, fatty acyl chains, choline, cholesterol, and AA, which in turn contribute to NAFLD pathogenesis favoring liver steatosis, inflammation, and fibrosis [163,164,177,178,179].
Although few studies investigated the sex differences associated with MPs/NPs exposure, evidence suggests that liver metabolic and toxic effects may be worse for females [180,181], given the major impact of these plastic particles and their associated chemical additives (i.e., EDCs) on female reproductive parameters and functions [181,182,183,184].

3.4. Air Particulate Matter

Particulate matter (PM) refers to solid particles and liquid droplets that are discharged into the air as a result of diesel use, road and agricultural dust, coal and biomass combustion, and emissions from industrial activities [185]. PM is composed of microscopic carbonaceous particles and the adsorbed chemicals, such PAHs, aryl hydrocarbons, volatile organic hydrocarbons, heavy metals (especially Fe, iron; Cu, copper; Ni), organic compounds, minerals, inorganic ions, and biological materials [185]. PM is classified depending on particle diameter: PM10 (<10 μm), PM2.5 (<2.5 μm), PM1 (<1 μm), and PM0.1 (<0.1 μm), also termed ultrafine particles (UFPs). The toxicity of specific subclasses of inhaled PM depends on particle size (with the smaller ones being the more toxic) and shape and on the composition of organic/inorganic fractions of PM as well as chemicals adsorbed to particle surface [186,187]. Being more abundant than UFPs, PM2.5 is considered to be the most harmful to human health [185].
In the respiratory compartment, <PM2.5 particles dissolve in the aqueous lining and reach the alveoli, where they induce the activation of macrophages towards an activated phenotype, triggering local and systemic inflammation [185]. Once crossed the alveolar barrier, <PM2.5 might enter the systemic circulation and, in a secondary phase, reach the liver, where it could be internalized via endocytosis-mediated mechanisms and favor metabolic dysfunctions and hepatic inflammation [188,189,190,191]. Three-week exposure to PM2.5 leads to the development of low-grade liver inflammation and enhances pro-inflammatory cytokines in plasma, while long-term exposure to “real world” PM2.5 leads to several molecular and metabolic derangements, including the rise in plasma TG, low/very low-density lipoproteins ratio (LDL/VLDL), IR, altered glucose metabolism, and a more sustained inflammatory response [191,192]. In rodent models, PM2.5 exposure synergizes with unbalanced dietary regimens (i.e., HFD) and aggravates metabolic and inflammatory dysfunctions, accelerating NAFLD progression to NASH [188,193].
From a mechanistic point of view, PM2.5 exposure can inhibit the expression of PPARα and PPARγ, impairing the regulation of glucose and lipid metabolism, FAO, and immune response, thus leading to hepatic steatosis, inflammation, and IR [194]. PM-induced metabolic effects are, indeed, mediated at least in part by the downregulation of PPARα and their lipid metabolism-related target genes in the liver (i.e., Cyp4a14; Cd36; Slc27a1, solute carrier family 27 member 1) and in BAT (i.e., Ucp1, uncoupling protein 1), which favor hepatic steatosis and BAT whitening [194]. In mice, exposure to PM2.5 for 16 weeks decreases glycolysis and Krebs cycle intermediates, while increasing the incorporation of 13C in the oxidative branch of the pentose phosphate pathway, suggesting that this metabolic shift can support de novo synthesis of fatty acids prior to the onset of IR [195]. PM2.5 treatment induces lipid synthesis in human hepatic HepG2 cells as well [196].
Both short-term acute and long-term chronic exposure to PM2.5 stimulate inflammatory cells and induce local tissue and systemic inflammation by increasing the levels of pro-inflammatory cytokines (IL-1β, IL-18, IL-6, and TNF-α) and liver injury associated with dyslipidemia [191]. In the liver, PM-induced cytokines activate Kupffer cells and promote inflammation through the activation of several molecular pathways, including c-JNKs-activator protein 1 (AP1), toll-like receptor 4 (TLR4), and NF-κB, thus favoring NASH development [197]. Additionally in adipocytes, together with impaired glucose tolerance, IR, and mitochondrial alterations, PM2.5 may prompt the expression of pro-inflammatory factors, such as TNFα and IL-6, and reduce anti-inflammatory factors, such as IL-10, further contributing to the propagation of systemic inflammation [198]. In turn, pro-inflammatory cytokines such as IL-6 may aggravate hepatic IR by inducing the activation of the STAT3/SOCS3 (signal transducer and activator of transcription 3/suppressor of cytokine signaling 3) pathway [199].
Prolonged PM2.5 exposure affects liver health also through the generation of ROS, which elevates the risk of oxidative stress-driven NAFLD by triggering lipid accumulation in the liver [192]. ROS production can also be mediated by heavy metals, especially Cr(VI), Pb, and As, contained within or adsorbed to particles as well as by electrophilic reaction metabolites derived from organic compounds attached to the surface of particles [200]. PM2.5 -induced production of ROS may reduce cell antioxidant capacity by impairing the translocation of nuclear transcription factor NRF-2 (nuclear factor erythroid 2–related factor 2) to the nucleus, thus affecting the expression of their target genes and the activity of antioxidant enzymes such as SOD, CAT (catalase), GPX (glutathione peroxidase), further propagating the oxidative stress [192,201]. Excessive ROS production and insufficient antioxidant activity may damage hepatic cells by altering the structure and function of biological macromolecules such as DNA, proteins, and lipids and by activating NF-κB, apoptosis, JNK, and p53 signaling pathways [191]. In turn, PM2.5-induced activation of inflammation can boost the production of ROS and reactive nitrogen species [191,192], further fostering oxidative stress. Oxidative stress derived from long-term exposure to PM2.5 can cause ER stress, that, in turn, aggravates liver lipid accumulation and IR through the selective activation of the unfolded protein response (UPR) signaling pathways, thus accelerating the progression of NAFLD [202,203].
PM2.5 exposure can promote NAFLD development and progression even as a consequence of changes in the intestinal microflora and gut dysbiosis [20,191,204,205]. Altered intestinal microflora may, indeed, increase the production and secretion of harmful substances, especially LPS [204]. Once entered the blood and reached the liver through the portal vein, intestinal bacteria-derived toxic derivatives may intensify a pro-inflammatory response, further exacerbating NAFLD progression [191,206]. Exposure to high doses of PM promotes a pro-oxidative and pro-inflammatory response and tight junction damage, leading to the death of gastrointestinal epithelial cells and to increased intestinal permeability [205]. In diet-induced mouse models of NAFLD, as well as in NAFLD patients, intestinal barrier dysfunctions and altered intestinal permeability may facilitate the transfer of LPS into the systemic circulation, exacerbating liver inflammation and the progression of NAFLD toward NASH and fibrosis [20,207,208].
Evidence suggests that long-term exposure to PM2.5 might have different metabolic effects in the two sexes [209]. Compared to their male counterparts, female mice showed greater IR, increased levels of hepatic TG, free fatty acids (FFA), and cholesterol (CH), and enhanced hepatic expression of ApoB (apolipoprotein B) and MTTP [209]. The greater vulnerability of females towards PM2.5 can be due to the inhibition of the hypothalamus–pituitary–adrenal (HPA) axis and to the decreased glucocorticoids levels, which may contribute to IR and to the disorders of hepatic lipid metabolism [209].
In a rodent model of menopause, the lack of estrogen action predisposes females to PM-negative effects by altering metabolic, oxidative, pro-inflammatory, and heat shock protein levels [210], further pointing to the greater susceptibility towards air pollution of females with unbalanced hormonal and reproductive systems.

4. Climate Change, Food Insecurity, and NAFLD

Although very poorly investigated, even climate change and global warming as consequences of environmental pollution may further account for the increasing incidence of NAFLD worldwide [211]. Global warming, indeed, threatens agriculture production leading to food insecurity and shortage of food supplies and favors the consumption of processed and imported foods with little nutritional value, thus accounting for an increased risk for humans to develop metabolic disorders, including NAFLD [211,212,213,214].
Several studies have shown that global warming favors the growth rate and production of microcystins (MCs), the most common class of liver toxins produced as secondary metabolites by a number of widely distributed freshwater cyanobacteria [215,216,217]. MCs are cyclic heptapeptides with two conventional amino acids in positions X and Y and a unique β-amino acid ADDA (3-Amino-9-methoxy-2,6,8-trimethyl-10-phenyldeca-4,6-dienoic acid) [215,218]. Based on their hydrophobicity and ability to form a chemical bond between the toxin and the protein phosphatases within cells, the two conventional amino acids differently contribute to MC toxicity and cell damage [218]. Among over 300 different MCs identified to date, microcystin-LR (MC-LR, so-called because it contains amino acids leucine (L) and arginine (R) in the X and Y positions, respectively), is the most abundant, persistent, and toxic MC variant, categorized as group 2B carcinogen by the International Agency for Research on Cancer (IARC) [218].
Once synthesized, MCs are stored intracellularly and only released into the water following cell lysis, either by a viral infection or during cell senescence. Human exposure to MCs occurs through the chronic and accidental ingestion of contaminated drinking or recreational water, inhalation or contact with the nasal mucous membrane, dermal contact with toxins during recreational activities, and consumption of contaminated food (i.e., vegetables, fruit, fish, and shellfish) irrigated with or grown in contaminated water [219]. Although the majority of MCs is eliminated with the feces, part of MCs is absorbed in the intestine and distributed to other organs, in particular to the liver [219], where MCs can be easily uptaken due to the higher expression of the MC organic anion transporting polypeptides (OATPs), especially Oatp1b2 as demonstrated for the mouse liver [218]. Once inside and taken up by the cells, the toxins promote cell damage by specifically inhibiting the serine/threonine protein phosphatases (PP)-PP1 and PP2A, leading to protein hyperphosphorylation, and alterations in the cytoskeleton, and increasing oxidative stress, cell death, cytoskeleton disruption, and cell lysis [219,220].
Being the liver the primary target organ for MC toxicity [218,219], several epidemiological and pre-clinical studies found a positive relationship between MCs and biomarkers of liver damage as well as dysfunctions in glucose, triglyceride, lipid, and cholesterol metabolic pathways [218,219,221,222,223,224,225]. Long-term environmental exposure to MCs increases the risk of NAFLD in humans [221] and exacerbates hepatic injury in rodent models of NAFLD [222,223,224,226], by increasing the expression of genes related to fatty acid biosynthesis and uptake, oxidative stress, pro-inflammation, necrosis, fibrosis, collagen deposition, hepatotoxicity, and by reducing the expression of genes involved in fatty acid β-oxidation, lipoprotein transport, and anti-inflammatory response. The relationship between MC exposure and NAFLD seems to be bi-directional since it has been reported that NAFLD may alter MC-LR toxicokinetics and acute toxicity [227]. Interestingly, MC-LR’s impact on liver functions in mice is different between the two sexes, with females showing higher susceptibility to MC-LR compared to males [228].
The increase in mean temperature consequent to global warming may promote NAFLD development even through the reduction of BAT activity with an impact on energy expenditure and thermogenesis [229,230]. Limited capacity and functionality of BAT might furthermore alter the regulation of glucose and lipid metabolism [231,232], triggering metabolic dysfunctions (i.e., increased adiposity, IR, T2D, and gestational diabetes) [229,233,234,235] and NAFLD development [236,237] due to the cross-talk between the BAT and the liver [231,238,239]. Environmental-induced BAT dysfunctionality may differently account for NAFLD susceptibility in the two sexes, as suggested by increasing evidence on sexual dimorphic and hormone-dependent regulation of BAT metabolism [240,241,242,243,244].

5. Dietary Intake of Environmental Pollutants, Female Subfertility, and NAFLD

Given the strict relationship between reproduction and the regulation of energy metabolism in female mammals [245], exposure to environmental pollutants may be detrimental, especially in women with reproductive dysfunctions; conversely, acting as EDCs, environmental pollutants may promote reproductive dysfunctions, further favoring metabolic disorders. A canonical example is represented by PCOS, an endocrine and metabolic condition affecting 5–18% of women, which diagnosis—according to the 2003 Rotterdam criteria—is confirmed with two of the three following criteria: clinical or biochemical hyperandrogenism, irregular cycles, and polycystic ovary morphology [246,247]. PCOS women show an increased susceptibly to the development of metabolic dysfunctions, including fatty liver, NAFLD, and NASH [248,249,250], likely a consequence of altered hormone levels (increased androgens and androgens/estrogens ratio; decreased estrogens) and IR [251,252,253]. Beyond genetic and epigenetic susceptibility, hypothalamic and ovarian dysfunctions, exposure to high androgen levels, IR, increased adiposity, and obesity [246,247], environmental pollutants have a role in PCOS physiopathology [254,255,256,257,258,259] and, thus, may further contribute to NAFLD incidence in PCOS women (Figure 5).
Exposure to BPA, the main component of plastic containers, enhances the production of androgens in ovarian theca cells and affects their hepatic metabolism by interacting with SHBG (sex hormone-binding globulin) and with enzymes regulating their hydroxylation at the hepatic level [254]. Androgens per se can downregulate BPA liver catabolism, increasing its circulating levels and, thereby, promoting PCOS [254].
A positive association has been found between PCOS and heavy metals, especially Cr, Cu, As, Cd, Pb, and Hg [258,260]. Acting as EDCs, heavy metals impair hypothalamic–pituitary–gonadal (HPG) axis, promote androgen synthesis, and several inflammatory and metabolic alterations, including the generation of oxidative stress [258,260]. In PCOS women, indeed, serum As, Cd, Pb, and Hg levels are increased and negatively correlated with serum levels of glutathione (GSH) and SOD [260].
Women exposed to high concentrations of fine air pollutants, including PM2.5, had an increased risk of developing PCOS [259]. Organic solvents found in indoor decoration may activate TNFα as well, and lead to excessive hepatic glucose production, low muscular glucose uptake, and impaired insulin sensitivity, further increasing metabolic and ovulatory dysfunctions in women suffering from PCOS [261].

6. Maternal Exposure to Pollutants and Developmental Origins of NAFLD

The epidemic and still growing prevalence of NAFLD worldwide [1] cannot be merely explained by changes in diet, lifestyle, and environmental factors that occurred in the last few decades, nor from their association with genetic risk factors that should have undergone few changes in such a limited time frame. In addition to these risk factors, it is conceivable that transgenerational effects may account for the developmental origins of NAFLD [25], contributing to the spreading of this pathology. There is a growing body of evidence showing that prenatal exposure to endocrine disruptors may alter liver metabolic programs and represent a risk factor for NAFLD development later in life [262,263] (Figure 6). In this view, prenatal exposure to environmental pollutants acting as endocrine and/or metabolic disruptors may contribute to or aggravate NAFLD development and progression, as suggested by several studies in rodents [264,265,266,267,268,269,270,271,272] and some clinical studies [273,274,275].
Gestational exposure to BPA induces fatty liver development in male offspring rodents by altering the expression and activity of several nuclear transcription factors [264,265,266]. Prenatal exposure to BPA promotes lipid deposition in the liver of offspring male mice through impairment of NR signaling, such as the inhibition of Hnf1b (hepatocyte nuclear factor 1b) and the upregulation of Pparγ [264]. In obese and diet-induced rodent models of NAFLD, perinatal exposure to BPA leads to the induction of NRF2 signaling, aggravates NAFLD onset, and exacerbates NAFLD progression toward a NASH-like phenotype [266,267,268]. The consumption of drinking water containing BPA during gestation impairs the PI3K/Akt/mTOR (PI3K/Akt/mammalian target of rapamycin) and TLR4/NF-κB pathways in the liver of offspring rats, resulting in the upregulation of lipogenic genes, activation of the inflammatory response, dysregulation of autophagy and development of NAFLD [265]. Possibly because the majority of pre-clinical studies have been conducted in males, it has not been clarified whether prenatal exposure to BPA might lead to sex differences in the offspring. In fact, while some studies have shown similar effects in the liver of both sexes [265], other studies have reported that gestational and developmental exposure to BPA leads to sexually dimorphic changes in hepatic gene expression and epigenome at birth and may exacerbate HFD-induced hepatic steatosis in a sex-specific fashion [269,270]. Perinatal exposure to endocrine-disrupting pollutants with estrogenic activity may, indeed, have a greater impact on male offspring, likely interfering with the liver metabolic programming prompted by estrogen signaling through organizational effects at this developmental stage [28].
In humans, higher exposure to PFAAs during pregnancy has been found to be associated with increased susceptibility to liver injury in children, who show higher serum levels of ALT, AST, and GGT (gamma-glutamyltransferase), and increased serum levels of aromatic AA and, especially, BCAA [273], the last ones having a role in NAFLD onset and progression [276,277,278,279].
Beyond direct effects on the liver, fetal exposure to environmental pollutants such as triphenyl phosphate (an organophosphate flame retardant) and nitenpyram (an insecticide used in agriculture) can trigger NAFLD and NASH development later in life as a consequence of impaired intestinal dysbiosis and colonic mucosal damage [271,280].
In utero exposure to heavy metals, possibly acting as EDCs, such as As and Hg increases offspring susceptibly to NAFLD, IR, liver inflammation, and injury [272,274].
In mice, gestational exposure to polystyrene NPs induces hepatic steatosis in the dams and in adult female offspring but not male offspring, by enhancing the expression of genes involved in DNL, FA uptake, and TG synthesis [281].
Maternal exposure to ≤PM2.5 predisposes adult male mice offspring to NAFLD and other long-term metabolic dysfunctions such as obesity, T2D, insulin resistance, hypertension, hyperlipidemia, and metabolic syndrome, likely affecting organogenesis and tissue functions [282,283,284,285]. In mice, prenatal exposure to diesel exhaust PM2.5 (DEP) leads to increased lipogenesis and worsens fatty acid oxidation, favoring hepatic lipid deposition in the adult male offspring [286]. In humans, the association between prenatal exposure to ≤PM2.5 and the risk of developing NAFLD later in life is more consistent, likely due to the very few population-based investigations completed [282]. Nevertheless, accordingly, with the multiple-hits hypothesis of NAFLD pathogenesis [287], some studies support the idea that early life PM2.5 exposure induces liver damage, especially in overweight/obese children [275]. Interestingly, prenatal DEP exposure has been shown to relieve hepatic steatosis and liver function in the offspring of mice fed with HFD, suggesting a complex interaction between nutritional and environmental conditions in prompting NAFLD [286]. Although very little investigated, maternal exposure to PM2.5 seems to have sex-specific effects in the offspring with a more severe impact on females, which show increased hepatic expression of markers of oxidative stress and inflammation [288].

7. Discussion

The prevalence of NAFLD and its associated diseases has become a significant health and economic burden nowadays and it is still rising. Although the well-known role of EDCs in NAFLD development, the specific contribution of dietary intake of environmental endocrine pollutants to the increasing incidence of NAFLD still represents a significant gap in our knowledge. Although rodent studies have provided the strongest evidence for this cause–effect association, only a small percentage of these studies have been focused on females, limiting the understanding of the molecular mechanisms through which females can be eventually more vulnerable to dietary exposure to environmental pollutants depending on their hormonal status. Given the strict interplay between metabolism and reproduction in females, dietary intake of contaminated food and water can be, indeed, particularly detrimental to the liver health of females with impaired hormonal signaling and reproductive dysfunctions. Conversely, environmental EDCs can trigger reproductive alterations in females, that, in turn, facilitate NAFLD development and progression. In addition to that, dietary intake of environmental endocrine pollutants during gestation can be a risk factor for developing NAFLD for the mothers as well as for the offspring, likely affecting the estrogen-driven programming of liver metabolism especially in males. Given this evidence, additional rodent studies are warranted to expand our knowledge of the consequences of dietary intake of environmental endocrine pollutants, with the aim to elucidate the underlying mechanisms of action and to identify valuable biomarkers and interventional strategies for the treatment of NAFLD.

Author Contributions

Conceptualization, S.D.T.; writing—original draft preparation, S.D.T.; writing—review and editing, A.D. and S.D.T.; visualization, S.D.T.; supervision, S.D.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data are publicly available.

Acknowledgments

To the Department of Pharmaceutical Sciences (DISFARM, University of Milan) for administrative support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Younossi, Z.M.; Golabi, P.; Paik, J.M.; Henry, A.; Van Dongen, C.; Henry, L. The Global Epidemiology of Nonalcoholic Fatty Liver Disease (NAFLD) and Nonalcoholic Steatohepatitis (NASH): A Systematic Review. Hepatol. Baltim. Md 2023, 77, 1335–1347. [Google Scholar] [CrossRef] [PubMed]
  2. Targher, G.; Corey, K.E.; Byrne, C.D.; Roden, M. The Complex Link between NAFLD and Type 2 Diabetes Mellitus—Mechanisms and Treatments. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 599–612. [Google Scholar] [CrossRef] [PubMed]
  3. Mantovani, A.; Scorletti, E.; Mosca, A.; Alisi, A.; Byrne, C.D.; Targher, G. Complications, Morbidity and Mortality of Nonalcoholic Fatty Liver Disease. Metabolism 2020, 111, 154170. [Google Scholar] [CrossRef]
  4. Adams, L.A.; Anstee, Q.M.; Tilg, H.; Targher, G. Non-Alcoholic Fatty Liver Disease and Its Relationship with Cardiovascular Disease and Other Extrahepatic Diseases. Gut 2017, 66, 1138–1153. [Google Scholar] [CrossRef] [PubMed]
  5. Powell, E.E.; Wong, V.W.-S.; Rinella, M. Non-Alcoholic Fatty Liver Disease. Lancet 2021, 397, 2212–2224. [Google Scholar] [CrossRef]
  6. Masarone, M.; Rosato, V.; Dallio, M.; Gravina, A.G.; Aglitti, A.; Loguercio, C.; Federico, A.; Persico, M. Role of Oxidative Stress in Pathophysiology of Nonalcoholic Fatty Liver Disease. Oxid. Med. Cell. Longev. 2018, 2018, 9547613. [Google Scholar] [CrossRef] [PubMed]
  7. Monserrat-Mesquida, M.; Quetglas-Llabrés, M.; Abbate, M.; Montemayor, S.; Mascaró, C.M.; Casares, M.; Tejada, S.; Abete, I.; Zulet, M.A.; Tur, J.A.; et al. Oxidative Stress and Pro-Inflammatory Status in Patients with Non-Alcoholic Fatty Liver Disease. Antioxidants 2020, 9, 759. [Google Scholar] [CrossRef]
  8. Lebeaupin, C.; Vallée, D.; Hazari, Y.; Hetz, C.; Chevet, E.; Bailly-Maitre, B. Endoplasmic Reticulum Stress Signalling and the Pathogenesis of Non-Alcoholic Fatty Liver Disease. J. Hepatol. 2018, 69, 927–947. [Google Scholar] [CrossRef]
  9. Schuster, S.; Cabrera, D.; Arrese, M.; Feldstein, A.E. Triggering and Resolution of Inflammation in NASH. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 349–364. [Google Scholar] [CrossRef] [PubMed]
  10. Kazankov, K.; Jørgensen, S.M.D.; Thomsen, K.L.; Møller, H.J.; Vilstrup, H.; George, J.; Schuppan, D.; Grønbæk, H. The Role of Macrophages in Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 145–159. [Google Scholar] [CrossRef]
  11. Luci, C.; Bourinet, M.; Leclère, P.S.; Anty, R.; Gual, P. Chronic Inflammation in Non-Alcoholic Steatohepatitis: Molecular Mechanisms and Therapeutic Strategies. Front. Endocrinol. 2020, 11, 597648. [Google Scholar] [CrossRef] [PubMed]
  12. Riazi, K.; Azhari, H.; Charette, J.H.; Underwood, F.E.; King, J.A.; Afshar, E.E.; Swain, M.G.; Congly, S.E.; Kaplan, G.G.; Shaheen, A.-A. The Prevalence and Incidence of NAFLD Worldwide: A Systematic Review and Meta-Analysis. Lancet Gastroenterol. Hepatol. 2022, 7, 851–861. [Google Scholar] [CrossRef] [PubMed]
  13. Carter, J.; Wang, S.; Friedman, S.L. Ten Thousand Points of Light: Heterogeneity Among the Stars of NASH Fibrosis. Hepatology 2021, 74, 543–546. [Google Scholar] [CrossRef] [PubMed]
  14. Jonas, W.; Schürmann, A. Genetic and Epigenetic Factors Determining NAFLD Risk. Mol. Metab. 2021, 50, 101111. [Google Scholar] [CrossRef]
  15. Choudhary, N.S.; Duseja, A. Genetic and Epigenetic Disease Modifiers: Non-Alcoholic Fatty Liver Disease (NAFLD) and Alcoholic Liver Disease (ALD). Transl. Gastroenterol. Hepatol. 2021, 6, 2. [Google Scholar] [CrossRef]
  16. Berná, G.; Romero-Gomez, M. The Role of Nutrition in Non-Alcoholic Fatty Liver Disease: Pathophysiology and Management. Liver Int. Off. J. Int. Assoc. Study Liver 2020, 40 (Suppl. 1), 102–108. [Google Scholar] [CrossRef]
  17. Yki-Järvinen, H.; Luukkonen, P.K.; Hodson, L.; Moore, J.B. Dietary Carbohydrates and Fats in Nonalcoholic Fatty Liver Disease. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 770–786. [Google Scholar] [CrossRef]
  18. Hallsworth, K.; Adams, L.A. Lifestyle Modification in NAFLD/NASH: Facts and Figures. JHEP Rep. Innov. Hepatol. 2019, 1, 468–479. [Google Scholar] [CrossRef]
  19. Aron-Wisnewsky, J.; Vigliotti, C.; Witjes, J.; Le, P.; Holleboom, A.G.; Verheij, J.; Nieuwdorp, M.; Clément, K. Gut Microbiota and Human NAFLD: Disentangling Microbial Signatures from Metabolic Disorders. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 279–297. [Google Scholar] [CrossRef]
  20. Kolodziejczyk, A.A.; Zheng, D.; Shibolet, O.; Elinav, E. The Role of the Microbiome in NAFLD and NASH. EMBO Mol. Med. 2019, 11, e9302. [Google Scholar] [CrossRef]
  21. Smith, G.I.; Shankaran, M.; Yoshino, M.; Schweitzer, G.G.; Chondronikola, M.; Beals, J.W.; Okunade, A.L.; Patterson, B.W.; Nyangau, E.; Field, T.; et al. Insulin Resistance Drives Hepatic de Novo Lipogenesis in Nonalcoholic Fatty Liver Disease. J. Clin. Investig. 2020, 130, 1453–1460. [Google Scholar] [CrossRef] [PubMed]
  22. Jensen, T.; Abdelmalek, M.F.; Sullivan, S.; Nadeau, K.J.; Green, M.; Roncal, C.; Nakagawa, T.; Kuwabara, M.; Sato, Y.; Kang, D.-H.; et al. Fructose and Sugar: A Major Mediator of Non-Alcoholic Fatty Liver Disease. J. Hepatol. 2018, 68, 1063–1075. [Google Scholar] [CrossRef] [PubMed]
  23. Della Torre, S. Beyond the X Factor: Relevance of Sex Hormones in NAFLD Pathophysiology. Cells 2021, 10, 2502. [Google Scholar] [CrossRef]
  24. Della Torre, S. Non-Alcoholic Fatty Liver Disease as a Canonical Example of Metabolic Inflammatory-Based Liver Disease Showing a Sex-Specific Prevalence: Relevance of Estrogen Signaling. Front. Endocrinol. 2020, 11, 572490. [Google Scholar] [CrossRef] [PubMed]
  25. Villa, A.; Della Torre, S.; Stell, A.; Cook, J.; Brown, M.; Maggi, A. Tetradian Oscillation of Estrogen Receptor Is Necessary to Prevent Liver Lipid Deposition. Proc. Natl. Acad. Sci. USA 2012, 109, 11806–11811. [Google Scholar] [CrossRef]
  26. Della Torre, S.; Mitro, N.; Fontana, R.; Gomaraschi, M.; Favari, E.; Recordati, C.; Lolli, F.; Quagliarini, F.; Meda, C.; Ohlsson, C.; et al. An Essential Role for Liver ERα in Coupling Hepatic Metabolism to the Reproductive Cycle. Cell Rep. 2016, 15, 360–371. [Google Scholar] [CrossRef]
  27. Della Torre, S.; Benedusi, V.; Pepe, G.; Meda, C.; Rizzi, N.; Uhlenhaut, N.H.; Maggi, A. Dietary Essential Amino Acids Restore Liver Metabolism in Ovariectomized Mice via Hepatic Estrogen Receptor α. Nat. Commun. 2021, 12, 6883. [Google Scholar] [CrossRef]
  28. Della Torre, S.; Mitro, N.; Meda, C.; Lolli, F.; Pedretti, S.; Barcella, M.; Ottobrini, L.; Metzger, D.; Caruso, D.; Maggi, A. Short-Term Fasting Reveals Amino Acid Metabolism as a Major Sex-Discriminating Factor in the Liver. Cell Metab. 2018, 28, 256–267. [Google Scholar] [CrossRef]
  29. Meda, C.; Barone, M.; Mitro, N.; Lolli, F.; Pedretti, S.; Caruso, D.; Maggi, A.; Della Torre, S. Hepatic ERα Accounts for Sex Differences in the Ability to Cope with an Excess of Dietary Lipids. Mol. Metab. 2020, 32, 97–108. [Google Scholar] [CrossRef]
  30. Gaggini, M.; Carli, F.; Rosso, C.; Buzzigoli, E.; Marietti, M.; Della Latta, V.; Ciociaro, D.; Abate, M.L.; Gambino, R.; Cassader, M.; et al. Altered Amino Acid Concentrations in NAFLD: Impact of Obesity and Insulin Resistance. Hepatology 2018, 67, 145–158. [Google Scholar] [CrossRef]
  31. Lake, A.D.; Novak, P.; Shipkova, P.; Aranibar, N.; Robertson, D.G.; Reily, M.D.; Lehman-McKeeman, L.D.; Vaillancourt, R.R.; Cherrington, N.J. Branched Chain Amino Acid Metabolism Profiles in Progressive Human Nonalcoholic Fatty Liver Disease. Amino Acids 2015, 47, 603–615. [Google Scholar] [CrossRef] [PubMed]
  32. Grzych, G.; Vonghia, L.; Bout, M.-A.; Weyler, J.; Verrijken, A.; Dirinck, E.; Chevalier Curt, M.J.; Van Gaal, L.; Paumelle, R.; Francque, S.; et al. Plasma BCAA Changes in Patients With NAFLD Are Sex Dependent. J. Clin. Endocrinol. Metab. 2020, 105, dgaa175. [Google Scholar] [CrossRef] [PubMed]
  33. Honda, T.; Ishigami, M.; Luo, F.; Lingyun, M.; Ishizu, Y.; Kuzuya, T.; Hayashi, K.; Nakano, I.; Ishikawa, T.; Feng, G.-G.; et al. Branched-Chain Amino Acids Alleviate Hepatic Steatosis and Liver Injury in Choline-Deficient High-Fat Diet Induced NASH Mice. Metabolism 2017, 69, 177–187. [Google Scholar] [CrossRef] [PubMed]
  34. Della Torre, S.; Maggi, A. Sex Differences: A Resultant of an Evolutionary Pressure? Cell Metab. 2017, 25, 499–505. [Google Scholar] [CrossRef]
  35. Maggi, A.; Della Torre, S. Sex, Metabolism and Health. Mol. Metab. 2018, 15, 3–7. [Google Scholar] [CrossRef] [PubMed]
  36. Erkan, G.; Yilmaz, G.; Konca Degertekin, C.; Akyol, G.; Ozenirler, S. Presence and Extent of Estrogen Receptor-Alpha Expression in Patients with Simple Steatosis and NASH. Pathol. Res. Pract. 2013, 209, 429–432. [Google Scholar] [CrossRef]
  37. Beulens, J.W.J.; Pinho, M.G.M.; Abreu, T.C.; den Braver, N.R.; Lam, T.M.; Huss, A.; Vlaanderen, J.; Sonnenschein, T.; Siddiqui, N.Z.; Yuan, Z.; et al. Environmental Risk Factors of Type 2 Diabetes—An Exposome Approach. Diabetologia 2022, 65, 263–274. [Google Scholar] [CrossRef] [PubMed]
  38. Mukherji, A.; Dachraoui, M.; Baumert, T.F. Perturbation of the Circadian Clock and Pathogenesis of NAFLD. Metabolism 2020, 111S, 154337. [Google Scholar] [CrossRef]
  39. Guo, W.; Pan, B.; Sakkiah, S.; Yavas, G.; Ge, W.; Zou, W.; Tong, W.; Hong, H. Persistent Organic Pollutants in Food: Contamination Sources, Health Effects and Detection Methods. Int. J. Environ. Res. Public. Health 2019, 16, 4361. [Google Scholar] [CrossRef]
  40. Myrmel, L.S.; Fjære, E.; Midtbø, L.K.; Bernhard, A.; Petersen, R.K.; Sonne, S.B.; Mortensen, A.; Hao, Q.; Brattelid, T.; Liaset, B.; et al. Macronutrient Composition Determines Accumulation of Persistent Organic Pollutants from Dietary Exposure in Adipose Tissue of Mice. J. Nutr. Biochem. 2016, 27, 307–316. [Google Scholar] [CrossRef]
  41. Reina-Pérez, I.; Artacho-Cordón, F.; Mustieles, V.; Castellano-Castillo, D.; Cardona, F.; Jiménez-Díaz, I.; López-Medina, J.A.; Alcaide, J.; Ocaña-Wilhelmi, L.; Iribarne-Durán, L.M.; et al. Cross-Sectional Associations of Persistent Organic Pollutants Measured in Adipose Tissue and Metabolic Syndrome in Clinically Diagnosed Middle-Aged Adults. Environ. Res. 2023, 222, 115350. [Google Scholar] [CrossRef]
  42. Rolle-Kampczyk, U.; Gebauer, S.; Haange, S.-B.; Schubert, K.; Kern, M.; Moulla, Y.; Dietrich, A.; Schön, M.R.; Klöting, N.; von Bergen, M.; et al. Accumulation of Distinct Persistent Organic Pollutants Is Associated with Adipose Tissue Inflammation. Sci. Total Environ. 2020, 748, 142458. [Google Scholar] [CrossRef]
  43. Moriceau, M.-A.; Cano-Sancho, G.; Kim, M.; Coumoul, X.; Emond, C.; Arrebola, J.-P.; Antignac, J.-P.; Audouze, K.; Rousselle, C. Partitioning of Persistent Organic Pollutants between Adipose Tissue and Serum in Human Studies. Toxics 2022, 11, 41. [Google Scholar] [CrossRef] [PubMed]
  44. Kumar, J.; Lind, L.; Salihovic, S.; van Bavel, B.; Ingelsson, E.; Lind, P.M. Persistent Organic Pollutants and Liver Dysfunction Biomarkers in a Population-Based Human Sample of Men and Women. Environ. Res. 2014, 134, 251–256. [Google Scholar] [CrossRef]
  45. Deierlein, A.L.; Rock, S.; Park, S. Persistent Endocrine-Disrupting Chemicals and Fatty Liver Disease. Curr. Environ. Health Rep. 2017, 4, 439–449. [Google Scholar] [CrossRef]
  46. Cano, R.; Pérez, J.L.; Dávila, L.A.; Ortega, Á.; Gómez, Y.; Valero-Cedeño, N.J.; Parra, H.; Manzano, A.; Véliz Castro, T.I.; Albornoz, M.P.D.; et al. Role of Endocrine-Disrupting Chemicals in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: A Comprehensive Review. Int. J. Mol. Sci. 2021, 22, 4807. [Google Scholar] [CrossRef] [PubMed]
  47. La Merrill, M.A.; Vandenberg, L.N.; Smith, M.T.; Goodson, W.; Browne, P.; Patisaul, H.B.; Guyton, K.Z.; Kortenkamp, A.; Cogliano, V.J.; Woodruff, T.J.; et al. Consensus on the Key Characteristics of Endocrine-Disrupting Chemicals as a Basis for Hazard Identification. Nat. Rev. Endocrinol. 2020, 16, 45–57. [Google Scholar] [CrossRef] [PubMed]
  48. Casals-Casas, C.; Desvergne, B. Endocrine Disruptors: From Endocrine to Metabolic Disruption. Annu. Rev. Physiol. 2011, 73, 135–162. [Google Scholar] [CrossRef]
  49. Foulds, C.E.; Treviño, L.S.; York, B.; Walker, C.L. Endocrine-Disrupting Chemicals and Fatty Liver Disease. Nat. Rev. Endocrinol. 2017, 13, 445–457. [Google Scholar] [CrossRef]
  50. Lv, Q.; Gao, R.; Peng, C.; Yi, J.; Liu, L.; Yang, S.; Li, D.; Hu, J.; Luo, T.; Mei, M.; et al. Bisphenol A Promotes Hepatic Lipid Deposition Involving Kupffer Cells M1 Polarization in Male Mice. J. Endocrinol. 2017, 234, 143–154. [Google Scholar] [CrossRef]
  51. Manzoor, M.F.; Tariq, T.; Fatima, B.; Sahar, A.; Tariq, F.; Munir, S.; Khan, S.; Nawaz Ranjha, M.M.A.; Sameen, A.; Zeng, X.-A.; et al. An Insight into Bisphenol A, Food Exposure and Its Adverse Effects on Health: A Review. Front. Nutr. 2022, 9, 1047827. [Google Scholar] [CrossRef] [PubMed]
  52. Vom Saal, F.S.; Myers, J.P. Bisphenol A and Risk of Metabolic Disorders. JAMA 2008, 300, 1353–1355. [Google Scholar] [CrossRef]
  53. Dallio, M.; Diano, N.; Masarone, M.; Gravina, A.G.; Patanè, V.; Romeo, M.; Di Sarno, R.; Errico, S.; Nicolucci, C.; Abenavoli, L.; et al. Chemical Effect of Bisphenol A on Non-Alcoholic Fatty Liver Disease. Int. J. Environ. Res. Public. Health 2019, 16, 3134. [Google Scholar] [CrossRef] [PubMed]
  54. An, S.J.; Yang, E.-J.; Oh, S.; Park, K.J.; Kim, T.; Hong, Y.-P.; Yang, Y.-J. 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 Prev. Med. 2021, 26, 91. [Google Scholar] [CrossRef] [PubMed]
  55. Kim, D.; Yoo, E.R.; Li, A.A.; Cholankeril, G.; Tighe, S.P.; Kim, W.; Harrison, S.A.; Ahmed, A. Elevated Urinary Bisphenol A Levels Are Associated with Non-Alcoholic Fatty Liver Disease among Adults in the United States. Liver Int. Off. J. Int. Assoc. Study Liver 2019, 39, 1335–1342. [Google Scholar] [CrossRef]
  56. Ismael, L.Q.; Abdulhameed, A.R.; Keong, Y.Y.; Abdullah, M.N.H.; Bahari, H.; Jie, T.J.; Yin, K.B. Bisphenol A Is a Carcinogen That Induces Lipid Accumulation, Peroxisome Proliferator-activated Receptor-γ Expression and Liver Disease. Exp. Ther. Med. 2022, 24, 735. [Google Scholar] [CrossRef]
  57. Song, D.; Chen, Y.; Wang, B.; Li, D.; Xu, C.; Huang, H.; Huang, S.; Liu, R. Bisphenol A Inhibits Autophagosome-Lysosome Fusion and Lipid Droplet Degradation. Ecotoxicol. Environ. Saf. 2019, 183, 109492. [Google Scholar] [CrossRef]
  58. Grasselli, E.; Cortese, K.; Voci, A.; Vergani, L.; Fabbri, R.; Barmo, C.; Gallo, G.; Canesi, L. Direct Effects of Bisphenol A on Lipid Homeostasis in Rat Hepatoma Cells. Chemosphere 2013, 91, 1123–1129. [Google Scholar] [CrossRef]
  59. Dallio, M.; Masarone, M.; Errico, S.; Gravina, A.G.; Nicolucci, C.; Di Sarno, R.; Gionti, L.; Tuccillo, C.; Persico, M.; Stiuso, P.; et al. Role of Bisphenol A as Environmental Factor in the Promotion of Non-Alcoholic Fatty Liver Disease: In Vitro and Clinical Study. Aliment. Pharmacol. Ther. 2018, 47, 826–837. [Google Scholar] [CrossRef]
  60. Federico, A.; Dallio, M.; Gravina, A.G.; Diano, N.; Errico, S.; Masarone, M.; Romeo, M.; Tuccillo, C.; Stiuso, P.; Morisco, F.; et al. The Bisphenol A Induced Oxidative Stress in Non-Alcoholic Fatty Liver Disease Male Patients: A Clinical Strategy to Antagonize the Progression of the Disease. Int. J. Environ. Res. Public. Health 2020, 17, 3369. [Google Scholar] [CrossRef]
  61. Elswefy, S.E.-S.; Abdallah, F.R.; Atteia, H.H.; Wahba, A.S.; Hasan, R.A. Inflammation, Oxidative Stress and Apoptosis Cascade Implications in Bisphenol A-Induced Liver Fibrosis in Male Rats. Int. J. Exp. Pathol. 2016, 97, 369–379. [Google Scholar] [CrossRef] [PubMed]
  62. Lee, J.-L.; Wang, Y.-C.; Hsu, Y.-A.; Chen, C.-S.; Weng, R.-C.; Lu, Y.-P.; Chuang, C.-Y.; Wan, L. Bisphenol A Coupled with a High-Fat Diet Promotes Hepatosteatosis through Reactive-Oxygen-Species-Induced CD36 Overexpression. Toxics 2022, 10, 208. [Google Scholar] [CrossRef] [PubMed]
  63. Feng, D.; Zhang, H.; Jiang, X.; Zou, J.; Li, Q.; Mai, H.; Su, D.; Ling, W.; Feng, X. Bisphenol A Exposure Induces Gut Microbiota Dysbiosis and Consequent Activation of Gut-Liver Axis Leading to Hepatic Steatosis in CD-1 Mice. Environ. Pollut. 2020, 265, 114880. [Google Scholar] [CrossRef]
  64. Liu, R.; Liu, B.; Tian, L.; Jiang, X.; Li, X.; Cai, D.; Sun, J.; Bai, W.; Jin, Y. Exposure to Bisphenol A Caused Hepatoxicity and Intestinal Flora Disorder in Rats. Int. J. Mol. Sci. 2022, 23, 8042. [Google Scholar] [CrossRef] [PubMed]
  65. Figueiredo, L.S.; Oliveira, K.M.; Freitas, I.N.; Silva, J.A.; Silva, J.N.; Favero-Santos, B.C.; Bonfleur, M.L.; Carneiro, E.M.; Ribeiro, R.A. 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. [Google Scholar] [CrossRef] [PubMed]
  66. Papadopoulou, E.; Poothong, S.; Koekkoek, J.; Lucattini, L.; Padilla-Sánchez, J.A.; Haugen, M.; Herzke, D.; Valdersnes, S.; Maage, A.; Cousins, I.T.; et al. Estimating Human Exposure to Perfluoroalkyl Acids via Solid Food and Drinks: Implementation and Comparison of Different Dietary Assessment Methods. Environ. Res. 2017, 158, 269–276. [Google Scholar] [CrossRef]
  67. Xu, Y.; Fletcher, T.; Pineda, D.; Lindh, C.H.; Nilsson, C.; Glynn, A.; Vogs, C.; Norström, K.; Lilja, K.; Jakobsson, K.; et al. Serum Half-Lives for Short- and Long-Chain Perfluoroalkyl Acids after Ceasing Exposure from Drinking Water Contaminated by Firefighting Foam. Environ. Health Perspect. 2020, 128, 077004. [Google Scholar] [CrossRef]
  68. Zhang, X.; Zhao, L.; Ducatman, A.; Deng, C.; von Stackelberg, K.E.; Danford, C.J.; Zhang, X. Association of Per- and Polyfluoroalkyl Substance Exposure with Fatty Liver Disease Risk in US Adults. JHEP Rep. Innov. Hepatol. 2023, 5, 100694. [Google Scholar] [CrossRef]
  69. Jin, R.; McConnell, R.; Catherine, C.; Xu, S.; Walker, D.I.; Stratakis, N.; Jones, D.P.; Miller, G.W.; Peng, C.; Conti, D.V.; et al. Perfluoroalkyl Substances and Severity of Nonalcoholic Fatty Liver in Children: An Untargeted Metabolomics Approach. Environ. Int. 2020, 134, 105220. [Google Scholar] [CrossRef]
  70. Costello, E.; Rock, S.; Stratakis, N.; Eckel, S.P.; Walker, D.I.; Valvi, D.; Cserbik, D.; Jenkins, T.; Xanthakos, S.A.; Kohli, R.; et al. Exposure to Per- and Polyfluoroalkyl Substances and Markers of Liver Injury: A Systematic Review and Meta-Analysis. Environ. Health Perspect. 2022, 130, 46001. [Google Scholar] [CrossRef]
  71. Wan, H.T.; Zhao, Y.G.; Wei, X.; Hui, K.Y.; Giesy, J.P.; Wong, C.K.C. PFOS-Induced Hepatic Steatosis, the Mechanistic Actions on β-Oxidation and Lipid Transport. Biochim. Biophys. Acta 2012, 1820, 1092–1101. [Google Scholar] [CrossRef] [PubMed]
  72. Marques, E.; Pfohl, M.; Auclair, A.; Jamwal, R.; Barlock, B.J.; Sammoura, F.M.; Goedken, M.; Akhlaghi, F.; Slitt, A.L. Perfluorooctanesulfonic Acid (PFOS) Administration Shifts the Hepatic Proteome and Augments Dietary Outcomes Related to Hepatic Steatosis in Mice. Toxicol. Appl. Pharmacol. 2020, 408, 115250. [Google Scholar] [CrossRef] [PubMed]
  73. Qin, Y.; Gu, T.; Ling, J.; Luo, J.; Zhao, J.; Hu, B.; Hua, L.; Wan, C.; Jiang, S. PFOS Facilitates Liver Inflammation and Steatosis: An Involvement of NLRP3 Inflammasome-Mediated Hepatocyte Pyroptosis. J. Appl. Toxicol. JAT 2022, 42, 806–817. [Google Scholar] [CrossRef] [PubMed]
  74. Louisse, J.; Rijkers, D.; Stoopen, G.; Janssen, A.; Staats, M.; Hoogenboom, R.; Kersten, S.; Peijnenburg, A. Perfluorooctanoic Acid (PFOA), Perfluorooctane Sulfonic Acid (PFOS), and Perfluorononanoic Acid (PFNA) Increase Triglyceride Levels and Decrease Cholesterogenic Gene Expression in Human HepaRG Liver Cells. Arch. Toxicol. 2020, 94, 3137–3155. [Google Scholar] [CrossRef]
  75. Li, X.; Wang, Z.; Klaunig, J.E. The Effects of Perfluorooctanoate on High Fat Diet Induced Non-Alcoholic Fatty Liver Disease in Mice. Toxicology 2019, 416, 1–14. [Google Scholar] [CrossRef] [PubMed]
  76. Pfohl, M.; Marques, E.; Auclair, A.; Barlock, B.; Jamwal, R.; Goedken, M.; Akhlaghi, F.; Slitt, A.L. An ’Omics Approach to Unraveling the Paradoxical Effect of Diet on Perfluorooctanesulfonic Acid (PFOS) and Perfluorononanoic Acid (PFNA)-Induced Hepatic Steatosis. Toxicol. Sci. Off. J. Soc. Toxicol. 2021, 180, 277–294. [Google Scholar] [CrossRef]
  77. Das, K.P.; Wood, C.R.; Lin, M.T.; Starkov, A.A.; Lau, C.; Wallace, K.B.; Corton, J.C.; Abbott, B.D. Perfluoroalkyl Acids-Induced Liver Steatosis: Effects on Genes Controlling Lipid Homeostasis. Toxicology 2017, 378, 37–52. [Google Scholar] [CrossRef]
  78. Bjork, J.A.; Butenhoff, J.L.; Wallace, K.B. Multiplicity of Nuclear Receptor Activation by PFOA and PFOS in Primary Human and Rodent Hepatocytes. Toxicology 2011, 288, 8–17. [Google Scholar] [CrossRef]
  79. Rosen, M.B.; Das, K.P.; Rooney, J.; Abbott, B.; Lau, C.; Corton, J.C. PPARα-Independent Transcriptional Targets of Perfluoroalkyl Acids Revealed by Transcript Profiling. Toxicology 2017, 387, 95–107. [Google Scholar] [CrossRef]
  80. Kim, S.-J.; Heo, S.-H.; Lee, D.-S.; Hwang, I.G.; Lee, Y.-B.; Cho, H.-Y. Gender Differences in Pharmacokinetics and Tissue Distribution of 3 Perfluoroalkyl and Polyfluoroalkyl Substances in Rats. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2016, 97, 243–255. [Google Scholar] [CrossRef]
  81. Huang, M.C.; Dzierlenga, A.L.; Robinson, V.G.; Waidyanatha, S.; DeVito, M.J.; Eifrid, M.A.; Granville, C.A.; Gibbs, S.T.; Blystone, C.R. Toxicokinetics of Perfluorobutane Sulfonate (PFBS), Perfluorohexane-1-Sulphonic Acid (PFHxS), and Perfluorooctane Sulfonic Acid (PFOS) in Male and Female Hsd:Sprague Dawley SD Rats after Intravenous and Gavage Administration. Toxicol. Rep. 2019, 6, 645–655. [Google Scholar] [CrossRef] [PubMed]
  82. Attanasio, R. Sex Differences in the Association between Perfluoroalkyl Acids and Liver Function in US Adolescents: Analyses of NHANES 2013-2016. Environ. Pollut. 2019, 254, 113061. [Google Scholar] [CrossRef] [PubMed]
  83. Limei, E.; Zhang, S.; Jiang, X. Association between Perfluoroalkyl Substances Exposure and the Prevalence of Nonalcoholic Fatty Liver Disease in the Different Sexes: A Study from the National Health and Nutrition Examination Survey 2005–2018. Environ. Sci. Pollut. Res. Int. 2023, 30, 44292–44303. [Google Scholar] [CrossRef]
  84. Roth, K.; Yang, Z.; Agarwal, M.; Liu, W.; Peng, Z.; Long, Z.; Birbeck, J.; Westrick, J.; Liu, W.; Petriello, M.C. Exposure to a Mixture of Legacy, Alternative, and Replacement per- and Polyfluoroalkyl Substances (PFAS) Results in Sex-Dependent Modulation of Cholesterol Metabolism and Liver Injury. Environ. Int. 2021, 157, 106843. [Google Scholar] [CrossRef] [PubMed]
  85. Al-Eryani, L.; Wahlang, B.; Falkner, K.C.; Guardiola, J.J.; Clair, H.B.; Prough, R.A.; Cave, M. Identification of Environmental Chemicals Associated with the Development of Toxicant-Associated Fatty Liver Disease in Rodents. Toxicol. Pathol. 2015, 43, 482–497. [Google Scholar] [CrossRef]
  86. Li, W.; Xiao, H.; Wu, H.; Pan, C.; Deng, K.; Xu, X.; Zhang, Y. Analysis of Environmental Chemical Mixtures and Nonalcoholic Fatty Liver Disease: NHANES 1999–2014. Environ. Pollut. 2022, 311, 119915. [Google Scholar] [CrossRef]
  87. Rajak, S.; Raza, S.; Tewari, A.; Sinha, R.A. Environmental Toxicants and NAFLD: A Neglected yet Significant Relationship. Dig. Dis. Sci. 2022, 67, 3497–3507. [Google Scholar] [CrossRef]
  88. Armstrong, L.E.; Guo, G.L. Understanding Environmental Contaminants’ Direct Effects on Non-Alcoholic Fatty Liver Disease Progression. Curr. Environ. Health Rep. 2019, 6, 95–104. [Google Scholar] [CrossRef]
  89. Sang, H.; Lee, K.-N.; Jung, C.H.; Han, K.; Koh, E.H. Association between Organochlorine Pesticides and Nonalcoholic Fatty Liver Disease in the National Health and Nutrition Examination Survey 2003–2004. Sci. Rep. 2022, 12, 11590. [Google Scholar] [CrossRef]
  90. Jellali, R.; Jacques, S.; Essaouiba, A.; Gilard, F.; Letourneur, F.; Gakière, B.; Legallais, C.; Leclerc, E. Investigation of Steatosis Profiles Induced by Pesticides Using Liver Organ-on-Chip Model and Omics Analysis. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2021, 152, 112155. [Google Scholar] [CrossRef]
  91. Li, M.; Liu, T.; Yang, T.; Zhu, J.; Zhou, Y.; Wang, M.; Wang, Q. Gut Microbiota Dysbiosis Involves in Host Non-Alcoholic Fatty Liver Disease upon Pyrethroid Pesticide Exposure. Environ. Sci. Ecotechnol. 2022, 11, 100185. [Google Scholar] [CrossRef] [PubMed]
  92. Wang, B.; Tsakiridis, E.E.; Zhang, S.; Llanos, A.; Desjardins, E.M.; Yabut, J.M.; Green, A.E.; Day, E.A.; Smith, B.K.; Lally, J.S.V.; et al. The Pesticide Chlorpyrifos Promotes Obesity by Inhibiting Diet-Induced Thermogenesis in Brown Adipose Tissue. Nat. Commun. 2021, 12, 5163. [Google Scholar] [CrossRef] [PubMed]
  93. Wasef, L.; Nassar, A.M.K.; El-Sayed, Y.S.; Samak, D.; Noreldin, A.; Elshony, N.; Saleh, H.; Elewa, Y.H.A.; Hassan, S.M.A.; Saati, A.A.; et al. The Potential Ameliorative Impacts of Cerium Oxide Nanoparticles against Fipronil-Induced Hepatic Steatosis. Sci. Rep. 2021, 11, 1310. [Google Scholar] [CrossRef]
  94. Yang, D.; Zhang, X.; Yue, L.; Hu, H.; Wei, X.; Guo, Q.; Zhang, B.; Fan, X.; Xin, Y.; Oh, Y.; et al. Thiamethoxam Induces Nonalcoholic Fatty Liver Disease in Mice via Methionine Metabolism Disturb via Nicotinamide N-Methyltransferase Overexpression. Chemosphere 2021, 273, 129727. [Google Scholar] [CrossRef]
  95. Taylor, R.; Armstrong, L.; Bhattacharya, A.; Henry, Z.; Brinker, A.; Buckley, B.; Kong, B.; Guo, G. Myclobutanil-Mediated Alteration of Liver-Gut FXR Signaling in Mice. Toxicol. Sci. Off. J. Soc. Toxicol. 2023, 191, 387–399. [Google Scholar] [CrossRef]
  96. Stellavato, A.; Lamberti, M.; Pirozzi, A.V.A.; de Novellis, F.; Schiraldi, C. Myclobutanil Worsens Nonalcoholic Fatty Liver Disease: An in Vitro Study of Toxicity and Apoptosis on HepG2 Cells. Toxicol. Lett. 2016, 262, 100–104. [Google Scholar] [CrossRef]
  97. Pirozzi, A.V.A.; Stellavato, A.; La Gatta, A.; Lamberti, M.; Schiraldi, C. Mancozeb, a Fungicide Routinely Used in Agriculture, Worsens Nonalcoholic Fatty Liver Disease in the Human HepG2 Cell Model. Toxicol. Lett. 2016, 249, 1–4. [Google Scholar] [CrossRef] [PubMed]
  98. Stossi, F.; Dandekar, R.D.; Johnson, H.; Lavere, P.; Foulds, C.E.; Mancini, M.G.; Mancini, M.A. Tributyltin Chloride (TBT) Induces RXRA down-Regulation and Lipid Accumulation in Human Liver Cells. PLoS ONE 2019, 14, e0224405. [Google Scholar] [CrossRef]
  99. González, N.; Domingo, J.L. Polychlorinated Dibenzo-p-Dioxins and Dibenzofurans (PCDD/Fs) in Food and Human Dietary Intake: An Update of the Scientific Literature. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2021, 157, 112585. [Google Scholar] [CrossRef]
  100. Shin, E.; Kim, J.; Choi, S.-D.; Kang, Y.-W.; Chang, Y.-S. Estimated Dietary Intake and Risk Assessment of Polychlorinated Dibenzo-p-Dioxins and Dibenzofurans and Dioxin-like Polychlorinated Biphenyls from Fish Consumption in the Korean General Population. Chemosphere 2016, 146, 419–425. [Google Scholar] [CrossRef]
  101. Houlahan, K.E.; Prokopec, S.D.; Sun, R.X.; Moffat, I.D.; Lindén, J.; Lensu, S.; Okey, A.B.; Pohjanvirta, R.; Boutros, P.C. Transcriptional Profiling of Rat White Adipose Tissue Response to 2,3,7,8-Tetrachlorodibenzo-ρ-Dioxin. Toxicol. Appl. Pharmacol. 2015, 288, 223–231. [Google Scholar] [CrossRef] [PubMed]
  102. Nwanaji-Enwerem, J.C.; Jenkins, T.G.; Colicino, E.; Cardenas, A.; Baccarelli, A.A.; Boyer, E.W. Serum Dioxin Levels and Sperm DNA Methylation Age: Findings in Vietnam War Veterans Exposed to Agent Orange. Reprod. Toxicol. 2020, 96, 27–35. [Google Scholar] [CrossRef]
  103. Angrish, M.M.; Dominici, C.Y.; Zacharewski, T.R. TCDD-Elicited Effects on Liver, Serum, and Adipose Lipid Composition in C57BL/6 Mice. Toxicol. Sci. Off. J. Soc. Toxicol. 2013, 131, 108–115. [Google Scholar] [CrossRef] [PubMed]
  104. Fletcher, N.; Wahlström, D.; Lundberg, R.; Nilsson, C.B.; Nilsson, K.C.; Stockling, K.; Hellmold, H.; Håkansson, H. 2,3,7,8-Tetrachlorodibenzo-p-Dioxin (TCDD) Alters the MRNA Expression of Critical Genes Associated with Cholesterol Metabolism, Bile Acid Biosynthesis, and Bile Transport in Rat Liver: A Microarray Study. Toxicol. Appl. Pharmacol. 2005, 207, 1–24. [Google Scholar] [CrossRef]
  105. Fader, K.A.; Nault, R.; Zhang, C.; Kumagai, K.; Harkema, J.R.; Zacharewski, T.R. 2,3,7,8-Tetrachlorodibenzo-p-Dioxin (TCDD)-Elicited Effects on Bile Acid Homeostasis: Alterations in Biosynthesis, Enterohepatic Circulation, and Microbial Metabolism. Sci. Rep. 2017, 7, 5921. [Google Scholar] [CrossRef]
  106. Fling, R.R.; Zacharewski, T.R. Aryl Hydrocarbon Receptor (AhR) Activation by 2,3,7,8-Tetrachlorodibenzo-p-Dioxin (TCDD) Dose-Dependently Shifts the Gut Microbiome Consistent with the Progression of Non-Alcoholic Fatty Liver Disease. Int. J. Mol. Sci. 2021, 22, 12431. [Google Scholar] [CrossRef] [PubMed]
  107. Fling, R.R.; Doskey, C.M.; Fader, K.A.; Nault, R.; Zacharewski, T.R. 2,3,7,8-Tetrachlorodibenzo-p-Dioxin (TCDD) Dysregulates Hepatic One Carbon Metabolism during the Progression of Steatosis to Steatohepatitis with Fibrosis in Mice. Sci. Rep. 2020, 10, 14831. [Google Scholar] [CrossRef]
  108. Wahlang, B.; Hardesty, J.E.; Jin, J.; Falkner, K.C.; Cave, M.C. Polychlorinated Biphenyls and Nonalcoholic Fatty Liver Disease. Curr. Opin. Toxicol. 2019, 14, 21–28. [Google Scholar] [CrossRef] [PubMed]
  109. Duval, C.; Teixeira-Clerc, F.; Leblanc, A.F.; Touch, S.; Emond, C.; Guerre-Millo, M.; Lotersztajn, S.; Barouki, R.; Aggerbeck, M.; Coumoul, X. Chronic Exposure to Low Doses of Dioxin Promotes Liver Fibrosis Development in the C57BL/6J Diet-Induced Obesity Mouse Model. Environ. Health Perspect. 2017, 125, 428–436. [Google Scholar] [CrossRef]
  110. Jin, J.; Wahlang, B.; Shi, H.; Hardesty, J.E.; Falkner, K.C.; Head, K.Z.; Srivastava, S.; Merchant, M.L.; Rai, S.N.; Cave, M.C.; et al. Dioxin-like and Non-Dioxin-like PCBs Differentially Regulate the Hepatic Proteome and Modify Diet-Induced Nonalcoholic Fatty Liver Disease Severity. Med. Chem. Res. Int. J. Rapid Commun. Des. Mech. Action Biol. Act. Agents 2020, 29, 1247–1263. [Google Scholar] [CrossRef]
  111. Wahlang, B.; Falkner, K.C.; Gregory, B.; Ansert, D.; Young, D.; Conklin, D.J.; Bhatnagar, A.; McClain, C.J.; Cave, M. Polychlorinated Biphenyl 153 Is a Diet-Dependent Obesogen That Worsens Nonalcoholic Fatty Liver Disease in Male C57BL6/J Mice. J. Nutr. Biochem. 2013, 24, 1587–1595. [Google Scholar] [CrossRef] [PubMed]
  112. Lee, J.; Prokopec, S.D.; Watson, J.D.; Sun, R.X.; Pohjanvirta, R.; Boutros, P.C. Male and Female Mice Show Significant Differences in Hepatic Transcriptomic Response to 2,3,7,8-Tetrachlorodibenzo-p-Dioxin. BMC Genomics 2015, 16, 625. [Google Scholar] [CrossRef] [PubMed]
  113. Nault, R.; Fader, K.A.; Harkema, J.R.; Zacharewski, T. Loss of Liver-Specific and Sexually Dimorphic Gene Expression by Aryl Hydrocarbon Receptor Activation in C57BL/6 Mice. PLoS ONE 2017, 12, e0184842. [Google Scholar] [CrossRef] [PubMed]
  114. Prokopec, S.D.; Watson, J.D.; Lee, J.; Pohjanvirta, R.; Boutros, P.C. Sex-Related Differences in Murine Hepatic Transcriptional and Proteomic Responses to TCDD. Toxicol. Appl. Pharmacol. 2015, 284, 188–196. [Google Scholar] [CrossRef] [PubMed]
  115. Matteo, G.; Hoyeck, M.P.; Blair, H.L.; Zebarth, J.; Rick, K.R.C.; Williams, A.; Gagné, R.; Buick, J.K.; Yauk, C.L.; Bruin, J.E. Prolonged Low-Dose Dioxin Exposure Impairs Metabolic Adaptability to High-Fat Diet Feeding in Female but Not Male Mice. Endocrinology 2021, 162, bqab050. [Google Scholar] [CrossRef] [PubMed]
  116. Vega, N.; Pinteur, C.; Buffelan, G.; Loizon, E.; Vidal, H.; Naville, D.; Le Magueresse-Battistoni, B. Exposure to Pollutants Altered Glucocorticoid Signaling and Clock Gene Expression in Female Mice. Evidence of Tissue- and Sex-Specificity. Chemosphere 2021, 262, 127841. [Google Scholar] [CrossRef]
  117. Sampaio, G.R.; Guizellini, G.M.; da Silva, S.A.; de Almeida, A.P.; Pinaffi-Langley, A.C.C.; Rogero, M.M.; de Camargo, A.C.; Torres, E.A.F.S. Polycyclic Aromatic Hydrocarbons in Foods: Biological Effects, Legislation, Occurrence, Analytical Methods, and Strategies to Reduce Their Formation. Int. J. Mol. Sci. 2021, 22, 6010. [Google Scholar] [CrossRef]
  118. Goedtke, L.; Sprenger, H.; Hofmann, U.; Schmidt, F.F.; Hammer, H.S.; Zanger, U.M.; Poetz, O.; Seidel, A.; Braeuning, A.; Hessel-Pras, S. Polycyclic Aromatic Hydrocarbons Activate the Aryl Hydrocarbon Receptor and the Constitutive Androstane Receptor to Regulate Xenobiotic Metabolism in Human Liver Cells. Int. J. Mol. Sci. 2020, 22, 372. [Google Scholar] [CrossRef]
  119. Li, F.; Xiang, B.; Jin, Y.; Li, C.; Ren, S.; Wu, Y.; Li, J.; Luo, Q. Hepatotoxic Effects of Inhalation Exposure to Polycyclic Aromatic Hydrocarbons on Lipid Metabolism of C57BL/6 Mice. Environ. Int. 2020, 134, 105000. [Google Scholar] [CrossRef]
  120. Zhu, X.-Y.; Xia, H.-G.; Wang, Z.-H.; Li, B.; Jiang, H.-Y.; Li, D.-L.; Jin, R.; Jin, Y. In Vitro and in Vivo Approaches for Identifying the Role of Aryl Hydrocarbon Receptor in the Development of Nonalcoholic Fatty Liver Disease. Toxicol. Lett. 2020, 319, 85–94. [Google Scholar] [CrossRef]
  121. Kim, K.; Melough, M.M.; Vance, T.M.; Noh, H.; Koo, S.I.; Chun, O.K. Dietary Cadmium Intake and Sources in the US. Nutrients 2018, 11, 2. [Google Scholar] [CrossRef]
  122. Zhang, X.; Wang, Z.; Liu, L.; Zhan, N.; Qin, J.; Lu, X.; Cheng, M. Assessment of the Risks from Dietary Lead Exposure in China. J. Hazard. Mater. 2021, 418, 126134. [Google Scholar] [CrossRef] [PubMed]
  123. Cinnirella, S.; Hedgecock, I.M.; Sprovieri, F. Heavy Metals in the Environment: Sources, Interactions and Human Health. Environ. Sci. Pollut. Res. Int. 2014, 21, 3997–3998. [Google Scholar] [CrossRef] [PubMed]
  124. Lin, Y.-C.; Lian, I.-B.; Kor, C.-T.; Chang, C.-C.; Su, P.-Y.; Chang, W.-T.; Liang, Y.-F.; Su, W.-W.; Soon, M.-S. Association between Soil Heavy Metals and Fatty Liver Disease in Men in Taiwan: A Cross Sectional Study. BMJ Open 2017, 7, e014215. [Google Scholar] [CrossRef] [PubMed]
  125. Sadighara, P.; Abedini, A.H.; Irshad, N.; Ghazi-Khansari, M.; Esrafili, A.; Yousefi, M. Association Between Non-Alcoholic Fatty Liver Disease and Heavy Metal Exposure: A Systematic Review. Biol. Trace Elem. Res. 2023. [Google Scholar] [CrossRef]
  126. Hong, D.; Min, J.-Y.; Min, K.-B. Association Between Cadmium Exposure and Liver Function in Adults in the United States: A Cross-Sectional Study. J. Prev. Med. Pub. Health 2021, 54, 471–480. [Google Scholar] [CrossRef]
  127. Park, E.; Kim, J.; Kim, B.; Park, E.Y. Association between Environmental Exposure to Cadmium and Risk of Suspected Non-Alcoholic Fatty Liver Disease. Chemosphere 2021, 266, 128947. [Google Scholar] [CrossRef]
  128. Li, Y.; Chen, C.; Lu, L.; Guo, W.; VanWagner, L.B.; Shikany, J.M.; Zhang, S.; Kahe, K. Cadmium Exposure in Young Adulthood Is Associated with Risk of Nonalcoholic Fatty Liver Disease in Midlife. Dig. Dis. Sci. 2022, 67, 689–696. [Google Scholar] [CrossRef]
  129. Xu, Z.; Weng, Z.; Liang, J.; Liu, Q.; Zhang, X.; Xu, J.; Xu, C.; Gu, A. Association between Urinary Cadmium Concentrations and Liver Function in Adolescents. Environ. Sci. Pollut. Res. Int. 2022, 29, 39768–39776. [Google Scholar] [CrossRef]
  130. Yang, C.; Li, Y.; Ding, R.; Xing, H.; Wang, R.; Zhang, M. Lead Exposure as a Causative Factor for Metabolic Associated Fatty Liver Disease (MAFLD) and a Lead Exposure Related Nomogram for MAFLD Prevalence. Front. Public Health 2022, 10, 1000403. [Google Scholar] [CrossRef]
  131. Nguyen, H.D.; Kim, M.-S. Cadmium, Lead, and Mercury Mixtures Interact with Non-Alcoholic Fatty Liver Diseases. Environ. Pollut. 2022, 309, 119780. [Google Scholar] [CrossRef] [PubMed]
  132. Xie, Z.; Aimuzi, R.; Si, M.; Qu, Y.; Jiang, Y. Associations of Metal Mixtures with Metabolic-Associated Fatty Liver Disease and Non-Alcoholic Fatty Liver Disease: NHANES 2003–2018. Front. Public Health 2023, 11, 1133194. [Google Scholar] [CrossRef] [PubMed]
  133. Frediani, J.K.; Naioti, E.A.; Vos, M.B.; Figueroa, J.; Marsit, C.J.; Welsh, J.A. Arsenic Exposure and Risk of Nonalcoholic Fatty Liver Disease (NAFLD) among U.S. Adolescents and Adults: An Association Modified by Race/Ethnicity, NHANES 2005-2014. Environ. Health Glob. Access Sci. Source 2018, 17, 6. [Google Scholar] [CrossRef] [PubMed]
  134. Genchi, G.; Sinicropi, M.S.; Lauria, G.; Carocci, A.; Catalano, A. The Effects of Cadmium Toxicity. Int. J. Environ. Res. Public. Health 2020, 17, 3782. [Google Scholar] [CrossRef]
  135. Zhao, M.; Ge, X.; Xu, J.; Li, A.; Mei, Y.; Yin, G.; Wu, J.; Liu, X.; Wei, L.; Xu, Q. Association between Urine Metals and Liver Function Biomarkers in Northeast China: A Cross-Sectional Study. Ecotoxicol. Environ. Saf. 2022, 231, 113163. [Google Scholar] [CrossRef]
  136. Gu, J.; Kong, A.; Guo, C.; Liu, J.; Li, K.; Ren, Z.; Zhou, Y.; Tang, M.; Shi, H. Cadmium Perturbed Lipid Profile and Induced Liver Dysfunction in Mice through Phosphatidylcholine Remodeling and Promoting Arachidonic Acid Synthesis and Metabolism. Ecotoxicol. Environ. Saf. 2022, 247, 114254. [Google Scholar] [CrossRef]
  137. Zhu, Y.; Zhao, Y.; Chai, X.-X.; Zhou, J.; Shi, M.-J.; Zhao, Y.; Tian, Y.; Wang, X.-M.; Ying, T.-X.; Feng, Q.; et al. Chronic Exposure to Low-Dose Cadmium Facilitated Nonalcoholic Steatohepatitis in Mice by Suppressing Fatty Acid Desaturation. Ecotoxicol. Environ. Saf. 2022, 233, 113306. [Google Scholar] [CrossRef]
  138. Ren, C.; Ren, L.; Yan, J.; Bai, Z.; Zhang, L.; Zhang, H.; Xie, Y.; Li, X. Transcription Profiling of Cadmium-Exposed Livers Reveals Alteration of Lipid Metabolism and Predisposition to Hepatic Steatosis. Xenobiotica Fate Foreign Compd. Biol. Syst. 2021, 51, 1271–1281. [Google Scholar] [CrossRef]
  139. Young, J.L.; Cave, M.C.; Xu, Q.; Kong, M.; Xu, J.; Lin, Q.; Tan, Y.; Cai, L. Whole Life Exposure to Low Dose Cadmium Alters Diet-Induced NAFLD. Toxicol. Appl. Pharmacol. 2022, 436, 115855. [Google Scholar] [CrossRef]
  140. He, X.; Gao, J.; Hou, H.; Qi, Z.; Chen, H.; Zhang, X.-X. Inhibition of Mitochondrial Fatty Acid Oxidation Contributes to Development of Nonalcoholic Fatty Liver Disease Induced by Environmental Cadmium Exposure. Environ. Sci. Technol. 2019, 53, 13992–14000. [Google Scholar] [CrossRef]
  141. Obeng-Gyasi, E. Sources of Lead Exposure in Various Countries. Rev. Environ. Health 2019, 34, 25–34. [Google Scholar] [CrossRef]
  142. Eslam, M.; Newsome, P.N.; Sarin, S.K.; Anstee, Q.M.; Targher, G.; Romero-Gomez, M.; Zelber-Sagi, S.; Wai-Sun Wong, V.; Dufour, J.-F.; Schattenberg, J.M.; et al. A New Definition for Metabolic Dysfunction-Associated Fatty Liver Disease: An International Expert Consensus Statement. J. Hepatol. 2020, 73, 202–209. [Google Scholar] [CrossRef] [PubMed]
  143. Cave, M.; Appana, S.; Patel, M.; Falkner, K.C.; McClain, C.J.; Brock, G. Polychlorinated Biphenyls, Lead, and Mercury Are Associated with Liver Disease in American Adults: NHANES 2003–2004. Environ. Health Perspect. 2010, 118, 1735–1742. [Google Scholar] [CrossRef]
  144. Betanzos-Robledo, L.; Cantoral, A.; Peterson, K.E.; Hu, H.; Hernández-Ávila, M.; Perng, W.; Jansen, E.; Ettinger, A.S.; Mercado-García, A.; Solano-González, M.; et al. Association between Cumulative Childhood Blood Lead Exposure and Hepatic Steatosis in Young Mexican Adults. Environ. Res. 2021, 196, 110980. [Google Scholar] [CrossRef] [PubMed]
  145. Vineeth Daniel, P.; Kamthan, M.; Gera, R.; Dogra, S.; Gautam, K.; Ghosh, D.; Mondal, P. Chronic Exposure to Pb2+ Perturbs ChREBP Transactivation and Coerces Hepatic Dyslipidemia. FEBS Lett. 2019, 593, 3084–3097. [Google Scholar] [CrossRef]
  146. Milosevic, N.; Maier, P. Lead Stimulates Intercellular Signalling between Hepatocytes and Kupffer Cells. Eur. J. Pharmacol. 2000, 401, 317–328. [Google Scholar] [CrossRef] [PubMed]
  147. Wan, H.; Wang, Y.; Zhang, H.; Zhang, K.; Chen, Y.; Chen, C.; Zhang, W.; Xia, F.; Wang, N.; Lu, Y. Chronic Lead Exposure Induces Fatty Liver Disease Associated with the Variations of Gut Microbiota. Ecotoxicol. Environ. Saf. 2022, 232, 113257. [Google Scholar] [CrossRef]
  148. Shi, X.; Wei, X.; Koo, I.; Schmidt, R.H.; Yin, X.; Kim, S.H.; Vaughn, A.; McClain, C.J.; Arteel, G.E.; Zhang, X.; et al. Metabolomic Analysis of the Effects of Chronic Arsenic Exposure in a Mouse Model of Diet-Induced Fatty Liver Disease. J. Proteome Res. 2014, 13, 547–554. [Google Scholar] [CrossRef]
  149. Jia, X.; Qiu, T.; Yao, X.; Jiang, L.; Wang, N.; Wei, S.; Tao, Y.; Pei, P.; Wang, Z.; Zhang, J.; et al. Arsenic Induces Hepatic Insulin Resistance via MtROS-NLRP3 Inflammasome Pathway. J. Hazard. Mater. 2020, 399, 123034. [Google Scholar] [CrossRef]
  150. Qiu, T.; Pei, P.; Yao, X.; Jiang, L.; Wei, S.; Wang, Z.; Bai, J.; Yang, G.; Gao, N.; Yang, L.; et al. Taurine Attenuates Arsenic-Induced Pyroptosis and Nonalcoholic Steatohepatitis by Inhibiting the Autophagic-Inflammasomal Pathway. Cell Death Dis. 2018, 9, 946. [Google Scholar] [CrossRef]
  151. Wei, S.; Qiu, T.; Wang, N.; Yao, X.; Jiang, L.; Jia, X.; Tao, Y.; Zhang, J.; Zhu, Y.; Yang, G.; et al. Ferroptosis Mediated by the Interaction between Mfn2 and IREα Promotes Arsenic-Induced Nonalcoholic Steatohepatitis. Environ. Res. 2020, 188, 109824. [Google Scholar] [CrossRef] [PubMed]
  152. Chen, H.; Zhu, C.; Zhou, X. Effects of Lead and Cadmium Combined Heavy Metals on Liver Function and Lipid Metabolism in Mice. Biol. Trace Elem. Res. 2023, 201, 2864–2876. [Google Scholar] [CrossRef]
  153. Campanale, C.; Massarelli, C.; Savino, I.; Locaputo, V.; Uricchio, V.F. A Detailed Review Study on Potential Effects of Microplastics and Additives of Concern on Human Health. Int. J. Environ. Res. Public. Health 2020, 17, 1212. [Google Scholar] [CrossRef]
  154. Frias, J.P.G.L.; Nash, R. Microplastics: Finding a Consensus on the Definition. Mar. Pollut. Bull. 2019, 138, 145–147. [Google Scholar] [CrossRef] [PubMed]
  155. Yee, M.S.-L.; Hii, L.-W.; Looi, C.K.; Lim, W.-M.; Wong, S.-F.; Kok, Y.-Y.; Tan, B.-K.; Wong, C.-Y.; Leong, C.-O. Impact of Microplastics and Nanoplastics on Human Health. Nanomaterials 2021, 11, 496. [Google Scholar] [CrossRef] [PubMed]
  156. Karbalaei, S.; Hanachi, P.; Walker, T.R.; Cole, M. Occurrence, Sources, Human Health Impacts and Mitigation of Microplastic Pollution. Environ. Sci. Pollut. Res. Int. 2018, 25, 36046–36063. [Google Scholar] [CrossRef]
  157. Yin, J.; Ju, Y.; Qian, H.; Wang, J.; Miao, X.; Zhu, Y.; Zhou, L.; Ye, L. Nanoplastics and Microplastics May Be Damaging Our Livers. Toxics 2022, 10, 586. [Google Scholar] [CrossRef]
  158. Prata, J.C.; da Costa, J.P.; Lopes, I.; Duarte, A.C.; Rocha-Santos, T. Environmental Exposure to Microplastics: An Overview on Possible Human Health Effects. Sci. Total Environ. 2020, 702, 134455. [Google Scholar] [CrossRef]
  159. Prata, J.C. Airborne Microplastics: Consequences to Human Health? Environ. Pollut. 2018, 234, 115–126. [Google Scholar] [CrossRef]
  160. De-la-Torre, G.E. Microplastics: An Emerging Threat to Food Security and Human Health. J. Food Sci. Technol. 2020, 57, 1601–1608. [Google Scholar] [CrossRef]
  161. Schwabl, P.; Köppel, S.; Königshofer, P.; Bucsics, T.; Trauner, M.; Reiberger, T.; Liebmann, B. Detection of Various Microplastics in Human Stool: A Prospective Case Series. Ann. Intern. Med. 2019, 171, 453–457. [Google Scholar] [CrossRef]
  162. Horvatits, T.; Tamminga, M.; Liu, B.; Sebode, M.; Carambia, A.; Fischer, L.; Püschel, K.; Huber, S.; Fischer, E.K. Microplastics Detected in Cirrhotic Liver Tissue. EBioMedicine 2022, 82, 104147. [Google Scholar] [CrossRef] [PubMed]
  163. Auguet, T.; Bertran, L.; Barrientos-Riosalido, A.; Fabregat, B.; Villar, B.; Aguilar, C.; Sabench, F. Are Ingested or Inhaled Microplastics Involved in Nonalcoholic Fatty Liver Disease? Int. J. Environ. Res. Public. Health 2022, 19, 13495. [Google Scholar] [CrossRef] [PubMed]
  164. Okamura, T.; Hamaguchi, M.; Hasegawa, Y.; Hashimoto, Y.; Majima, S.; Senmaru, T.; Ushigome, E.; Nakanishi, N.; Asano, M.; Yamazaki, M.; et al. Oral Exposure to Polystyrene Microplastics of Mice on a Normal or High-Fat Diet and Intestinal and Metabolic Outcomes. Environ. Health Perspect. 2023, 131, 027006. [Google Scholar] [CrossRef] [PubMed]
  165. Li, L.; Xu, M.; He, C.; Wang, H.; Hu, Q. Polystyrene Nanoplastics Potentiate the Development of Hepatic Fibrosis in High Fat Diet Fed Mice. Environ. Toxicol. 2022, 37, 362–372. [Google Scholar] [CrossRef]
  166. Fan, X.; Wei, X.; Hu, H.; Zhang, B.; Yang, D.; Du, H.; Zhu, R.; Sun, X.; Oh, Y.; Gu, N. Effects of Oral Administration of Polystyrene Nanoplastics on Plasma Glucose Metabolism in Mice. Chemosphere 2022, 288, 132607. [Google Scholar] [CrossRef]
  167. Wang, H.; Shi, X.; Gao, Y.; Zhang, X.; Zhao, H.; Wang, L.; Zhang, X.; Chen, R. Polystyrene Nanoplastics Induce Profound Metabolic Shift in Human Cells as Revealed by Integrated Proteomic and Metabolomic Analysis. Environ. Int. 2022, 166, 107349. [Google Scholar] [CrossRef]
  168. He, Y.; Li, J.; Chen, J.; Miao, X.; Li, G.; He, Q.; Xu, H.; Li, H.; Wei, Y. Cytotoxic Effects of Polystyrene Nanoplastics with Different Surface Functionalization on Human HepG2 Cells. Sci. Total Environ. 2020, 723, 138180. [Google Scholar] [CrossRef]
  169. Lin, S.; Zhang, H.; Wang, C.; Su, X.-L.; Song, Y.; Wu, P.; Yang, Z.; Wong, M.-H.; Cai, Z.; Zheng, C. Metabolomics Reveal Nanoplastic-Induced Mitochondrial Damage in Human Liver and Lung Cells. Environ. Sci. Technol. 2022, 56, 12483–12493. [Google Scholar] [CrossRef]
  170. Cheng, W.; Li, X.; Zhou, Y.; Yu, H.; Xie, Y.; Guo, H.; Wang, H.; Li, Y.; Feng, Y.; Wang, Y. Polystyrene Microplastics Induce Hepatotoxicity and Disrupt Lipid Metabolism in the Liver Organoids. Sci. Total Environ. 2022, 806, 150328. [Google Scholar] [CrossRef]
  171. Lai, W.; Xu, D.; Li, J.; Wang, Z.; Ding, Y.; Wang, X.; Li, X.; Xu, N.; Mai, K.; Ai, Q. Dietary Polystyrene Nanoplastics Exposure Alters Liver Lipid Metabolism and Muscle Nutritional Quality in Carnivorous Marine Fish Large Yellow Croaker (Larimichthys Crocea). J. Hazard. Mater. 2021, 419, 126454. [Google Scholar] [CrossRef] [PubMed]
  172. Deng, Y.; Zhang, Y.; Lemos, B.; Ren, H. Tissue Accumulation of Microplastics in Mice and Biomarker Responses Suggest Widespread Health Risks of Exposure. Sci. Rep. 2017, 7, 46687. [Google Scholar] [CrossRef] [PubMed]
  173. Mu, Y.; Sun, J.; Li, Z.; Zhang, W.; Liu, Z.; Li, C.; Peng, C.; Cui, G.; Shao, H.; Du, Z. Activation of Pyroptosis and Ferroptosis Is Involved in the Hepatotoxicity Induced by Polystyrene Microplastics in Mice. Chemosphere 2022, 291, 132944. [Google Scholar] [CrossRef]
  174. Chen, L.; Qi, M.; Zhang, L.; Yu, F.; Tao, D.; Xu, C.; Xu, S. Di(2-Ethylhexyl) Phthalate and Microplastics Cause Necroptosis and Apoptosis in Hepatocytes of Mice by Inducing Oxidative Stress. Environ. Toxicol. 2023. [Google Scholar] [CrossRef]
  175. Wang, F.; Zhang, Q.; Cui, J.; Bao, B.; Deng, X.; Liu, L.; Guo, M.-Y. Polystyrene Microplastics Induce Endoplasmic Reticulum Stress, Apoptosis and Inflammation by Disrupting the Gut Microbiota in Carp Intestines. Environ. Pollut. 2023, 323, 121233. [Google Scholar] [CrossRef] [PubMed]
  176. Perl, A.; Hanczko, R.; Telarico, T.; Oaks, Z.; Landas, S. Oxidative Stress, Inflammation and Carcinogenesis Are Controlled through the Pentose Phosphate Pathway by Transaldolase. Trends Mol. Med. 2011, 17, 395–403. [Google Scholar] [CrossRef]
  177. Lu, L.; Wan, Z.; Luo, T.; Fu, Z.; Jin, Y. Polystyrene Microplastics Induce Gut Microbiota Dysbiosis and Hepatic Lipid Metabolism Disorder in Mice. Sci. Total Environ. 2018, 631–632, 449–458. [Google Scholar] [CrossRef]
  178. Chen, X.; Zhuang, J.; Chen, Q.; Xu, L.; Yue, X.; Qiao, D. Chronic Exposure to Polyvinyl Chloride Microplastics Induces Liver Injury and Gut Microbiota Dysbiosis Based on the Integration of Liver Transcriptome Profiles and Full-Length 16S RRNA Sequencing Data. Sci. Total Environ. 2022, 839, 155984. [Google Scholar] [CrossRef]
  179. Huang, D.; Zhang, Y.; Long, J.; Yang, X.; Bao, L.; Yang, Z.; Wu, B.; Si, R.; Zhao, W.; Peng, C.; et al. Polystyrene Microplastic Exposure Induces Insulin Resistance in Mice via Dysbacteriosis and Pro-Inflammation. Sci. Total Environ. 2022, 838, 155937. [Google Scholar] [CrossRef]
  180. Yang, X.; Jiang, J.; Wang, Q.; Duan, J.; Chen, N.; Wu, D.; Xia, Y. Gender Difference in Hepatic AMPK Pathway Activated Lipid Metabolism Induced by Aged Polystyrene Microplastics Exposure. Ecotoxicol. Environ. Saf. 2022, 245, 114105. [Google Scholar] [CrossRef]
  181. Saeed, A.; Akhtar, M.F.; Saleem, A.; Akhtar, B.; Sharif, A. Reproductive and Metabolic Toxic Effects of Polystyrene Microplastics in Adult Female Wistar Rats: A Mechanistic Study. Environ. Sci. Pollut. Res. Int. 2023, 30, 63185–63199. [Google Scholar] [CrossRef] [PubMed]
  182. Liu, Z.; Zhuan, Q.; Zhang, L.; Meng, L.; Fu, X.; Hou, Y. Polystyrene Microplastics Induced Female Reproductive Toxicity in Mice. J. Hazard. Mater. 2022, 424, 127629. [Google Scholar] [CrossRef]
  183. Wang, J.; Li, Y.; Lu, L.; Zheng, M.; Zhang, X.; Tian, H.; Wang, W.; Ru, S. Polystyrene Microplastics Cause Tissue Damages, Sex-Specific Reproductive Disruption and Transgenerational Effects in Marine Medaka (Oryzias Melastigma). Environ. Pollut. 2019, 254, 113024. [Google Scholar] [CrossRef] [PubMed]
  184. Wei, Z.; Wang, Y.; Wang, S.; Xie, J.; Han, Q.; Chen, M. Comparing the Effects of Polystyrene Microplastics Exposure on Reproduction and Fertility in Male and Female Mice. Toxicology 2022, 465, 153059. [Google Scholar] [CrossRef]
  185. Thangavel, P.; Park, D.; Lee, Y.-C. Recent Insights into Particulate Matter (PM2.5)-Mediated Toxicity in Humans: An Overview. Int. J. Environ. Res. Public. Health 2022, 19, 7511. [Google Scholar] [CrossRef]
  186. Chen, H.; Oliver, B.G.; Pant, A.; Olivera, A.; Poronnik, P.; Pollock, C.A.; Saad, S. Effects of Air Pollution on Human Health—Mechanistic Evidence Suggested by in Vitro and in Vivo Modelling. Environ. Res. 2022, 212, 113378. [Google Scholar] [CrossRef] [PubMed]
  187. Manisalidis, I.; Stavropoulou, E.; Stavropoulos, A.; Bezirtzoglou, E. Environmental and Health Impacts of Air Pollution: A Review. Front. Public Health 2020, 8, 14. [Google Scholar] [CrossRef]
  188. Guo, B.; Guo, Y.; Nima, Q.; Feng, Y.; Wang, Z.; Lu, R.; Baimayangji; Ma, Y.; Zhou, J.; Xu, H.; et al. Exposure to Air Pollution Is Associated with an Increased Risk of Metabolic Dysfunction-Associated Fatty Liver Disease. J. Hepatol. 2022, 76, 518–525. [Google Scholar] [CrossRef] [PubMed]
  189. Sun, S.; Yang, Q.; Zhou, Q.; Cao, W.; Yu, S.; Zhan, S.; Sun, F. Long-Term Exposure to Fine Particulate Matter and Non-Alcoholic Fatty Liver Disease: A Prospective Cohort Study. Gut 2022, 71, 443–445. [Google Scholar] [CrossRef]
  190. Li, F.-R.; Liao, J.; Zhu, B.; Li, X.; Cheng, Z.; Jin, C.; Mo, C.; Wu, X.; Li, Q.; Liang, F. Long-Term Exposure to Air Pollution and Incident Non-Alcoholic Fatty Liver Disease and Cirrhosis: A Cohort Study. Liver Int. Off. J. Int. Assoc. Study Liver 2023, 43, 299–307. [Google Scholar] [CrossRef]
  191. Chen, J.; Wu, L.; Yang, G.; Zhang, C.; Liu, X.; Sun, X.; Chen, X.; Wang, N. The Influence of PM2.5 Exposure on Non-Alcoholic Fatty Liver Disease. Life Sci. 2021, 270, 119135. [Google Scholar] [CrossRef] [PubMed]
  192. Xu, M.-X.; Ge, C.-X.; Qin, Y.-T.; Gu, T.-T.; Lou, D.-S.; Li, Q.; Hu, L.-F.; Feng, J.; Huang, P.; Tan, J. Prolonged PM2.5 Exposure Elevates Risk of Oxidative Stress-Driven Nonalcoholic Fatty Liver Disease by Triggering Increase of Dyslipidemia. Free Radic. Biol. Med. 2019, 130, 542–556. [Google Scholar] [CrossRef] [PubMed]
  193. Ding, S.; Yuan, C.; Si, B.; Wang, M.; Da, S.; Bai, L.; Wu, W. Combined Effects of Ambient Particulate Matter Exposure and a High-Fat Diet on Oxidative Stress and Steatohepatitis in Mice. PLoS ONE 2019, 14, e0214680. [Google Scholar] [CrossRef] [PubMed]
  194. Xu, Z.; Shi, L.; Li, D.; Wu, Q.; Zhang, Y.; Gao, M.; Ji, A.; Jiang, Q.; Chen, R.; Zhang, R.; et al. Real Ambient Particulate Matter-Induced Lipid Metabolism Disorder: Roles of Peroxisome Proliferators-Activated Receptor Alpha. Ecotoxicol. Environ. Saf. 2022, 231, 113173. [Google Scholar] [CrossRef]
  195. Reyes-Caballero, H.; Rao, X.; Sun, Q.; Warmoes, M.O.; Lin, P.; Sussan, T.E.; Park, B.; Fan, T.W.-M.; Maiseyeu, A.; Rajagopalan, S.; et al. Air Pollution-Derived Particulate Matter Dysregulates Hepatic Krebs Cycle, Glucose and Lipid Metabolism in Mice. Sci. Rep. 2019, 9, 17423. [Google Scholar] [CrossRef] [PubMed]
  196. Ogino, N.; Miyagawa, K.; Nagaoka, K.; Sumida, K.; Kusanaga, M.; Oe, S.; Honma, Y.; Shibata, M.; Harada, M.; Suganuma, N.; et al. Airborne Fine Particulate Matter in Japan Induces Lipid Synthesis and Inhibits Autophagy in HepG2 Cells. Int. J. Biochem. Cell Biol. 2021, 141, 106099. [Google Scholar] [CrossRef] [PubMed]
  197. Tan, H.-H.; Fiel, M.I.; Sun, Q.; Guo, J.; Gordon, R.E.; Chen, L.-C.; Friedman, S.L.; Odin, J.A.; Allina, J. Kupffer Cell Activation by Ambient Air Particulate Matter Exposure May Exacerbate Non-Alcoholic Fatty Liver Disease. J. Immunotoxicol. 2009, 6, 266–275. [Google Scholar] [CrossRef]
  198. Xu, X.; Liu, C.; Xu, Z.; Tzan, K.; Zhong, M.; Wang, A.; Lippmann, M.; Chen, L.-C.; Rajagopalan, S.; Sun, Q. Long-Term Exposure to Ambient Fine Particulate Pollution Induces Insulin Resistance and Mitochondrial Alteration in Adipose Tissue. Toxicol. Sci. Off. J. Soc. Toxicol. 2011, 124, 88–98. [Google Scholar] [CrossRef]
  199. Long, M.-H.; Zhang, C.; Xu, D.-Q.; Fu, W.-L.; Gan, X.-D.; Li, F.; Wang, Q.; Xia, W.; Xu, D.-G. PM2.5 Aggravates Diabetes via the Systemically Activated IL-6-Mediated STAT3/SOCS3 Pathway in Rats’ Liver. Environ. Pollut. 2020, 256, 113342. [Google Scholar] [CrossRef]
  200. Pan, X.; Yu, Q.; Chen, S.; Li, Y.; Jiao, T.; Li, W.; Zhang, C.; Kureshi, A.; Cheng, L.; Xu, Q. Dissecting Contributions of Representative Heavy Metal Components in PM2.5 to Its Cytotoxicity. Ecotoxicol. Environ. Saf. 2023, 251, 114562. [Google Scholar] [CrossRef]
  201. Xu, J.; Zhang, W.; Lu, Z.; Zhang, F.; Ding, W. Airborne PM2.5-Induced Hepatic Insulin Resistance by Nrf2/JNK-Mediated Signaling Pathway. Int. J. Environ. Res. Public. Health 2017, 14, 787. [Google Scholar] [CrossRef] [PubMed]
  202. Laing, S.; Wang, G.; Briazova, T.; Zhang, C.; Wang, A.; Zheng, Z.; Gow, A.; Chen, A.F.; Rajagopalan, S.; Chen, L.C.; et al. Airborne Particulate Matter Selectively Activates Endoplasmic Reticulum Stress Response in the Lung and Liver Tissues. Am. J. Physiol. Cell Physiol. 2010, 299, C736–C749. [Google Scholar] [CrossRef] [PubMed]
  203. Flessa, C.-M.; Kyrou, I.; Nasiri-Ansari, N.; Kaltsas, G.; Papavassiliou, A.G.; Kassi, E.; Randeva, H.S. Endoplasmic Reticulum Stress and Autophagy in the Pathogenesis of Non-Alcoholic Fatty Liver Disease (NAFLD): Current Evidence and Perspectives. Curr. Obes. Rep. 2021, 10, 134–161. [Google Scholar] [CrossRef] [PubMed]
  204. Kish, L.; Hotte, N.; Kaplan, G.G.; Vincent, R.; Tso, R.; Gänzle, M.; Rioux, K.P.; Thiesen, A.; Barkema, H.W.; Wine, E.; et al. Environmental Particulate Matter Induces Murine Intestinal Inflammatory Responses and Alters the Gut Microbiome. PLoS ONE 2013, 8, e62220. [Google Scholar] [CrossRef]
  205. Mutlu, E.A.; Engen, P.A.; Soberanes, S.; Urich, D.; Forsyth, C.B.; Nigdelioglu, R.; Chiarella, S.E.; Radigan, K.A.; Gonzalez, A.; Jakate, S.; et al. Particulate Matter Air Pollution Causes Oxidant-Mediated Increase in Gut Permeability in Mice. Part. Fibre Toxicol. 2011, 8, 19. [Google Scholar] [CrossRef] [PubMed]
  206. Ferro, D.; Baratta, F.; Pastori, D.; Cocomello, N.; Colantoni, A.; Angelico, F.; Del Ben, M. New Insights into the Pathogenesis of Non-Alcoholic Fatty Liver Disease: Gut-Derived Lipopolysaccharides and Oxidative Stress. Nutrients 2020, 12, 2762. [Google Scholar] [CrossRef]
  207. Leung, C.; Rivera, L.; Furness, J.B.; Angus, P.W. The Role of the Gut Microbiota in NAFLD. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 412–425. [Google Scholar] [CrossRef]
  208. Safari, Z.; Gérard, P. The Links between the Gut Microbiome and Non-Alcoholic Fatty Liver Disease (NAFLD). Cell. Mol. Life Sci. CMLS 2019, 76, 1541–1558. [Google Scholar] [CrossRef]
  209. Li, R.; Sun, Q.; Lam, S.M.; Chen, R.; Zhu, J.; Gu, W.; Zhang, L.; Tian, H.; Zhang, K.; Chen, L.-C.; et al. Sex-Dependent Effects of Ambient PM2.5 Pollution on Insulin Sensitivity and Hepatic Lipid Metabolism in Mice. Part. Fibre Toxicol. 2020, 17, 14. [Google Scholar] [CrossRef]
  210. Goettems-Fiorin, P.B.; Costa-Beber, L.C.; Dos Santos, J.B.; Friske, P.T.; Sulzbacher, L.M.; Frizzo, M.N.; Ludwig, M.S.; Rhoden, C.R.; Heck, T.G. Ovariectomy Predisposes Female Rats to Fine Particulate Matter Exposure’s Effects by Altering Metabolic, Oxidative, pro-Inflammatory, and Heat-Shock Protein Levels. Environ. Sci. Pollut. Res. Int. 2019, 26, 20581–20594. [Google Scholar] [CrossRef]
  211. Donnelly, M.C.; Stableforth, W.; Krag, A.; Reuben, A. The Negative Bidirectional Interaction between Climate Change and the Prevalence and Care of Liver Disease: A Joint BSG, BASL, EASL, and AASLD Commentary. J. Hepatol. 2022, 76, 995–1000. [Google Scholar] [CrossRef] [PubMed]
  212. Hadley, K.; Wheat, S.; Rogers, H.H.; Balakumar, A.; Gonzales-Pacheco, D.; Davis, S.S.; Linstadt, H.; Cushing, T.; Ziska, L.H.; Piper, C.; et al. Mechanisms Underlying Food Insecurity in the Aftermath of Climate-Related Shocks: A Systematic Review. Lancet Planet. Health 2023, 7, e242–e250. [Google Scholar] [CrossRef] [PubMed]
  213. Fanzo, J.C.; Downs, S.M. Climate Change and Nutrition-Associated Diseases. Nat. Rev. Dis. Primer 2021, 7, 90. [Google Scholar] [CrossRef] [PubMed]
  214. Koch, C.A.; Sharda, P.; Patel, J.; Gubbi, S.; Bansal, R.; Bartel, M.J. Climate Change and Obesity. Horm. Metab. Res. 2021, 53, 575–587. [Google Scholar] [CrossRef]
  215. Krishnan, A.; Mou, X. A Brief Review of the Structure, Cytotoxicity, Synthesis, and Biodegradation of Microcystins. Water 2021, 13, 2147. [Google Scholar] [CrossRef]
  216. Chorus, I.; Fastner, J.; Welker, M. Cyanobacteria and Cyanotoxins in a Changing Environment: Concepts, Controversies, Challenges. Water 2021, 13, 2463. [Google Scholar] [CrossRef]
  217. Bui, T.; Dao, T.-S.; Vo, T.-G.; Lürling, M. Warming Affects Growth Rates and Microcystin Production in Tropical Bloom-Forming Microcystis Strains. Toxins 2018, 10, 123. [Google Scholar] [CrossRef]
  218. Lad, A.; Breidenbach, J.D.; Su, R.C.; Murray, J.; Kuang, R.; Mascarenhas, A.; Najjar, J.; Patel, S.; Hegde, P.; Youssef, M.; et al. As We Drink and Breathe: Adverse Health Effects of Microcystins and Other Harmful Algal Bloom Toxins in the Liver, Gut, Lungs and Beyond. Life 2022, 12, 418. [Google Scholar] [CrossRef]
  219. Arman, T.; Clarke, J. Microcystin Toxicokinetics, Molecular Toxicology, and Pathophysiology in Preclinical Rodent Models and Humans. Toxins 2021, 13, 537. [Google Scholar] [CrossRef]
  220. Ma, Y.; Liu, H.; Du, X.; Shi, Z.; Liu, X.; Wang, R.; Zhang, S.; Tian, Z.; Shi, L.; Guo, H.; et al. Advances in the Toxicology Research of Microcystins Based on Omics Approaches. Environ. Int. 2021, 154, 106661. [Google Scholar] [CrossRef]
  221. Zhao, Y.; Yan, Y.; Xie, L.; Wang, L.; He, Y.; Wan, X.; Xue, Q. Long-Term Environmental Exposure to Microcystins Increases the Risk of Nonalcoholic Fatty Liver Disease in Humans: A Combined Fisher-Based Investigation and Murine Model Study. Environ. Int. 2020, 138, 105648. [Google Scholar] [CrossRef]
  222. Lad, A.; Su, R.; Breidenbach, J.; Stemmer, P.; Carruthers, N.; Sanchez, N.; Khalaf, F.; Zhang, S.; Kleinhenz, A.; Dube, P.; et al. Chronic Low Dose Oral Exposure to Microcystin-LR Exacerbates Hepatic Injury in a Murine Model of Non-Alcoholic Fatty Liver Disease. Toxins 2019, 11, 486. [Google Scholar] [CrossRef] [PubMed]
  223. He, J.; Li, G.; Chen, J.; Lin, J.; Zeng, C.; Chen, J.; Deng, J.; Xie, P. Prolonged Exposure to Low-Dose Microcystin Induces Nonalcoholic Steatohepatitis in Mice: A Systems Toxicology Study. Arch. Toxicol. 2017, 91, 465–480. [Google Scholar] [CrossRef] [PubMed]
  224. Arman, T.; Lynch, K.D.; Montonye, M.L.; Goedken, M.; Clarke, J.D. Sub-Chronic Microcystin-LR Liver Toxicity in Preexisting Diet-Induced Nonalcoholic Steatohepatitis in Rats. Toxins 2019, 11, 398. [Google Scholar] [CrossRef] [PubMed]
  225. Zhang, Z.; Zhang, X.-X.; Wu, B.; Yin, J.; Yu, Y.; Yang, L. Comprehensive Insights into Microcystin-LR Effects on Hepatic Lipid Metabolism Using Cross-Omics Technologies. J. Hazard. Mater. 2016, 315, 126–134. [Google Scholar] [CrossRef]
  226. Zheng, S.; Yang, Y.; Wen, C.; Liu, W.; Cao, L.; Feng, X.; Chen, J.; Wang, H.; Tang, Y.; Tian, L.; et al. Effects of Environmental Contaminants in Water Resources on Nonalcoholic Fatty Liver Disease. Environ. Int. 2021, 154, 106555. [Google Scholar] [CrossRef]
  227. Clarke, J.D.; Dzierlenga, A.; Arman, T.; Toth, E.; Li, H.; Lynch, K.D.; Tian, D.-D.; Goedken, M.; Paine, M.F.; Cherrington, N. Nonalcoholic Fatty Liver Disease Alters Microcystin-LR Toxicokinetics and Acute Toxicity. Toxicon 2019, 162, 1–8. [Google Scholar] [CrossRef]
  228. Mrdjen, I.; Morse, M.; Ruch, R.; Knobloch, T.; Choudhary, S.; Weghorst, C.; Lee, J. Impact of Microcystin-LR on Liver Function Varies by Dose and Sex in Mice. Toxins 2018, 10, 435. [Google Scholar] [CrossRef]
  229. Symonds, M.E.; Farhat, G.; Aldiss, P.; Pope, M.; Budge, H. Brown Adipose Tissue and Glucose Homeostasis—The Link between Climate Change and the Global Rise in Obesity and Diabetes. Adipocyte 2019, 8, 46–50. [Google Scholar] [CrossRef]
  230. Turner, J.B.; Kumar, A.; Koch, C.A. The Effects of Indoor and Outdoor Temperature on Metabolic Rate and Adipose Tissue—The Mississippi Perspective on the Obesity Epidemic. Rev. Endocr. Metab. Disord. 2016, 17, 61–71. [Google Scholar] [CrossRef]
  231. Hankir, M.K.; Klingenspor, M. Brown Adipocyte Glucose Metabolism: A Heated Subject. EMBO Rep. 2018, 19, e46404. [Google Scholar] [CrossRef]
  232. Wang, Z.; Wang, Q.A.; Liu, Y.; Jiang, L. Energy Metabolism in Brown Adipose Tissue. FEBS J. 2021, 288, 3647–3662. [Google Scholar] [CrossRef] [PubMed]
  233. Wibmer, A.G.; Becher, T.; Eljalby, M.; Crane, A.; Andrieu, P.C.; Jiang, C.S.; Vaughan, R.; Schöder, H.; Cohen, P. Brown Adipose Tissue Is Associated with Healthier Body Fat Distribution and Metabolic Benefits Independent of Regional Adiposity. Cell Rep. Med. 2021, 2, 100332. [Google Scholar] [CrossRef] [PubMed]
  234. Cheng, L.; Wang, J.; Dai, H.; Duan, Y.; An, Y.; Shi, L.; Lv, Y.; Li, H.; Wang, C.; Ma, Q.; et al. Brown and Beige Adipose Tissue: A Novel Therapeutic Strategy for Obesity and Type 2 Diabetes Mellitus. Adipocyte 2021, 10, 48–65. [Google Scholar] [CrossRef] [PubMed]
  235. Pace, N.P.; Vassallo, J.; Calleja-Agius, J. Gestational Diabetes, Environmental Temperature and Climate Factors—From Epidemiological Evidence to Physiological Mechanisms. Early Hum. Dev. 2021, 155, 105219. [Google Scholar] [CrossRef]
  236. Greenhill, C. Low Brown Adipose Tissue Activity Linked to NAFLD. Nat. Rev. Endocrinol. 2021, 17, 707. [Google Scholar] [CrossRef]
  237. Ahmed, B.A.; Ong, F.J.; Barra, N.G.; Blondin, D.P.; Gunn, E.; Oreskovich, S.M.; Szamosi, J.C.; Syed, S.A.; Hutchings, E.K.; Konyer, N.B.; et al. Lower Brown Adipose Tissue Activity Is Associated with Non-Alcoholic Fatty Liver Disease but Not Changes in the Gut Microbiota. Cell Rep. Med. 2021, 2, 100397. [Google Scholar] [CrossRef]
  238. Scheele, C.; Wolfrum, C. Brown Adipose Crosstalk in Tissue Plasticity and Human Metabolism. Endocr. Rev. 2020, 41, 53–65. [Google Scholar] [CrossRef]
  239. Gavaldà-Navarro, A.; Villarroya, J.; Cereijo, R.; Giralt, M.; Villarroya, F. The Endocrine Role of Brown Adipose Tissue: An Update on Actors and Actions. Rev. Endocr. Metab. Disord. 2022, 23, 31–41. [Google Scholar] [CrossRef]
  240. Keuper, M.; Jastroch, M. The Good and the BAT of Metabolic Sex Differences in Thermogenic Human Adipose Tissue. Mol. Cell. Endocrinol. 2021, 533, 111337. [Google Scholar] [CrossRef]
  241. Kaikaew, K.; Grefhorst, A.; Visser, J.A. Sex Differences in Brown Adipose Tissue Function: Sex Hormones, Glucocorticoids, and Their Crosstalk. Front. Endocrinol. 2021, 12, 652444. [Google Scholar] [CrossRef] [PubMed]
  242. Herz, C.T.; Kulterer, O.C.; Prager, M.; Marculescu, R.; Langer, F.B.; Prager, G.; Kautzky-Willer, A.; Haug, A.R.; Kiefer, F.W. Sex Differences in Brown Adipose Tissue Activity and Cold-Induced Thermogenesis. Mol. Cell. Endocrinol. 2021, 534, 111365. [Google Scholar] [CrossRef] [PubMed]
  243. Gómez-García, I.; Trepiana, J.; Fernández-Quintela, A.; Giralt, M.; Portillo, M.P. Sexual Dimorphism in Brown Adipose Tissue Activation and White Adipose Tissue Browning. Int. J. Mol. Sci. 2022, 23, 8250. [Google Scholar] [CrossRef] [PubMed]
  244. Fletcher, L.A.; Kim, K.; Leitner, B.P.; Cassimatis, T.M.; O’Mara, A.E.; Johnson, J.W.; Halprin, M.S.; McGehee, S.M.; Brychta, R.J.; Cypess, A.M.; et al. Sexual Dimorphisms in Adult Human Brown Adipose Tissue. Obes. Silver Spring Md 2020, 28, 241–246. [Google Scholar] [CrossRef] [PubMed]
  245. Della Torre, S.; Benedusi, V.; Fontana, R.; Maggi, A. Energy Metabolism and Fertility—A Balance Preserved for Female Health. Nat. Rev. Endocrinol. 2014, 10, 13–23. [Google Scholar] [CrossRef] [PubMed]
  246. McCartney, C.R.; Marshall, J.C. Polycystic Ovary Syndrome. N. Engl. J. Med. 2016, 375, 54–64. [Google Scholar] [CrossRef]
  247. Joham, A.E.; Norman, R.J.; Stener-Victorin, E.; Legro, R.S.; Franks, S.; Moran, L.J.; Boyle, J.; Teede, H.J. Polycystic Ovary Syndrome. Lancet Diabetes Endocrinol. 2022, 10, 668–680. [Google Scholar] [CrossRef]
  248. Spremović Rađenović, S.; Pupovac, M.; Andjić, M.; Bila, J.; Srećković, S.; Gudović, A.; Dragaš, B.; Radunović, N. Prevalence, Risk Factors, and Pathophysiology of Nonalcoholic Fatty Liver Disease (NAFLD) in Women with Polycystic Ovary Syndrome (PCOS). Biomedicines 2022, 10, 131. [Google Scholar] [CrossRef]
  249. Falzarano, C.; Lofton, T.; Osei-Ntansah, A.; Oliver, T.; Southward, T.; Stewart, S.; Andrisse, S. Nonalcoholic Fatty Liver Disease in Women and Girls with Polycystic Ovary Syndrome. J. Clin. Endocrinol. Metab. 2022, 107, 258–272. [Google Scholar] [CrossRef]
  250. Sarkar, M.; Terrault, N.; Chan, W.; Cedars, M.I.; Huddleston, H.G.; Duwaerts, C.C.; Balitzer, D.; Gill, R.M. Polycystic Ovary Syndrome (PCOS) Is Associated with NASH Severity and Advanced Fibrosis. Liver Int. Off. J. Int. Assoc. Study Liver 2020, 40, 355–359. [Google Scholar] [CrossRef]
  251. Cui, P.; Hu, W.; Ma, T.; Hu, M.; Tong, X.; Zhang, F.; Shi, J.; Xu, X.; Li, X.; Shao, L.R.; et al. Long-Term Androgen Excess Induces Insulin Resistance and Non-Alcoholic Fatty Liver Disease in PCOS-like Rats. J. Steroid Biochem. Mol. Biol. 2021, 208, 105829. [Google Scholar] [CrossRef] [PubMed]
  252. Condorelli, R.A.; Calogero, A.E.; Di Mauro, M.; Mongioi’, L.M.; Cannarella, R.; Rosta, G.; La Vignera, S. Androgen Excess and Metabolic Disorders in Women with PCOS: Beyond the Body Mass Index. J. Endocrinol. Investig. 2018, 41, 383–388. [Google Scholar] [CrossRef]
  253. Roy, S.; Abudu, A.; Salinas, I.; Sinha, N.; Cline-Fedewa, H.; Yaw, A.M.; Qi, W.; Lydic, T.A.; Takahashi, D.L.; Hennebold, J.D.; et al. Androgen-Mediated Perturbation of the Hepatic Circadian System Through Epigenetic Modulation Promotes NAFLD in PCOS Mice. Endocrinology 2022, 163, bqac127. [Google Scholar] [CrossRef] [PubMed]
  254. Palioura, E.; Diamanti-Kandarakis, E. Polycystic Ovary Syndrome (PCOS) and Endocrine Disrupting Chemicals (EDCs). Rev. Endocr. Metab. Disord. 2015, 16, 365–371. [Google Scholar] [CrossRef]
  255. Hammarstrand, S.; Jakobsson, K.; Andersson, E.; Xu, Y.; Li, Y.; Olovsson, M.; Andersson, E.M. Perfluoroalkyl Substances (PFAS) in Drinking Water and Risk for Polycystic Ovarian Syndrome, Uterine Leiomyoma, and Endometriosis: A Swedish Cohort Study. Environ. Int. 2021, 157, 106819. [Google Scholar] [CrossRef] [PubMed]
  256. Al-Saleh, I. The Relationship between Urinary Phthalate Metabolites and Polycystic Ovary Syndrome in Women Undergoing in Vitro Fertilization: Nested Case-Control Study. Chemosphere 2022, 286, 131495. [Google Scholar] [CrossRef]
  257. Kim, K.; Pollack, A.Z.; Nobles, C.J.; Sjaarda, L.A.; Zolton, J.R.; Radoc, J.G.; Schisterman, E.F.; Mumford, S.L. Associations between Blood Cadmium and Endocrine Features Related to PCOS-Phenotypes in Healthy Women of Reproductive Age: A Prospective Cohort Study. Environ. Health Glob. Access Sci. Source 2021, 20, 64. [Google Scholar] [CrossRef]
  258. Zhang, C.; Xu, L.; Zhao, Y.; Wang, Y. Changes in Serum Heavy Metals in Polycystic Ovary Syndrome and Their Association with Endocrine, Lipid-Metabolism, Inflammatory Characteristics and Pregnancy Outcomes. Reprod. Toxicol. 2022, 111, 20–26. [Google Scholar] [CrossRef]
  259. Lin, S.-Y.; Yang, Y.-C.; Chang, C.Y.-Y.; Lin, C.-C.; Hsu, W.-H.; Ju, S.-W.; Hsu, C.-Y.; Kao, C.-H. Risk of Polycystic Ovary Syndrome in Women Exposed to Fine Air Pollutants and Acidic Gases: A Nationwide Cohort Analysis. Int. J. Environ. Res. Public. Health 2019, 16, 4816. [Google Scholar] [CrossRef]
  260. Abudawood, M.; Tabassum, H.; Alanazi, A.H.; Almusallam, F.; Aljaser, F.; Ali, M.N.; Alenzi, N.D.; Alanazi, S.T.; Alghamdi, M.A.; Altoum, G.H.; et al. Antioxidant Status in Relation to Heavy Metals Induced Oxidative Stress in Patients with Polycystic Ovarian Syndrome (PCOS). Sci. Rep. 2021, 11, 22935. [Google Scholar] [CrossRef]
  261. Zhang, B.; Zhou, W.; Shi, Y.; Zhang, J.; Cui, L.; Chen, Z.-J. Lifestyle and Environmental Contributions to Ovulatory Dysfunction in Women of Polycystic Ovary Syndrome. BMC Endocr. Disord. 2020, 20, 19. [Google Scholar] [CrossRef] [PubMed]
  262. Treviño, L.S.; Katz, T.A. Endocrine Disruptors and Developmental Origins of Nonalcoholic Fatty Liver Disease. Endocrinology 2018, 159, 20–31. [Google Scholar] [CrossRef] [PubMed]
  263. Lynch, C.; Chan, C.S.; Drake, A.J. Early Life Programming and the Risk of Non-Alcoholic Fatty Liver Disease. J. Dev. Orig. Health Dis. 2017, 8, 263–272. [Google Scholar] [CrossRef] [PubMed]
  264. Long, Z.; Fan, J.; Wu, G.; Liu, X.; Wu, H.; Liu, J.; Chen, Y.; Su, S.; Cheng, X.; Xu, Z.; 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. [Google Scholar] [CrossRef]
  265. Lin, R.; Wu, D.; Wu, F.-J.; Meng, Y.; Zhang, J.-H.; Wang, X.-G.; Jia, L.-H. Non-Alcoholic Fatty Liver Disease Induced by Perinatal Exposure to Bisphenol a Is Associated With Activated MTOR and TLR4/NF-ΚB Signaling Pathways in Offspring Rats. Front. Endocrinol. 2019, 10, 620. [Google Scholar] [CrossRef]
  266. Shimpi, P.C.; More, V.R.; Paranjpe, M.; Donepudi, A.C.; Goodrich, J.M.; Dolinoy, D.C.; Rubin, B.; Slitt, A.L. 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. [Google Scholar] [CrossRef] [PubMed]
  267. Dabeer, S.; Raisuddin, S. Perinatal Exposure to Environmental Endocrine Disruptor Bisphenol A Aggravates the Onset of Non-Alcoholic Fatty Liver Disease (NAFLD) in Weanling F1 Offspring of Obese Rats. Environ. Sci. Pollut. Res. 2023, 30, 3146–3165. [Google Scholar] [CrossRef]
  268. Wei, J.; Sun, X.; Chen, Y.; Li, Y.; Song, L.; Zhou, Z.; Xu, B.; Lin, Y.; Xu, S. Perinatal Exposure to Bisphenol A Exacerbates Nonalcoholic Steatohepatitis-like Phenotype in Male Rat Offspring Fed on a High-Fat Diet. J. Endocrinol. 2014, 222, 313–325. [Google Scholar] [CrossRef]
  269. Strakovsky, R.S.; Wang, H.; Engeseth, N.J.; Flaws, J.A.; Helferich, W.G.; Pan, Y.-X.; Lezmi, S. Developmental Bisphenol A (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, 101–112. [Google Scholar] [CrossRef]
  270. Marchlewicz, E.; McCabe, C.; Djuric, Z.; Hoenerhoff, M.; Barks, J.; Tang, L.; Song, P.X.; Peterson, K.; Padmanabhan, V.; Dolinoy, D.C. 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. [Google Scholar] [CrossRef]
  271. Wang, D.; Yan, S.; Yan, J.; Teng, M.; Meng, Z.; Li, R.; Zhou, Z.; Zhu, W. Effects of Triphenyl Phosphate Exposure during Fetal Development on Obesity and Metabolic Dysfunctions in Adult Mice: Impaired Lipid Metabolism and Intestinal Dysbiosis. Environ. Pollut. 2019, 246, 630–638. [Google Scholar] [CrossRef] [PubMed]
  272. Ditzel, E.J.; Nguyen, T.; Parker, P.; Camenisch, T.D. Effects of Arsenite Exposure during Fetal Development on Energy Metabolism and Susceptibility to Diet-Induced Fatty Liver Disease in Male Mice. Environ. Health Perspect. 2016, 124, 201–209. [Google Scholar] [CrossRef] [PubMed]
  273. Stratakis, N.; Conti, D.V.; Jin, R.; Margetaki, K.; Valvi, D.; Siskos, A.P.; Maitre, L.; Garcia, E.; Varo, N.; Zhao, Y.; et al. Prenatal Exposure to Perfluoroalkyl Substances Associated with Increased Susceptibility to Liver Injury in Children. Hepatology 2020, 72, 1758–1770. [Google Scholar] [CrossRef] [PubMed]
  274. Stratakis, N.; Golden-Mason, L.; Margetaki, K.; Zhao, Y.; Valvi, D.; Garcia, E.; Maitre, L.; Andrusaityte, S.; Basagana, X.; Borràs, E.; et al. In Utero Exposure to Mercury Is Associated with Increased Susceptibility to Liver Injury and Inflammation in Childhood. Hepatology 2021, 74, 1546–1559. [Google Scholar] [CrossRef]
  275. Garcia, E.; Stratakis, N.; Valvi, D.; Maitre, L.; Varo, N.; Aasvang, G.M.; Andrusaityte, S.; Basagana, X.; Casas, M.; de Castro, M.; et al. Prenatal and Childhood Exposure to Air Pollution and Traffic and the Risk of Liver Injury in European Children. Environ. Epidemiol. 2021, 5, e153. [Google Scholar] [CrossRef]
  276. Lo, E.K.K.; Felicianna; Xu, J.-H.; Zhan, Q.; Zeng, Z.; El-Nezami, H. The Emerging Role of Branched-Chain Amino Acids in Liver Diseases. Biomedicines 2022, 10, 1444. [Google Scholar] [CrossRef]
  277. Grenier-Larouche, T.; Coulter Kwee, L.; Deleye, Y.; Leon-Mimila, P.; Walejko, J.M.; McGarrah, R.W.; Marceau, S.; Trahan, S.; Racine, C.; Carpentier, A.C.; et al. Altered Branched-Chain α-Keto Acid Metabolism Is a Feature of NAFLD in Individuals with Severe Obesity. JCI Insight 2022, 7, e159204. [Google Scholar] [CrossRef]
  278. van den Berg, E.H.; Flores-Guerrero, J.L.; Gruppen, E.G.; de Borst, M.H.; Wolak-Dinsmore, J.; Connelly, M.A.; Bakker, S.J.L.; Dullaart, R.P.F. Non-Alcoholic Fatty Liver Disease and Risk of Incident Type 2 Diabetes: Role of Circulating Branched-Chain Amino Acids. Nutrients 2019, 11, 705. [Google Scholar] [CrossRef]
  279. Lischka, J.; Schanzer, A.; Hojreh, A.; Ba Ssalamah, A.; Item, C.B.; de Gier, C.; Walleczek, N.-K.; Metz, T.F.; Jakober, I.; Greber-Platzer, S.; et al. A Branched-Chain Amino Acid-Based Metabolic Score Can Predict Liver Fat in Children and Adolescents with Severe Obesity. Pediatr. Obes. 2021, 16, e12739. [Google Scholar] [CrossRef]
  280. Yan, S.; Tian, S.; Meng, Z.; Teng, M.; Sun, W.; Jia, M.; Zhou, Z.; Bi, S.; Zhu, W. Exposure to Nitenpyram during Pregnancy Causes Colonic Mucosal Damage and Non-Alcoholic Steatohepatitis in Mouse Offspring: The Role of Gut Microbiota. Environ. Pollut. 2021, 271, 116306. [Google Scholar] [CrossRef]
  281. Wang, X.; Zhao, Z.; Wang, X.; Hu, W.; Chu, X.; Qian, M.; Wang, R.; Yu, S.; Wu, Q.; Tang, J.; et al. Effects of Polystyrene Nanoplastic Gestational Exposure on Mice. Chemosphere 2023, 324, 138255. [Google Scholar] [CrossRef]
  282. Sun, J.; Liu, H.; Zhang, C.; Liu, X.; Sun, X.; Chen, X.; Yang, G.; Wang, N. Predisposed Obesity and Long-Term Metabolic Diseases from Maternal Exposure to Fine Particulate Matter (PM2.5)—A Review of Its Effect and Potential Mechanisms. Life Sci. 2022, 310, 121054. [Google Scholar] [CrossRef]
  283. Wu, G.; Brown, J.; Zamora, M.L.; Miller, A.; Satterfield, M.C.; Meininger, C.J.; Steinhauser, C.B.; Johnson, G.A.; Burghardt, R.C.; Bazer, F.W.; et al. Adverse Organogenesis and Predisposed Long-Term Metabolic Syndrome from Prenatal Exposure to Fine Particulate Matter. Proc. Natl. Acad. Sci. USA 2019, 116, 11590–11595. [Google Scholar] [CrossRef] [PubMed]
  284. Song, Y.; Chen, L.; Bennett, E.; Wheeler, A.J.; Southam, K.; Yen, S.; Johnston, F.; Zosky, G.R. Can Maternal Exposure to Air Pollution Affect Post-Natal Liver Development? Toxics 2023, 11, 61. [Google Scholar] [CrossRef] [PubMed]
  285. Pejhan, A.; Agah, J.; Adli, A.; Mehrabadi, S.; Raoufinia, R.; Mokamel, A.; Abroudi, M.; Ghalenovi, M.; Sadeghi, Z.; Bolghanabadi, Z.; et al. Exposure to Air Pollution during Pregnancy and Newborn Liver Function. Chemosphere 2019, 226, 447–453. [Google Scholar] [CrossRef] [PubMed]
  286. Wang, X.; Yang, Y.; Zhu, P.; Wu, Y.; Jin, Y.; Yu, S.; Wei, H.; Qian, M.; Cao, W.; Xu, S.; et al. Prenatal Exposure to Diesel Exhaust PM2.5 Programmed Non-Alcoholic Fatty Liver Disease Differently in Adult Male Offspring of Mice Fed Normal Chow and a High-Fat Diet. Environ. Pollut. 2019, 255, 113366. [Google Scholar] [CrossRef] [PubMed]
  287. Tilg, H.; Adolph, T.E.; Moschen, A.R. Multiple Parallel Hits Hypothesis in Nonalcoholic Fatty Liver Disease: Revisited After a Decade. Hepatology 2021, 73, 833–842. [Google Scholar] [CrossRef]
  288. Chen, H.; Van Reyk, D.; Oliveira, A.; Chan, Y.L.; Town, S.E.; Rayner, B.; Pollock, C.A.; Saad, S.; George, J.; Padula, M.P.; et al. Sex-Dependent Responses to Maternal Exposure to PM2.5 in the Offspring. Antioxidants 2022, 11, 2255. [Google Scholar] [CrossRef]
Figure 1. In addition to obesity, lifestyle, genetic and epigenetic factors, and gut microbiota dysbiosis, dietary intake of several environmental pollutants, including persistent organic pollutants (POPs), endocrine disrupting chemicals (EDCs), heavy metals, micro- and nanoplastics promotes NAFLD development and progression. In addition to ingestion, exposure to air particulate matter through inhalation can be another risk factor for developing NAFLD, especially in the context of high urbanization due to the combination with other risk factors, including obesogenic food environment. Figure created with BioRender (https://biorender.com/, accessed on 11 April 2023).
Figure 1. In addition to obesity, lifestyle, genetic and epigenetic factors, and gut microbiota dysbiosis, dietary intake of several environmental pollutants, including persistent organic pollutants (POPs), endocrine disrupting chemicals (EDCs), heavy metals, micro- and nanoplastics promotes NAFLD development and progression. In addition to ingestion, exposure to air particulate matter through inhalation can be another risk factor for developing NAFLD, especially in the context of high urbanization due to the combination with other risk factors, including obesogenic food environment. Figure created with BioRender (https://biorender.com/, accessed on 11 April 2023).
Nutrients 15 02335 g001
Figure 2. Major effects of persistent EDCs on the main pathways involved in NAFLD development. (a) The steatosis-inducing effect of EDCs is mediated by the binding with different nuclear receptors. (b) EDC-induced disruption of nuclear receptor signaling promotes lipid accumulation within hepatocytes through the increase in lipid uptake and DNL, the inhibition of FAO, and the reduction of lipid secretion in the form of VLDL. EDCs induce oxidative stress and hepatic inflammation by inducing the polarization of Kupffer cells towards a pro-inflammatory phenotype, increasing immune cell infiltration, and by transforming HSCs into myofibroblast-like cells, thus favoring liver damage and fibrosis. EDCs-induced gut dysbiosis contributes to NAFLD progression by sustaining a pro-inflammatory status. The pathways/factors that increased or decreased are shown in red or blue, respectively. Abbreviations: AhR: hydrocarbon receptor; BPA: Bisphenol A; CAR: constitutive activated receptor; DNL: de novo lipogenesis; ERs: estrogen receptors; FAs: fatty acids; FAO: fatty acid oxidation; FXR: farnesoid X receptor; HSCs: hepatic stellate cells; IR: insulin resistance; Kupffer M1: Kupffer cells with an M1 phenotype; LXR: liver X receptor; OCPs: organochlorine pesticides; PAHs:, polyaromatic hydrocarbons; PCBs: polychlorinated biphenyls; PFAAs: perfluoroalkyl acids; PPARs: peroxisome proliferator-activated receptors; PXR: pregnane X receptor; RXR: retinoid X receptor; TG: triglycerides; VLDL: very-low density lipoproteins. Figure created with BioRender (https://biorender.com/, accessed on 11 May 2023).
Figure 2. Major effects of persistent EDCs on the main pathways involved in NAFLD development. (a) The steatosis-inducing effect of EDCs is mediated by the binding with different nuclear receptors. (b) EDC-induced disruption of nuclear receptor signaling promotes lipid accumulation within hepatocytes through the increase in lipid uptake and DNL, the inhibition of FAO, and the reduction of lipid secretion in the form of VLDL. EDCs induce oxidative stress and hepatic inflammation by inducing the polarization of Kupffer cells towards a pro-inflammatory phenotype, increasing immune cell infiltration, and by transforming HSCs into myofibroblast-like cells, thus favoring liver damage and fibrosis. EDCs-induced gut dysbiosis contributes to NAFLD progression by sustaining a pro-inflammatory status. The pathways/factors that increased or decreased are shown in red or blue, respectively. Abbreviations: AhR: hydrocarbon receptor; BPA: Bisphenol A; CAR: constitutive activated receptor; DNL: de novo lipogenesis; ERs: estrogen receptors; FAs: fatty acids; FAO: fatty acid oxidation; FXR: farnesoid X receptor; HSCs: hepatic stellate cells; IR: insulin resistance; Kupffer M1: Kupffer cells with an M1 phenotype; LXR: liver X receptor; OCPs: organochlorine pesticides; PAHs:, polyaromatic hydrocarbons; PCBs: polychlorinated biphenyls; PFAAs: perfluoroalkyl acids; PPARs: peroxisome proliferator-activated receptors; PXR: pregnane X receptor; RXR: retinoid X receptor; TG: triglycerides; VLDL: very-low density lipoproteins. Figure created with BioRender (https://biorender.com/, accessed on 11 May 2023).
Nutrients 15 02335 g002
Figure 3. Major effects of heavy metals on the main pathways involved in NAFLD development. Dietary intake of heavy metals leads to impaired regulation of lipid metabolism, by promoting lipid synthesis and deposition and by inhibiting lipid catabolism. Heavy metals enhance DNL also through the increased transactivation of ChREBP, favoring TG synthesis and lipid deposition. Heavy metals impair mitochondrial functions and inhibit SIRT1, FAO, and autophagy, resulting in increased production of ROS and lipotoxicity, which in turn activate NLRP3 inflammasome and boost inflammation, further favoring NAFLD progression. Heavy metals contribute to NAFLD pathogenesis also by affecting gut permeability, thus increasing the flux of SCFA and inflammatory molecules toward the liver. The pathways/factors that increased or decreased are shown in red or blue, respectively. Abbreviations: ChREBP: carbohydrate responsive element binding protein; DNL: de novo lipogenesis; FAs: fatty acids; FAO: fatty acid oxidation; NLRP3: NLR family pyrin domain containing 3; ROS: reactive oxygen species; SCFA: short chain fatty acids; SIRT1: sirtuin 1; TG: triglyceride. Figure created with BioRender (https://biorender.com/, accessed on 11 May 2023).
Figure 3. Major effects of heavy metals on the main pathways involved in NAFLD development. Dietary intake of heavy metals leads to impaired regulation of lipid metabolism, by promoting lipid synthesis and deposition and by inhibiting lipid catabolism. Heavy metals enhance DNL also through the increased transactivation of ChREBP, favoring TG synthesis and lipid deposition. Heavy metals impair mitochondrial functions and inhibit SIRT1, FAO, and autophagy, resulting in increased production of ROS and lipotoxicity, which in turn activate NLRP3 inflammasome and boost inflammation, further favoring NAFLD progression. Heavy metals contribute to NAFLD pathogenesis also by affecting gut permeability, thus increasing the flux of SCFA and inflammatory molecules toward the liver. The pathways/factors that increased or decreased are shown in red or blue, respectively. Abbreviations: ChREBP: carbohydrate responsive element binding protein; DNL: de novo lipogenesis; FAs: fatty acids; FAO: fatty acid oxidation; NLRP3: NLR family pyrin domain containing 3; ROS: reactive oxygen species; SCFA: short chain fatty acids; SIRT1: sirtuin 1; TG: triglyceride. Figure created with BioRender (https://biorender.com/, accessed on 11 May 2023).
Nutrients 15 02335 g003
Figure 4. Major effects of MPs and NPs on the main pathways involved in NAFLD development. Dietary intake of MPs and NPs promotes NAFLD by increasing the hepatic expression of PPARγ and their target genes leading to enhanced DNL and TG synthesis, and by reducing FAO and lipid catabolism, mainly through the inhibition of PPARα. Chemical additives associated with MPs/NPs lead to mitochondrial damage, oxidative stress, and inflammation. Oxidative stress and inflammation boost IR, further favoring DNL and lipid deposition. Excessive oxidative stress and inflammation prompt programmed cell death through apoptosis, pyroptosis, and ferroptosis. MPs/NPs-induced gut dysbiosis also contribute to NAFLD pathogenesis by increasing inflammation and the production of SCFA that, once reached the liver, feeds the pool of FAs. The pathways/factors increased or decreased are shown in red or blue, respectively. Abbreviations: DNL: de novo lipogenesis; FAs: fatty acids; FAO: fatty acid oxidation; IR: insulin resistance; MPs: microplastics; NPs: nanoplastics; PPARα: peroxisome proliferator-activated receptor α; PPARγ: peroxisome proliferator-activated receptor γ; ROS: reactive oxygen species; SCFA: short chain fatty acids; TG: triglyceride. Figure created with BioRender (https://biorender.com/, accessed on 11 May 2023).
Figure 4. Major effects of MPs and NPs on the main pathways involved in NAFLD development. Dietary intake of MPs and NPs promotes NAFLD by increasing the hepatic expression of PPARγ and their target genes leading to enhanced DNL and TG synthesis, and by reducing FAO and lipid catabolism, mainly through the inhibition of PPARα. Chemical additives associated with MPs/NPs lead to mitochondrial damage, oxidative stress, and inflammation. Oxidative stress and inflammation boost IR, further favoring DNL and lipid deposition. Excessive oxidative stress and inflammation prompt programmed cell death through apoptosis, pyroptosis, and ferroptosis. MPs/NPs-induced gut dysbiosis also contribute to NAFLD pathogenesis by increasing inflammation and the production of SCFA that, once reached the liver, feeds the pool of FAs. The pathways/factors increased or decreased are shown in red or blue, respectively. Abbreviations: DNL: de novo lipogenesis; FAs: fatty acids; FAO: fatty acid oxidation; IR: insulin resistance; MPs: microplastics; NPs: nanoplastics; PPARα: peroxisome proliferator-activated receptor α; PPARγ: peroxisome proliferator-activated receptor γ; ROS: reactive oxygen species; SCFA: short chain fatty acids; TG: triglyceride. Figure created with BioRender (https://biorender.com/, accessed on 11 May 2023).
Nutrients 15 02335 g004
Figure 5. Exposure to environmental endocrine pollutants such as BPA increases the susceptibility of PCOS women to develop NAFLD. In ovarian theca cells, BPA enhances the production of androgens, further impairing T/E2 ratio. At the hepatic level, BPA decreases SHBG, thus contributing to high levels of free T. In the liver, androgens downregulate BPA liver catabolism, thereby increasing circulating BPA levels. Abbreviations: BPA: Bisphenol A; E2: estradiol/estrogens; PCOS: polycystic ovary syndrome; SHBG: sex hormone-binding globulin; T: testosterone/androgens. Figure created with BioRender (https://biorender.com/, accessed on 11 May 2023).
Figure 5. Exposure to environmental endocrine pollutants such as BPA increases the susceptibility of PCOS women to develop NAFLD. In ovarian theca cells, BPA enhances the production of androgens, further impairing T/E2 ratio. At the hepatic level, BPA decreases SHBG, thus contributing to high levels of free T. In the liver, androgens downregulate BPA liver catabolism, thereby increasing circulating BPA levels. Abbreviations: BPA: Bisphenol A; E2: estradiol/estrogens; PCOS: polycystic ovary syndrome; SHBG: sex hormone-binding globulin; T: testosterone/androgens. Figure created with BioRender (https://biorender.com/, accessed on 11 May 2023).
Nutrients 15 02335 g005
Figure 6. Gestational exposure to environmental pollutants acting as EDCs interferes with liver metabolism programming and increases the risk of developing NAFLD later in life. Figure created with BioRender (https://biorender.com/, accessed on 11 May 2023).
Figure 6. Gestational exposure to environmental pollutants acting as EDCs interferes with liver metabolism programming and increases the risk of developing NAFLD later in life. Figure created with BioRender (https://biorender.com/, accessed on 11 May 2023).
Nutrients 15 02335 g006
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Dolce, A.; Della Torre, S. Sex, Nutrition, and NAFLD: Relevance of Environmental Pollution. Nutrients 2023, 15, 2335. https://doi.org/10.3390/nu15102335

AMA Style

Dolce A, Della Torre S. Sex, Nutrition, and NAFLD: Relevance of Environmental Pollution. Nutrients. 2023; 15(10):2335. https://doi.org/10.3390/nu15102335

Chicago/Turabian Style

Dolce, Arianna, and Sara Della Torre. 2023. "Sex, Nutrition, and NAFLD: Relevance of Environmental Pollution" Nutrients 15, no. 10: 2335. https://doi.org/10.3390/nu15102335

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

Article Metrics

Back to TopTop