Next Article in Journal
Assessment of Organic Matter Influence on the Ecological Integrity of Poyang Lake Using O/E Model and Chemical–Biological Indices over the Past Two Decades
Previous Article in Journal
PAH Contamination, Sources and Health Risks in Black Soil Region of Jilin Province, China
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Benzo(a)pyrene and Gut Microbiome Crosstalk: Health Risk Implications

1
Center for Natural Product Systems Biology, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea
2
Department of Aquatic Life Medicine, College of Life Sciences, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
3
Department of Food Science, College of Life Sciences, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
4
Natural Products Applied Science, KIST School, University of Science and Technology, Gangneung 25451, Republic of Korea
5
Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, 20, Ilsan-ro, Wonju 26493, Republic of Korea
*
Author to whom correspondence should be addressed.
Toxics 2024, 12(12), 938; https://doi.org/10.3390/toxics12120938
Submission received: 6 November 2024 / Revised: 13 December 2024 / Accepted: 17 December 2024 / Published: 23 December 2024
(This article belongs to the Section Emerging Contaminants)

Abstract

This review delves into the impact of benzo(a)pyrene (B(a)P), which is a toxic and pervasive polycyclic aromatic hydrocarbon (PAH) and known carcinogen, on the human health risk from a gut microbiome perspective. We retrieved the relevant articles on each PAH and summarized the reporting to date, with a particular focus on benzo(a)pyrene, which has been reported to have a high risk of gut microbiome-related harm. B(a)P exposure can compromise the homeostasis of the gut microbiota, leading to dysbiosis, a state of microbial imbalance. The consequences of B(a)P-induced gut dysbiosis can be far-reaching, potentially contributing to inflammation, metabolic disorders, and an increased risk of various diseases. Additionally, due to the strong coupling between B(a)P and microparticles, the toxicity of B(a)P may be further compounded by its reaction with strong gut disruptors such as micro-/nanoplastics, which have recently become a serious environmental concern. This review summarizes current research on the impact of B(a)P on the gut microbiome, highlighting the intricate relationship between environmental exposure, gut health, and human disease. Further research is necessary to elucidate the underlying mechanisms and develop effective strategies to mitigate the adverse health effects of B(a)P exposure.

Graphical Abstract

1. Introduction

Research on the health impacts of environmental pollutants has evolved significantly over time. Polycyclic aromatic hydrocarbons (PAHs) are a class of ubiquitous environmental pollutants that pose significant health risks to humans. These persistent organic compounds, formed primarily through incomplete combustion of organic materials, are widespread in both urban and rural environments. PAHs have garnered considerable attention from the scientific community and regulatory bodies due to their potential to cause adverse health effects, even at low exposure levels [1,2]. The ubiquity of PAHs in the environment, coupled with their ability to bioaccumulate in the food chain, makes human exposure nearly unavoidable. Sources of exposure are diverse, ranging from air pollution and occupational settings to dietary intake and tobacco smoke. This widespread presence underscores the critical need for continued research, effective monitoring, and stringent regulatory measures to mitigate the health risks posed by these pervasive contaminants. The health risks associated with PAH exposure are of particular concern. Numerous studies have established strong links between PAH exposure and various forms of cancer, including lung, skin, and bladder cancers [3,4,5]. The International Agency for Research on Cancer (IARC) has classified several PAHs, most notably benzo[a]pyrene (B(a)P), as Group 1 carcinogens, indicating that they are carcinogenic to humans.
The gut microbiome, a complex community of microorganisms in the human gastrointestinal tract, plays a crucial role in various physiological processes, including nutrient absorption, immune system development, and protection against pathogens [6,7]. Disruptions to this complex ecosystem, often referred to as dysbiosis, can have far-reaching implications for human health. Understanding the complex interplay between environmental exposures, the gut microbiome, and human health has become increasingly important in recent years for a variety of toxicants, including heavy metals, persistent organic pollutants, and microplastics (MPs).
Figure 1 illustrates the publication trends in PAH research by 2023, highlighting the research field for various PAH compounds and increasing the focus on specific PAHs such as naphthalene, pyrene, and B(a)P. While other PAHs such as naphthalene, phenanthrene, and pyrene continue to be studied, research on B(a)P has expanded considerably, particularly regarding its impact on the gut microbiome. B(a)P exposure has been shown to induce gut dysbiosis, altering the composition and diversity of the gut microbiota and potentially impairing its functionality [8,9]. While previous B(a)P research characterized the metabolic disturbance, carcinogenic potential, and broader health impacts, recent B(a)P research has shifted its focus from traditional toxicity studies to exploring the gut microbiome’s role in mediating its effects. This emerging research underscores the critical importance of the gut microbiome in shaping our response to environmental pollutants such as B(a)P.
Moreover, oxidative stress, an imbalance between reactive oxygen species production and antioxidant defenses, can significantly impact gut microbiota. Excessive reactive oxygen species (ROS) can damage cellular components, including DNA, proteins, and lipids, potentially disrupting the gut microbiota balance and leading to dysbiosis. The gut microbiota itself plays a role in regulating oxidative stress, producing both ROS and antioxidants. This complex interplay between oxidative stress and gut microbiota is an important area of research, with implications for understanding various health and disease states [10].
Overall, this review delves into the impact of B(a)P exposure on the gut microbiome, exploring the mechanisms by which B(a)P disrupts microbial balance and the potential consequences for human health. We reviewed the effects of B(a)P on gut microbial diversity, composition, and functionality, drawing upon existing research to provide a comprehensive overview of this critical area of study. Understanding the interplay between B(a)P exposure and the gut microbiome is crucial for developing strategies to mitigate the health risks associated with this pervasive environmental pollutant.

2. Literature Search and Selection Methods

A thorough literature search was conducted across multiple databases, including PubMed, Scopus, ScienceDirect, Springer Online, and Google Scholar, to identify relevant studies published between 2012 and 2024. The search employed the following keywords: “Benzo(a)pyrene AND health risk AND gut microbiome AND heavy metals AND (microplastics OR nanoplastics)”. We excluded review and research articles that focused on gut microbiome studies but did not specifically address gut microbiome dysbiosis and its potential link to health risks. Over 50 research articles were reviewed to evaluate the effects of B(a)P and co-exposure to environmental pollutants, such as microplastics, on risks through gut microbiome disturbances.

3. B(a)P and Its Health Risks

3.1. History of B(a)P Health Risk Study

Research on B(a)P has a history spanning decades (Figure 2). It was first identified as a component of coal tar as early as 1913 and was scientifically reported in 1945 [11]. As illustrated in Figure 2, from 1946 to the 1980s, scientists established the study of B(a)P biochemistry and metabolic activation, where it becomes highly reactive in the body (in vivo), leading to DNA damage and cancer. In the 1990s, governments and regulatory bodies, such as the U.S. Environmental Protection Agency (EPA), established limits and guidelines for B(a)P exposure in air, water, burnt food, and soil to protect public health and the environment. Ongoing studies have developed biomarkers to measure B(a)P toxicity exposure in individuals, and ongoing research explores establishing a mode of action link between this compound and cancer [5,12]. B(a)P remains a focal point of research in toxicology and environmental science, where scientists are exploring its health hazards, routes of exposure, and strategies to mitigate its effects on human health and aquatic environments. For example, research on combined exposure to MPs and nanoplastics (NPs) in aquatic animals has gained significant attention in recent years due to growing concern about the environmental impact of plastic pollution [13,14,15].
Particularly in recent studies, researchers continue to investigate the extent of contamination, the mechanisms of toxicity, and the long-term ecological implications for the gut microbiome in individual animals, particularly in aquatic animals [43,44] and mice [45]. This area of research is shedding light on the complex interactions between pollutants, microbial communities, and ecological processes, with implications for both environmental health and human well-being.

3.2. Toxic Mode of Action of B(a)P

B(a)P undergoes metabolic activation in the human body, which can lead to the formation of highly reactive intermediate metabolites [46]. Briefly, the enzymes in the liver convert B(a)P into reactive intermediates that can covalently bind to DNA, leading to genetic mutations [4]. Once inside the body, B(a)P undergoes metabolic activation, primarily by enzymes such as cytochrome P450 enzymes (e.g., CYP1A1 and CYP1B1), which can convert it into highly carcinogenic reactive intermediates, such as B(a)P-7,8-dihydrodiol-9,10-epoxide (BPDE), in tissues such as the liver and others [47,48]. Reactive intermediates such as BPDE can covalently bind to DNA, forming DNA adducts that distort the standard DNA structure, and these DNA disruptions lead to mutations in critical genes, including tumor suppressor genes and oncogenes [49]. Furthermore, B(a)P exposure can generate reactive oxygen species (ROS) and induce oxidative stress within cells [50,51]. ROS can damage cellular components, including DNA, proteins, and lipids, increasing the risk of oxidative DNA damage and promoting inflammation [52,53,54]. Consequently, this imbalance has been implicated in the pathogenesis of various diseases, such as cancer, cardiovascular diseases, and respiratory disorders.
B(a)P is metabolically activated to a series of reactive intermediates by CYP450 enzymes, particularly CYP1A1 and CYP1B1, under the control of the aryl-hydrocarbon receptor (AhR). There is strong evidence that the benzo(a)pyrene diol epoxide (BPDE) mechanism operates in mouse lung tumorigenesis [55], while there is also strong evidence that both the radical-cation and the diol epoxide mechanisms are involved in mouse skin carcinogenesis [5]. The first step in the action of B(a)P metabolic transformation is the binding of the molecule to the aryl hydrocarbon receptor (AhR), which leads to the activation of CYP1A1 and the formation of reactive intermediates [56,57]. The AhR is a transcription factor that regulates gene expression in xenobiotic metabolisms, such as CYP1A1 and CYP1A2 [58,59]. The oxidation reactions of B(a)P activation are promoted by cytochrome P450s, with CYP1A1 and CYP1B1 exhibiting the highest catalytic specificity towards B(a)P [60,61]. In vitro experiments with recombinant human P450 enzymes from E. coli and Trichoplusia in cells have shown that CYP1A1 and CYP1B1 exhibit the highest catalytic specificity towards B(a)P [62]. CYP1A1 was considered uniquely responsible for PAH activation until the early 1990s, when CYP1B1 was identified. Thus, factors that influence this balance, including genetic variations in CYP enzymes and the presence of protective antioxidants, can modulate an individual’s susceptibility to B(a)P-induced toxicity [63]. Understanding these metabolic pathways is essential for assessing the health risks associated with B(a)P exposure and for developing strategies to mitigate its toxic effects.
Additionally, mitochondrial unfolded protein response (UPRmt) is a critical pathway implicated in various diseases, including metabolic disorders and cancer. Emerging evidence suggests an interesting connection between the UPRmt and the gut microbiota. The gut microbiota plays a vital role in host metabolism and immune modulation, influencing the development of both metabolic disorders and cancer. Certain pathogenic gut bacteria can trigger inflammatory responses and recruit specific immune cells, thereby impacting the progression of these diseases. Some bacterial pathogens release toxins that directly target mitochondria, leading to dysfunction, protein damage, and potential disruption of the mitochondrial membrane. These toxins can induce mtDNA damage and trigger apoptotic pathways [64,65]. Alterations in mitochondrial function, particularly oxidative phosphorylation (OxPhos), are key factors in the development of metabolic diseases such as obesity and insulin resistance. In the context of cancer, the inhibition of key UPRmt regulators, such as ATF5, ATF4, and SIRT, has been linked to cancer growth and progression. ATF5, a retrograde signaling molecule, is overexpressed during UPRmt activation and contributes to cancer cell invasiveness by inducing integrin expression and maintaining OxPhos function. ATF4 enhances the resistance of gastric cancer to chemotherapeutic drugs, such as cisplatin. Additionally, SIRT-mediated UPRmt can promote cancer cell metastasis by preserving mtDNA mutations, thereby maintaining mitochondrial function during metastasis [66].

3.3. Human Exposure and Epidemiological Research of B(a)P

Some clinical studies have evaluated the actual risk exposure of B(a)P, other PAHs, and other contaminants. The summarized data in Table 1 provide reference concentrations for pollutant compounds, including B(a)P, which refers to the Integrated Risk Information System (IRIS) and the United States EPA. B(a)P is classified as a Group 1 carcinogen by the International Agency for Research on Cancer (IARC) in human adverse-level effect at dose 1 per mg/kg-day. There is substantial evidence supporting its carcinogenicity in humans when compared to other PAHs, such as naphthalene, fluorene, anthracene, fluoranthene, phenanthrene, chrysene, pyrene, benzo(a)anthracene, 1-nitropyrene, and corannulene. Although these other PAHs may still pose health risks, particularly with chronic or high-level exposures, B(a)P remains the most concerning regarding its carcinogenic potential and well-documented health risks. B(a)P’s health risks are primarily associated with inhalation of contaminated air, often found in areas with elevated levels of air pollution, and dietary intake, particularly from grilled or smoked foods and tobacco smoke [63]. When considering their exposure routes closely related to daily exposure for organisms, including humans, B(a)P tends to have higher toxicity effects on health risks compared to other persistent organic pollutants (POPs) and heavy metals such as polychlorinated biphenyls (PCBs) and lead (Pb), which are mainly linked to a lesser extent exposure through inhalation in industrial or specific environmental settings [67].
B(a)P has a substantial impact on epidemiological research, primarily due to its well-established carcinogenic properties and its association with various adverse health outcomes, including cardiovascular diseases, immunotoxicity, reproductive issues, respiratory disorders, gastrointestinal disturbances, and DNA damage, which can lead to the development of cancer [68,69,70]. In the realm of cancer epidemiology, extensive studies have firmly established a strong link between B(a)P exposure and several types of cancer. Interestingly, epidemiological research on B(a)P in the context of the gut microbiome is a relatively emerging area of study, but holds significant potential implications for our understanding of environmental exposures and their effects on human health. Previous studies have revealed significant associations between gut microbiome dysbiosis and the development of cancers like colorectal and lung cancer [71]. Recent research suggests that the gut microbiome has local and systemic effects on cancer disease and that gut dysbiosis is involved in carcinogenesis induced by pollutants and may play a role in the progression of cancer development.
Table 1. Exposure levels and legally safe permissible concentrations for PAHs in humans.
Table 1. Exposure levels and legally safe permissible concentrations for PAHs in humans.
PollutantStructurePollutant SourcesHuman Daily Exposure LevelHuman Adverse Level Concentration 1
CarcinogenicOther Systems
NaphthaleneToxics 12 00938 i001Household and industryAssumed for 70 kg adult is 1.127 μg/kg per day from air, 0.237 μg/kg per day from food, and 0.235 μg/kg per day from house dust Probable human carcinogenRespiratory: 9.3 mg/m3 (LOAEL-HEC)
FluoreneToxics 12 00938 i002IndustryNot assessedNot assessedNot assessed
AnthraceneToxics 12 00938 i003Industry77.4 ng/m3 from the air [72]Not classifiable as to human carcinogenicity1.0 × 103 mg/kg-day (NOAEL)
FluorantheneToxics 12 00938 i004IndustryNot assessedNot assessedHepatic, urinary: 1.25 × 102 mg/kg-day (NOAEL)
PhenanthreneToxics 12 00938 i005IndustryNot assessedNot classifiable as to human carcinogenicity0.1–0.2 mg/m3 for airborne exposure limit
ChryseneToxics 12 00938 i006IndustryNot assessedProbable human carcinogenNot assessed
PyreneToxics 12 00938 i007IndustryNot assessedNot classifiable as to human carcinogenicityUrinary: 7.5 × 10 mg/kg-day (NOAEL)
Benz(a)anthraceneToxics 12 00938 i008IndustryNot assessedProbable human carcinogenNot assessed
1-NitropyreneToxics 12 00938 i009IndustryNot assessedNot assessedNot assessed
Benzo(a)pyreneToxics 12 00938 i010Household and industry52 to 95 ng/cigarette, 7.20 ± 1.11 μg/m3 in the air, 4.15 μg/kg in well-done steaks, and 4.00 to 8.33 ng/L in drinking water [63]1 per mg/kg-dayEmbryo: 4.6 × 103 mg/m3 (LOAEL)
CorannuleneToxics 12 00938 i011Lab- synthesized chemicalNot assessedNot assessedNot assessed
Polychlorinated biphenyl (PCBs)-IndustryFor adult exposure: 3.04 ng/kg-day indoor inhalation, 3.0 ± 2.2 ng/kg-day for dietary intake [73]1 µg/m3 (10-h time-weighted average) Not assessed
Lead (Pb)-Household and industryHuman daily intake from food: 0.1 to 0.3 µg/kg body weight/day [74]Not assessedBrain–intelligence: Blood lead level of <10 μg/dL in child study [75]
1 All human adverse-level concentration references were retrieved from the Integrated Risk Information System (IRIS) or Environmental Protection Agency (EPA) database; not assessed, and no references were found.

4. B(a)P and Gut Microbiome Interaction

The gut microbiome, a complex community of microorganisms in the gastrointestinal tract, plays a vital role in human health, including digestion, immune system regulation, and metabolic balance. Recent studies have explored how B(a)P exposure impacts the composition and diversity of this microbiome, potentially affecting its functionality and human health outcomes. The effects of B(a)P exposure on gut microbiome are summarized in Table 2.

4.1. B(a)P Exposure and Gut Dysbiosis

According to Table 2, although recent in vitro studies directly examining the relationship between B(a)P-induced gut microbiome dysbiosis and human health risks remain limited, existing research indicates that B(a)P exposure can alter the composition of the gut microbiome. These changes affect the volatile organic compounds or known as volatolome, which is linked to metabolic diseases. This suggests a potential pathway through which B(a)P exposure may contribute to metabolic disorders [18] and causing inflammatory bowel disease (IBD) through its impact on gut microbiome (discussed further in Section 4.2). Rodent studies have shown that B(a)P exposure leads to significant alterations in gut microbial composition, resulting in reduced alpha diversity. Ribiere et al. demonstrated that 28-day exposure to B(a)P (10 mL/kg BW) in murine models induced gut microbial shifts, decreasing Bacteroides, Parabacteroides, and Paraprevotella. Furthermore, He reported that long-term oral exposure of B(a)P at 50 mg/kg mice BW for 42 days induced gut microbiome alteration, especially the genera Faecalibaculum, Lactobacillus, Acinetobacter, Desulfovibrio, and Alistipes, which could induce colon inflammation. Similar to Du et al., the study evaluated long-term exposure in mice via oral gavage at low-dose B(a)P (50 u µg/mouse/day) for 23 days, resulting in gut dysbiosis, characterized by an increased abundance of Lachnospiraceae bacterium 3-2, Lachnospiraceae bacterium COE1, and Prevotella sp. MGM1, which is linked to allergic responses [45].
Studies on aquatic animals, including fish, scallops, and sea cucumbers, have shown that B(a)P exposure decreases alpha diversity while increasing the abundance of Fusobacteria, Proteobacteria, and Bacteroidetes [80,81,83]. These effects were observed through various exposure routes, including water exposure, feeding manipulation, and intraperitoneal injection. According to those studies, B(a)P and other environmental contaminants can exert a substantial influence on the gut microbiota, with their impact primarily contingent on the long duration and low concentration intensity of the host’s exposure to pollutants. These findings indicate a potential connection between B(a)P-induced gut dysbiosis and health risks. In particular, translational studies that bridge the gap between in vitro and in vivo research on human studies are needed to fully understand the health implications of B(a)P exposure. The current in vitro evidence provides a strong basis for exploring the effects of B(a)P on the gut microbiome in more advanced in vivo models and eventually human populations. This will require carefully designed human studies that functionally evaluate both B(a)P exposure and gut microbiome composition alongside key metabolic health markers. Such research is crucial to determine the true impact of B(a)P on human health and to inform strategies for minimizing potential risks.

4.2. Key Influential Factors for B(a)P-Induced Gut Microbiome Change

The B(a)P-associated gut microbiome changes may be linked to the lipophilic characteristics of B(a)P. As lipophilic substances, PAHs such as B(a)P can permeate the cytoplasmic membrane, increasing membrane fluidity and damaging bacterial cells, leading to bacterial death and an unbalanced gut composition [18,84]. The lipophilic nature of B(a)P influences its interaction with both Gram-positive and Gram-negative bacteria, although the structural differences between these bacteria types lead to some variations in their effects. In Gram-negative bacteria, B(a)P lipophilicity allows it to readily penetrate the outer membrane, which is rich in lipopolysaccharide. This can disrupt the outer membrane structure, potentially leading to the release of LPS, a potent endotoxin that triggers inflammation [85]. Furthermore, B(a)P can cross the inner membrane, affecting its fluidity and potentially damaging essential membrane proteins [86]. The periplasmic space, located between the outer and inner membranes, contains peptidoglycan, which can also be affected by B(a)P exposure, potentially triggering signaling through NOD receptors [87]. Despite lacking an outer membrane, Gram-positive bacteria possess a thick peptidoglycan layer external to their single-cell membrane. B(a)P’s lipophilic nature still allows it to penetrate this peptidoglycan layer and reach the cell membrane, where it can disrupt membrane fluidity and function, similar to its effects on Gram-negative bacteria. The absence of an outer membrane and LPS in Gram-positive bacteria means that B(a)P exposure will not lead to LPS release and the associated inflammatory effects seen in Gram-negative bacteria [88]. In both cases, B(a)P’s lipophilicity enables it to interact with and disrupt bacterial membranes, ultimately affecting cell viability and potentially contributing to gut dysbiosis.
Moreover, volatile organic compounds (VOCs) generated by the gut microbiota have been examined as potential contributors to pollutant-induced gut dysbiosis [18,89]. Six distinct VOCs, including benzaldehyde, 3-octane, 2-pentyl furan, butyl butanoate, 2-methyl-phenol, and 2-hexyl furan, were significantly detected in human fecal bacteria after a 24 h in vitro exposure [18]. De et al. observed shifts in metabolites within the benzenoid, ketone, and furan derivative categories, likely attributable to disruptions in typical bacterial ecology, particularly in pathologies such as inflammatory bowel disease (IBD) [90].
Sex differences in gut microbiota have been reported in B(a)P exposure studies using fish and mice as in vivo models. In juvenile fathead minnows, the gut microbiota community structure was notably changed in female fish but not in male fish after exposure to a low B(a)P dose [91]. These distinctions are ascribed to hormonal regulation, particularly the influence of androgen receptors. The gut microbiota controls estrogen levels by producing β-glucuronidase, which converts estrogens into active forms. Microbiota imbalance can disrupt this process, ultimately reducing circulating estrogen levels. Alterations in circulating estrogen levels can contribute to metabolic inflammation, leading to various diseases [92,93,94]. Beibei et al. demonstrated that oral administration of B(a)P in female mice led to increased susceptibility to allergic symptoms compared to male mice, associated with increases in specific bacterial species [45]. Although clinical studies comparing B(a)P exposure effects between males and females are limited, it is evident that pollutant exposure can disrupt the gut microbiome in a sex-specific manner, possibly influenced by hormonal regulation.

4.3. Microbiome-Related Health Risks of B(a)P

B(a)P has been shown to accumulate within the intestinal tract, resulting in significant alterations in the intricate balance of the gut microbiome. This intricate microbial ecosystem plays a pivotal role in maintaining human health, significantly influencing the development of various chronic diseases. These health conditions encompass a spectrum of disorders, ranging from localized intestinal issues such as colonic injury [33,77,95] to systemic diseases, including carcinogenic effects like lung cancer and colorectal cancer, cytotoxic responses linked to cancer, xenobiotic metabolism, and modulation of allergic responses [6,96].

4.3.1. Colonic Injury and Immunity Impairment

Sub-chronic oral exposure to B(a)P resulted in moderate inflammation, primarily affecting the ileal mucosa. In a murine study, this pollutant exposure led to changes in fecal and mucosa-associated microbiota composition [33]. Specifically, it reduced beneficial bacteria associated with short-chain fatty acids (SCFA), such as Clostridium and Coprococcus, from the Lachnospiraceae family. Conversely, there was an increase in the abundance of pro-inflammatory bacteria, such as Paraprevotellaceae, Turicibacter, and Desulfovibrionaceae, which are associated with colitis. Notably, Desulfovibrionaceae produces endotoxins and toxic sulfur, which can contribute to immune disorders and colon damage. A supporting study reported that B(a)P exposure in mice elevated metabolic pathways related to lipopolysaccharides (LPS) and sulfur compounds, along with increasing the presence of Desulfovibrionaceae [77]. Specific metabolites or elements of intestinal flora can cause immune cells to produce cytokines, which can modify the immunological response [97,98]. B(a)P treatment increased the prevalence of the pathogenic genus Staphylococcus, which is linked to increased inflammation and decreased immunomodulatory function of the intestines at the genus level [99]. Du et al. found that in mice exposed to intestinal allergic inflammation, the levels of inflammatory cytokines were linked to the presence of specific bacterial taxa, particularly Lachnospiraceae bacterium 28-4 and Alistipes inops [45]. This was paralleled by elevated allergic cytokines and intestinal permeability. The abundance of L. bacterium 3-2, L. bacterium COE1, and Prevotella sp. MGM1 positively correlated with the exacerbation of intestinal inflammation. These results suggest a correlation between inflammatory responses and specific bacterial taxa, indicating their potential role in gut health and immune response. A key area of concern is the link between environmental pollutants, chronic inflammation, and the loss of self-tolerance in autoimmune diseases. Pollutants can disrupt the gut microbiome, leading to dysbiosis and increased intestinal permeability. This can allow harmful substances, including bacterial components such as lipopolysaccharides, to enter the bloodstream, triggering systemic inflammation [100]. Chronic inflammation, in turn, can contribute to the breakdown of self-tolerance, where the immune system mistakenly attacks the body’s own tissues, characteristic of autoimmune diseases. Environmental factors, including xenobiotics, play a significant role in the development of autoimmunity, often acting as triggers in genetically susceptible individuals [101]. Further research is needed to elucidate the precise mechanisms by which pollutants contribute to autoimmunity in humans. Longitudinal studies assessing pollutant exposure, gut microbiome composition, inflammatory markers, and autoimmune disease development are crucial. This research will inform targeted interventions to mitigate the impact of environmental pollutants on human health, particularly in preventing or managing autoimmune diseases.

4.3.2. Carcinogenic and Xenobiotic Implications

In cancer research, the occurrence of particular bacterial strains (including Bifidobacterium, Intestinimonas, Alistipes, Odoribacter, and Acetatifactor) and the activation of specific metabolic pathways (including linoleic acid metabolism, unsaturated fatty acid production, and steroid hormone biosynthesis) were linked with the gut–lung axis correlation in a mouse model of lung cancer [71]. The gut microbiota can modulate the immune response in the lungs through various mechanisms, such as the activation of immune cells by T cells producing IL-17 and the production of cytokines and chemokines essential for regulating immune responses in the respiratory system [102,103,104]. Wu reported that B(a)P-induced colon cancer in mice showed significantly increased levels of Sphingobacteria, Gamma-proteobacteria, and Lactobacillales. These bacteria might help transform B(a)P into reactive metabolites that undergo enterohepatic circulation as they interact with gut microbiota, contributing to the reintroduction of reactive xenobiotic metabolites in the intestine [96]. Along with xenobiotic studies, Gentao et al. conducted the intraperitoneal exposure route B(a)P to rats at 100 mg/kg BW and showed there was a notable rise in the hepatic transcriptomic levels of carcinogenic biomarkers associated with xenobiotic metabolism influenced by the gut microbiome [78].

4.3.3. Metabolic Disturbance

Metabolic syndrome (MetS) is a risk factor for developing several complex human diseases, including type 2 diabetes, obesity, cardiovascular disorders, and cancer [7]. While specific studies on B(a)P’s effects on metabolic syndrome related to the gut microbiota are preliminary, previous research has shown that gut microbiota dysbiosis can lead to metabolic syndrome and is involved in bile acid metabolism, glucose, and energy homeostasis [105]. B(a)P exposure altered gut microbiome composition by decreasing Akkermansia muciniphila in murine models [33] and elevating the Firmicutes/Bacteroidetes ratio in various aquatic species. In mice treated with B(a)P, the abundance of Prevotella spp. was found to be significantly increased [45]. Dysbiosis of some colonizing bacteria, including Prevotella spp., has been associated with type 1 diabetes (T1D) [106]. Additionally, a higher Firmicutes/Bacteroidetes ratio, similar to that observed in obese animals or those fed a high-fat diet, is linked to metabolic disorders and weight gain [107,108]. This observation might be explained by the fact that Firmicutes are more efficient as an energy source than Bacteroidetes, resulting in more effective calorie absorption and subsequent weight gain.

5. Synergistic Toxic Effects of B(a)P and MPs

5.1. MPs as Vectors for B(a)P: Mechanisms, Environmental Fate, and Biological Impacts

The interactions between B(a)P and microplastics (MPs) are of growing concern in environmental science due to their potentially synergistic effects on ecosystems and human health [109]. MPs can act as vectors for contaminants such as B(a)P, disrupting gut microbiomes and causing various indirect hazards to organisms [110,111,112,113,114]. They have considerable adsorption capacity and can transport pollutants over long distances [115,116]. Shaoyong et al. found that B(a)P-loaded aged polystyrene MPs (PS-MPs) promote colonic barrier injury in mice, leading to increased permeability and inflammation mediated by oxidative stress-induced activation of the Notch signaling pathway [117]. The interaction between PS-MPs and B(a)P in acidic environments leads to excessive reactive oxygen species (ROS) production, causing synergistic toxic effects [118,119].
MPs can be ingested by aquatic animals, particularly fish, making them as suitable model organisms for studying the combined effects of MPs and B(a)P on health risks [120]. As MPs can adsorb and carry chemicals, including B(a)P, and deliver them to the host, simultaneous exposure to MPs and B(a)P may adversely affect organisms, particularly aquatic animals, as summarized in Table 3. The interaction between MPs and B(a)P is related to the binding affinity between these two substances. MPs can have varying binding affinity to B(a)P depending on the MP types, such as polystyrene, polyethylene, and polypropylene, and their sizes [121]. MPs/NPs have a strong binding affinity, which enables extensive interactions between them and the surrounding substances [122]. A study on mussels found that B(a)P concentrations increased over time, especially when sorbed to smaller MPs, and that MPs with sorbed B(a)P were more toxic than MPs alone [15]. The mobility of MPs depends on their size, with smaller MPs (<1 mm) showing high mobility in horizontal and vertical directions [123]. Smaller MPs are more mobile than larger MPs, which means that they can more easily penetrate biological barriers and accumulate in tissue, increasing their potential for toxicity [101,107,108].
PS-MPs have a higher binding affinity to B(a)P than other MPs, which might be attributed to their chemical properties. The PS-MP characteristics are also strongly influenced by the presence of the pendant phenyl (C6H5) groups, as the phenyl rings prevent the chains from rotating around the carbon–carbon bonds and give the polymer its well-known stiffness [108]. Another study also stated that the hydrophobicity of PS-MPs is due to its aromatic ring structure, which allows it to interact with other aromatic compounds, such as B(a)P [109,110,111]. Thus, the accumulation of MPs within cells can disrupt cellular functions and potentially lead to cytotoxicity. These findings emphasize the potential for the combined pollutants to exert substantial toxic effects on biological ecosystems.

5.2. Combined Effects of B(a)P and MPs on Gut Microbiome

The co-exposure of B(a)P and MPs potentially has more adverse effects on the gut microbiota compared to single exposure to B(a)P because the gut dysbiosis caused by MP exposure has been consistently reported in recent years. Exposure to MPs can cause changes in bacterial species, richness, beta diversity, and gut microbiota composition in various aquatic species, often reducing beneficial bacteria and increasing potential pathogens [133]. Usman et al. found that PS-MP exposure in Javanese medaka fish altered gut flora and caused metabolic abnormalities related to energy metabolism [134]. Also, recent studies have shown that MPs, particularly NPs, can affect the gut–brain axis by altering gut microbiota, intestinal barrier permeability, oxidative stress, inflammation, neurotoxicity, and behavioral disturbances in mice [135,136]. Yang et al. revealed that oral administration of NPs to mice influenced brain function by stimulating macrophage IL-1 signaling in the intestine [137]. Besides, MP ingestion in rats resulted in gut microbiome alterations at the family level (Muribaculaceae, Oscillospiraceae, Bacteroidaceae, Neisseriaceae, Prevotellaceae, and Veillonellaceae), affecting lipid metabolism and potentially impacting brain function and anxiety-related behaviors [138].
Although there are no direct studies of the combined effects of B(a)P and MPs on the gut microbiome, co-exposure to MPs and B(a)P may potentially induce gut microbiota dysbiosis by damaging the intestinal epithelium’s mucus layer, increasing intestinal permeability, and disrupting the gut microbiota barrier. This disruption can lead to changes in microbial diversity, composition, and metabolite profiles, as well as intestinal inflammation. While research in this field is still ongoing, studies examining the impact of MPs on gut microbiota have revealed an emerging and increasingly important area of research [139,140]. These findings highlight the potential health risks associated with exposure to B(a)P and MPs and the need for further research to fully understand the mechanisms underlying their toxic effects on the onset of various diseases linked with the gut microbiome in aquatic animals, humans, and other animals.
Research on B(a)P’s impact on the gut microbiome reveals both direct and indirect health effects, presenting a multifaceted risk profile (Figure 3).

6. Conclusions

In conclusion, B(a)P exposure can disrupt gut microbiota balance, leading to dysbiosis that can trigger gut inflammation and influence the host’s immune response, potentially promoting cancer development. Also, gut microbial dysbiosis due to B(a)P exposure may also aggravate metabolic processes, contributing to disorders such as obesity and insulin resistance. However, further in-depth analysis is needed to fully elucidate these complex relationships. Certain gut bacteria can metabolize B(a)P into toxic compounds, and shifts in microbiome composition may alter the balance of these metabolites, impacting cancer risk. Also, due to the strong reactivity of B(a)P with microparticles, B(a)P can act as a carrier of microplastics by binding to them, which have recently been demonstrated to be hazardous to the gut microbiome. As microbiome-related B(a)P research is still limited to several in vitro and in vivo studies, more approaches at different concentrations and treatment periods are needed, as well as combination studies with various microplastic materials and sizes. Most importantly, human studies will be required to determine whether the various B(a)P exposure levels result in differences in human gut microbiome.

Author Contributions

Conceptualization, I.R.M. and K.H.C.; validation, H.J.L.; formal analysis, I.R.M. and E.H.; investigation, A.N.T.K.; writing—original draft preparation, I.R.M.; writing—review and editing, S.Y.K., S.-M.L. and K.H.C.; supervision, K.H.C.; project administration, K.H.C.; funding acquisition, K.H.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Korea Institute of Planning and Evaluation for Technology in Food, Agriculture and Forestry (IPET) through the Agricultural Microbiome R&D Program for Advancing Innovative Technology, funded by the Ministry of Agriculture, Food and Rural Affairs (MAFRA) (RS-2024-00396964), and a National Research Foundation of Korea (NRF) grant, funded by the Korean government (MSIT) (2021R1C1C1007945).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data for the results presented in this article will be available upon request.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Wu, D.; Chen, L.; Ma, Z.; Zhou, D.; Fu, L.; Liu, M.; Zhang, T.; Yang, J.; Zhen, Q. Source analysis and health risk assessment of polycyclic aromatic hydrocarbon (PAHs) in total suspended particulate matter (TSP) from Bengbu, China. Sci. Rep. 2024, 14, 5080. [Google Scholar] [CrossRef] [PubMed]
  2. Wilson, M.J.; Frickel, S.; Nguyen, D.; Bui, T.; Echsner, S.; Simon, B.R.; Howard, J.L.; Miller, K.; Wickliffe, J.K. A targeted health risk assessment following the Deepwater Horizon oil spill: Polycyclic aromatic hydrocarbon exposure in Vietnamese-American shrimp consumers. Environ. Health Perspect. 2015, 123, 152–159. [Google Scholar] [CrossRef] [PubMed]
  3. Das, D.N.; Ravi, N. Influences of polycyclic aromatic hydrocarbon on the epigenome toxicity and its applicability in human health risk assessment. Environ. Res. 2022, 213, 113677. [Google Scholar] [CrossRef] [PubMed]
  4. Xue, W.; Warshawsky, D. Metabolic activation of polycyclic and heterocyclic aromatic hydrocarbons and DNA damage: A review. Toxicol. Appl. Pharmacol. 2005, 206, 73–93. [Google Scholar] [CrossRef]
  5. Chakravarti, D.; Venugopal, D.; Mailander, P.C.; Meza, J.L.; Higginbotham, S.; Cavalieri, E.L.; Rogan, E.G. The role of polycyclic aromatic hydrocarbon-DNA adducts in inducing mutations in mouse skin. Mutat. Res. 2008, 649, 161–178. [Google Scholar] [CrossRef]
  6. Vijay, A.; Valdes, A.M. Role of the gut microbiome in chronic diseases: A narrative review. Eur. J. Clin. Nutr. 2022, 76, 489–501. [Google Scholar] [CrossRef]
  7. Fan, Y.; Pedersen, O. Gut microbiota in human metabolic health and disease. Nat. Rev. Microbiol. 2021, 19, 55–71. [Google Scholar] [CrossRef]
  8. Wu, H.J.; Wu, E. The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes 2012, 3, 4–14. [Google Scholar] [CrossRef]
  9. Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef]
  10. Sun, Y.; Wang, X.; Li, L.; Zhong, C.; Zhang, Y.; Yang, X.; Li, M.; Yang, C. The role of gut microbiota in intestinal disease: From an oxidative stress perspective. Front. Microbiol. 2024, 15, 1328324. [Google Scholar] [CrossRef]
  11. Berenblum, I.; Schoental, R.; Brit, J. 3:4-Benzpyrene from Coal Tar. Nature 1945, 156, 601. [Google Scholar] [CrossRef]
  12. Diggs, D.L.; Harris, K.L.; Rekhadevi, P.V.; Ramesh, A. Tumor microsomal metabolism of the food toxicant, benzo(a)pyrene, in ApcMin mouse model of colon cancer. Tumour Biol. 2012, 33, 1255–1260. [Google Scholar] [CrossRef] [PubMed]
  13. Batel, A.; Linti, F.; Scherer, M.; Erdinger, L.; Braunbeck, T. Transfer of benzo[a]pyrene from microplastics to Artemia nauplii and further to zebrafish via a trophic food web experiment: CYP1A induction and visual tracking of persistent organic pollutants. Environ. Toxicol. Chem. 2016, 35, 1656–1666. [Google Scholar] [CrossRef] [PubMed]
  14. Batel, A.; Borchert, F.; Reinwald, H.; Erdinger, L.; Braunbeck, T. Microplastic accumulation patterns and transfer of benzo[a]pyrene to adult zebrafish (Danio rerio) gills and zebrafish embryos. Environ. Pollut. 2018, 235, 918–930. [Google Scholar] [CrossRef]
  15. Gonzalez-Soto, N.; Hatfield, J.; Katsumiti, A.; Duroudier, N.; Lacave, J.M.; Bilbao, E.; Orbea, A.; Navarro, E.; Cajaraville, M.P. Impacts of dietary exposure to different sized polystyrene microplastics alone and with sorbed benzo[a]pyrene on biomarkers and whole organism responses in mussels Mytilus galloprovincialis. Sci. Total Environ. 2019, 684, 548–566. [Google Scholar] [CrossRef]
  16. Buening, M.K.; Wislocki, P.G.; Levin, W.; Yagi, H.; Thakker, D.R.; Akagi, H.; Koreeda, M.; Jerina, D.M.; Conney, A.H. Tumorigenicity of the optical enantiomers of the diastereomeric benzo[a]pyrene 7,8-diol-9,10-epoxides in newborn mice: Exceptional activity of (+)-7beta,8alpha-dihydroxy-9alpha,10alpha-epoxy-7,8,9,10-tetrahydrobenzo[a]pyrene. Proc. Natl. Acad. Sci. USA 1978, 75, 5358–5361. [Google Scholar] [CrossRef]
  17. Conney, A.H.; Miller, E.C.; Miller, J.A. Substrate-induced synthesis and other properties of benzpyrene hydroxylase in rat liver. J. Biol. Chem. 1957, 228, 753–766. [Google Scholar] [CrossRef]
  18. Defois, C.; Ratel, J.; Denis, S.; Batut, B.; Beugnot, R.; Peyretaillade, E.; Engel, E.; Peyret, P. Environmental Pollutant Benzo[a]Pyrene Impacts the Volatile Metabolome and Transcriptome of the Human Gut Microbiota. Front. Microbiol. 2017, 8, 1562. [Google Scholar] [CrossRef]
  19. Denissenko, M.F.; Pao, A.; Tang, M.; Pfeifer, G.P. Preferential formation of benzo[a]pyrene adducts at lung cancer mutational hotspots in P53. Science 1996, 274, 430–432. [Google Scholar] [CrossRef]
  20. Fahey, J.W.; Haristoy, X.; Dolan, P.M.; Kensler, T.W.; Scholtus, I.; Stephenson, K.K.; Talalay, P.; Lozniewski, A. Sulforaphane inhibits extracellular, intracellular, and antibiotic-resistant strains of Helicobacter pylori and prevents benzo[a]pyrene-induced stomach tumors. Proc. Natl. Acad. Sci. USA 2002, 99, 7610–7615. [Google Scholar] [CrossRef]
  21. Gelboin, H.V. A microsome-dependent binding of benzo[a]pyrene to DNA. Cancer Res. 1969, 29, 1272–1276. [Google Scholar] [PubMed]
  22. Ha, Y.L.; Storkson, J.; Pariza, M.W. Inhibition of benzo(a)pyrene-induced mouse forestomach neoplasia by conjugated dienoic derivatives of linoleic acid. Cancer Res. 1990, 50, 1097–1101. [Google Scholar] [PubMed]
  23. Herbstman, J.B.; Tang, D.; Zhu, D.; Qu, L.; Sjodin, A.; Li, Z.; Camann, D.; Perera, F.P. Prenatal exposure to polycyclic aromatic hydrocarbons, benzo[a]pyrene-DNA adducts, and genomic DNA methylation in cord blood. Environ. Health Perspect. 2012, 120, 733–738. [Google Scholar] [CrossRef]
  24. Huberman, E.; Sachs, L. Cell susceptibility to transformation and cytotoxicity by the carcinogenic hydrocarbon benzo[a]pyrene. Proc. Natl. Acad. Sci. USA 1966, 56, 1123–1129. [Google Scholar] [CrossRef]
  25. Huberman, E.; Sachs, L.; Yang, S.K.; Gelboin, V. Identification of mutagenic metabolites of benzo(a)pyrene in mammalian cells. Proc. Natl. Acad. Sci. USA 1976, 73, 607–611. [Google Scholar] [CrossRef]
  26. Schneider, J.; Grosser, R.; Jayasimhulu, K.; Xue, W.; Warshawsky, D. Degradation of Pyrene, Benz[a]anthracene, and Benzo[a]pyrene by Mycobacterium sp. Strain RJGII-135, Isolated from a Former Coal Gasification Site. Appl. Environ. Microbiol. 1996, 62, 13–19. [Google Scholar] [CrossRef]
  27. Kazerouni, N.; Sinha, R.; Hsu, C.H.; Greenberg, A.; Rothman, N. Analysis of 200 food items for benzo[a]pyrene and estimation of its intake in an epidemiologic study. Food Chem. Toxicol. 2001, 39, 423–436. [Google Scholar] [CrossRef]
  28. Lu, P.Q.; Jeong, H.; Jankowiak, R.; Small, G.J.; Kim, S.K.; Cosman, M.; Gracintov, N.E. Comparative laser spectroscopic study of DNA and polynucleotide adducts from the (+)-anti-diol epoxide of benzo[a]pyrene. Chem. Res. Toxicol. 1991, 4, 58–69. [Google Scholar] [CrossRef]
  29. Miller, E.C. Studies on the formation of protein-bound derivatives of 3,4-benzpyrene in the epidermal fraction of mouse skin. Cancer Res. 1951, 11, 100–108. [Google Scholar]
  30. Cosman, M.; De Los Santos, C.; Fiala, R.; Hingerty, B.E.; Singh, S.B.; Ibanez, V.; Margulis, L.A.; Live, D.; Geacintov, N.E.; Broyde, S. Solution conformation of the major adduct between the carcinogen (+)-anti-benzo[a]pyrene diol epoxide and DNA. Proc. Natl. Acad. Sci. USA 1992, 89, 1914–1918. [Google Scholar] [CrossRef]
  31. Phillips, D.H. Fifty years of benzo(a)pyrene. Nature 1983, 303, 468–472. [Google Scholar] [CrossRef] [PubMed]
  32. Hannah, R.; Ramani, P.; Ramanathan, A.; Gheena, S.; Ramasubramanian, A.; Monika, K. CYP2 C9 polymorphism among patients with oral squamous cell carcinoma and its role in altering the metabolism of benzo[a]pyrene. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2020, 130, 306–312. [Google Scholar] [CrossRef]
  33. Ribiere, C.; Peyret, P.; Parisot, N.; Darcha, C.; Dechelotte, P.J.; Barnich, N.; Peyretaillade, E.; Boucher, D. Oral exposure to environmental pollutant benzo[a]pyrene impacts the intestinal epithelium and induces gut microbial shifts in murine model. Sci. Rep. 2016, 6, 31027. [Google Scholar] [CrossRef]
  34. Sims, P.; Grover, P.L.; Swaisland, A.; Pal, K.; Hewer, A. Metabolic activation of benzo(a)pyrene proceeds by a diol-epoxide. Nature 1974, 252, 326–328. [Google Scholar] [CrossRef]
  35. Sparnins, V.L.; Barany, G.; Wattenberg, L.W. Effects of organosulfur compounds from garlic and onions on benzo[a]pyrene-induced neoplasia and glutathione S-transferase activity in the mouse. Carcinogenesis 1988, 9, 131–134. [Google Scholar] [CrossRef]
  36. Sun, Y.; Zhou, Q.; Xu, Y.; Wang, L.; Liang, X. Phytoremediation for co-contaminated soils of benzo[a]pyrene (B[a]P) and heavy metals using ornamental plant Tagetes patula. J. Hazard. Mater. 2011, 186, 2075–2082. [Google Scholar] [CrossRef]
  37. Toyama, T.; Furukawa, T.; Maeda, N.; Inoue, D.; Sei, K.; Mori, K.; Kikuchi, S.; Ike, M. Accelerated biodegradation of pyrene and benzo[a]pyrene in the Phragmites australis rhizosphere by bacteria-root exudate interactions. Water Res. 2011, 45, 1629–1638. [Google Scholar] [CrossRef]
  38. Uno, S.; Dalton, T.P.; Derkenne, S.; Curran, C.P.; Miller, M.L.; Shertzer, H.G.; Nebert, D.W. Oral exposure to benzo[a]pyrene in the mouse: Detoxication by inducible cytochrome P450 is more important than metabolic activation. Mol. Pharmacol. 2004, 65, 1225–1237. [Google Scholar] [CrossRef]
  39. Uno, S.; Dalton, T.P.; Dragin, N.; Curran, C.P.; Derkenne, S.; Miller, M.L.; Shertzer, H.G.; Gonzalez, F.J.; Nebert, D.W. Oral benzo[a]pyrene in Cyp1 knockout mouse lines: CYP1A1 important in detoxication, CYP1B1 metabolism required for immune damage independent of total-body burden and clearance rate. Mol. Pharmacol. 2006, 69, 1103–1114. [Google Scholar] [CrossRef]
  40. Wattenberg, L.W.; Leong, J.L. Inhibition of the carcinogenic action of benzo(a)pyrene by flavones. Cancer Res. 1970, 30, 1922–1925. [Google Scholar]
  41. Shimizu, Y.; Nakatsuru, Y.; Ichinose, M.; Takahashi, Y.; Kume, H.; Mimura, J.; Fujii-Kuriyama, Y.; Ishikawa, T. Benzo[a]pyrene carcinogenicity is lost in mice lacking the aryl hydrocarbon receptor. Proc. Natl. Acad. Sci. USA 2000, 97, 4. [Google Scholar] [CrossRef] [PubMed]
  42. Weigert, F.; Mottram, J.C. The Biochemistry of Benzpyrene II. The Course of Its Metabolism and the Chemical Nature of the Metabolites. Cancer Res. 1946, 6, 109–120. [Google Scholar] [PubMed]
  43. Zhao, Y.; Liu, H.; Wang, Q.; Li, B.; Zhang, H.; Pi, Y. The effects of benzo[a]pyrene on the composition of gut microbiota and the gut health of the juvenile sea cucumber Apostichopus japonicus Selenka. Fish Shellfish Immunol. 2019, 93, 369–379. [Google Scholar] [CrossRef] [PubMed]
  44. DeBofsky, A.; Xie, Y.; Grimard, C.; Alcaraz, A.J.; Brinkmann, M.; Hecker, M.; Giesy, J.P. Differential responses of gut microbiota of male and female fathead minnow (Pimephales promelas) to a short-term environmentally-relevant, aqueous exposure to benzo[a]pyrene. Chemosphere 2020, 252, 126461. [Google Scholar] [CrossRef]
  45. Du, B.; Xiao, X.; Wang, H.; Li, W.; Xia, Z.; Yang, P.; Huang, S.K.; Yuan, R.; Liu, J.; Han, M.; et al. Evaluation of the Impact of BaP Exposure on the Gut Microbiota and Allergic Responses in an OVA-Sensitized Mouse Model. Environ. Health Perspect. 2023, 131, 67004. [Google Scholar] [CrossRef]
  46. Motwani, H.V.; Westberg, E.; Tornqvist, M. Interaction of benzo[a]pyrene diol epoxide isomers with human serum albumin: Site specific characterisation of adducts and associated kinetics. Sci. Rep. 2016, 6, 36243. [Google Scholar] [CrossRef]
  47. Reed, L.; Arlt, V.M.; Phillips, D.H. The role of cytochrome P450 enzymes in carcinogen activation and detoxication: An in vivo-in vitro paradox. Carcinogenesis 2018, 39, 851–859. [Google Scholar] [CrossRef]
  48. Sulc, M.; Indra, R.; Moserova, M.; Schmeiser, H.H.; Frei, E.; Arlt, V.M.; Stiborova, M. The impact of individual cytochrome P450 enzymes on oxidative metabolism of benzo[a]pyrene in human livers. Environ. Mol. Mutagen. 2016, 57, 229–235. [Google Scholar] [CrossRef]
  49. Einaudi, L.; Courbiere, B.; Tassistro, V.; Prevot, C.; Sari-Minodier, I.; Orsiere, T.; Perrin, J. In vivo exposure to benzo(a)pyrene induces significant DNA damage in mouse oocytes and cumulus cells. Hum. Reprod. 2014, 29, 548–554. [Google Scholar] [CrossRef]
  50. Bukowska, B.; Duchnowicz, P. Molecular Mechanisms of Action of Selected Substances Involved in the Reduction of Benzo[a]pyrene-Induced Oxidative Stress. Molecules 2022, 27, 1379. [Google Scholar] [CrossRef]
  51. Guo, B.; Feng, D.; Xu, Z.; Qi, P.; Yan, X. Acute benzo[a]pyrene exposure induced oxidative stress, neurotoxicity and epigenetic change in blood clam Tegillarca granosa. Sci. Rep. 2021, 11, 18744. [Google Scholar] [CrossRef] [PubMed]
  52. Shields, H.J.; Traa, A.; Van Raamsdonk, J.M. Beneficial and Detrimental Effects of Reactive Oxygen Species on Lifespan: A Comprehensive Review of Comparative and Experimental Studies. Front. Cell Dev. Biol. 2021, 9, 628157. [Google Scholar] [CrossRef] [PubMed]
  53. Juan, C.A.; Perez de la Lastra, J.M.; Plou, F.J.; Perez-Lebena, E. The Chemistry of Reactive Oxygen Species (ROS) Revisited: Outlining Their Role in Biological Macromolecules (DNA, Lipids and Proteins) and Induced Pathologies. Int. J. Mol. Sci. 2021, 22, 4642. [Google Scholar] [CrossRef] [PubMed]
  54. Davis, R.L.A.a.J.M. Oxygen Toxicity and Reactive Oxygen Species: The Devil Is in the Details. Pediatr. Res. 2009, 66, 2009. [Google Scholar]
  55. Shi, Q.; Fijten, R.R.; Spina, D.; Riffo Vasquez, Y.; Arlt, V.M.; Godschalk, R.W.; Van Schooten, F.J. Altered gene expression profiles in the lungs of benzo[a]pyrene-exposed mice in the presence of lipopolysaccharide-induced pulmonary inflammation. Toxicol. Appl. Pharmacol. 2017, 336, 8–19. [Google Scholar] [CrossRef]
  56. Vazquez-Gomez, G.; Rocha-Zavaleta, L.; Rodriguez-Sosa, M.; Petrosyan, P.; Rubio-Lightbourn, J. Benzo[a]pyrene activates an AhR/Src/ERK axis that contributes to CYP1A1 induction and stable DNA adducts formation in lung cells. Toxicol. Lett. 2018, 289, 54–62. [Google Scholar] [CrossRef]
  57. Coelho, N.R.; Pimpao, A.B.; Correia, M.J.; Rodrigues, T.C.; Monteiro, E.C.; Morello, J.; Pereira, S.A. Pharmacological blockage of the AHR-CYP1A1 axis: A call for in vivo evidence. J. Mol. Med. 2022, 100, 215–243. [Google Scholar] [CrossRef]
  58. Larigot, L.; Juricek, L.; Dairou, J.; Coumoul, X. AhR signaling pathways and regulatory functions. Biochim. Open 2018, 7, 1–9. [Google Scholar] [CrossRef]
  59. Granados, J.C.; Falah, K.; Koo, I.; Morgan, E.W.; Perdew, G.H.; Patterson, A.D.; Jamshidi, N.; Nigam, S.K. AHR is a master regulator of diverse pathways in endogenous metabolism. Sci. Rep. 2022, 12, 16625. [Google Scholar] [CrossRef]
  60. Uppstad, H.; Ovrebo, S.; Haugen, A.; Mollerup, S. Importance of CYP1A1 and CYP1B1 in bioactivation of benzo[a]pyrene in human lung cell lines. Toxicol. Lett. 2010, 192, 221–228. [Google Scholar] [CrossRef]
  61. Shiizaki, K.; Kawanishi, M.; Yagi, T. Modulation of benzo[a]pyrene-DNA adduct formation by CYP1 inducer and inhibitor. Genes Environ. 2017, 39, 14. [Google Scholar] [CrossRef] [PubMed]
  62. Androutsopoulos, V.P.; Tsatsakis, A.M.; Spandidos, D.A. Cytochrome P450 CYP1A1: Wider roles in cancer progression and prevention. BMC Cancer 2009, 9, 187. [Google Scholar] [CrossRef] [PubMed]
  63. Bukowska, B.; Mokra, K.; Michalowicz, J. Benzo[a]pyrene-Environmental Occurrence, Human Exposure, and Mechanisms of Toxicity. Int. J. Mol. Sci. 2022, 23, 6348. [Google Scholar] [CrossRef]
  64. Maurice, N.M.; Sadikot, R.T. Mitochondrial Dysfunction in Bacterial Infections. Pathogens 2023, 12, 1005. [Google Scholar] [CrossRef]
  65. Pellegrino, M.W.; Nargund, A.M.; Kirienko, N.V.; Gillis, R.; Fiorese, C.J.; Haynes, C.M. Mitochondrial UPR-regulated innate immunity provides resistance to pathogen infection. Nature 2014, 516, 414–417. [Google Scholar] [CrossRef]
  66. Zhu, L.; Zhou, Q.; He, L.; Chen, L. Mitochondrial unfolded protein response: An emerging pathway in human diseases. Free Radic. Biol. Med. 2021, 163, 125–134. [Google Scholar] [CrossRef]
  67. Montano, L.; Pironti, C.; Pinto, G.; Ricciardi, M.; Buono, A.; Brogna, C.; Venier, M.; Piscopo, M.; Amoresano, A.; Motta, O. Polychlorinated Biphenyls (PCBs) in the Environment: Occupational and Exposure Events, Effects on Human Health and Fertility. Toxics 2022, 10, 365. [Google Scholar] [CrossRef]
  68. Mallah, M.A.; Mallah, M.A.; Liu, Y.; Xi, H.; Wang, W.; Feng, F.; Zhang, Q. Relationship Between Polycyclic Aromatic Hydrocarbons and Cardiovascular Diseases: A Systematic Review. Front. Public Health 2021, 9, 763706. [Google Scholar] [CrossRef]
  69. Booc, F.; Thornton, C.; Lister, A.; MacLatchy, D.; Willett, K.L. Benzo[a]pyrene effects on reproductive endpoints in Fundulus heteroclitus. Toxicol. Sci. 2014, 140, 73–82. [Google Scholar] [CrossRef]
  70. Hu, X.; Geetha, R.V.; Surapaneni, K.M.; Veeraraghavan, V.P.; Chinnathambi, A.; Alahmadi, T.A.; Manikandan, V.; Manokaran, K. Lung cancer induced by Benzo(A)Pyrene: ChemoProtective effect of sinapic acid in swiss albino mice. Saudi J. Biol. Sci. 2021, 28, 7125–7133. [Google Scholar] [CrossRef]
  71. Qu, Z.; Zhang, L.; Hou, R.; Ma, X.; Yu, J.; Zhang, W.; Zhuang, C. Exposure to a mixture of cigarette smoke carcinogens disturbs gut microbiota and influences metabolic homeostasis in A/J mice. Chem. Biol. Interact. 2021, 344, 109496. [Google Scholar] [CrossRef] [PubMed]
  72. Zhang, X.; Yang, L.; Zhang, H.; Xing, W.; Wang, Y.; Bai, P.; Zhang, L.; Hayakawa, K.; Toriba, A.; Wei, Y.; et al. Assessing Approaches of Human Inhalation Exposure to Polycyclic Aromatic Hydrocarbons: A Review. Int. J. Environ. Res. Public Health 2021, 18, 3124. [Google Scholar] [CrossRef] [PubMed]
  73. Weitekamp, C.A.; Phillips, L.J.; Carlson, L.M.; DeLuca, N.M.; Cohen Hubal, E.A.; Lehmann, G.M. A state-of-the-science review of polychlorinated biphenyl exposures at background levels: Relative contributions of exposure routes. Sci. Total Environ. 2021, 776, 145912. [Google Scholar] [CrossRef] [PubMed]
  74. Zhang, T.; Zhang, Y.; Li, W.; Wang, L.; Jiao, Y.; Wang, Y.; Jiang, D.; Gao, X. Occurrence and dietary exposure of heavy metals in marketed vegetables and fruits of Shandong Province, China. Food Sci. Nutr. 2021, 9, 5166–5173. [Google Scholar] [CrossRef] [PubMed]
  75. Jusko, T.A.; Henderson, C.R.; Lanphear, B.P.; Cory-Slechta, D.A.; Parsons, P.J.; Canfield, R.L. Blood lead concentrations < 10 microg/dL and child intelligence at 6 years of age. Environ. Health Perspect. 2008, 116, 243–248. [Google Scholar] [CrossRef]
  76. Li, P.; Jiang, Y.; Fei, Y.Y.; Zeng, X.C. Dynamic Variation of Camel Gastrointestinal Bacterial Communities Contributing to Benzo(a)pyrene Degradation. Appl. Biochem. Microbiol. 2022, 58, 796–805. [Google Scholar] [CrossRef]
  77. He, S.; Li, X.; Li, C.; Deng, H.; Shao, Y.; Yuan, L. Isoorientin attenuates benzo[a]pyrene-induced colonic injury and gut microbiota disorders in mice. Food Res. Int. 2019, 126, 108599. [Google Scholar] [CrossRef]
  78. Li, G.; Xiong, H.; Saeed, K.; Ma, R.; Xing, Y.; Bi, Y.; Li, C.; Huang, J.; Zhang, Y. Comparative toxicity analysis of corannulene and benzo[a]pyrene in mice. Toxicol Lett 2020, 331, 130–142. [Google Scholar] [CrossRef]
  79. Garcia, W.L.; Miller, C.J.; Lomas, G.X.; Gaither, K.A.; Tyrrell, K.J.; Smith, J.N.; Brandvold, K.R.; Wright, A.T. Profiling How the Gut Microbiome Modulates Host Xenobiotic Metabolism in Response to Benzo[a]pyrene and 1-Nitropyrene Exposure. Chem. Res. Toxicol. 2022, 35, 585–596. [Google Scholar] [CrossRef]
  80. DeBofsky, A.; Xie, Y.; Challis, J.K.; Jain, N.; Brinkmann, M.; Jones, P.D.; Giesy, J.P. Responses of juvenile fathead minnow (Pimephales promelas) gut microbiome to a chronic dietary exposure of benzo[a]pyrene. Environ. Pollut. 2021, 278, 116821. [Google Scholar] [CrossRef]
  81. Quintanilla-Mena, M.; Vega-Arreguin, J.; Del Rio-Garcia, M.; Patino-Suarez, V.; Peraza-Echeverria, S.; Puch-Hau, C. The effect of benzo[a]pyrene on the gut microbiota of Nile tilapia (Oreochromis niloticus). Appl. Microbiol. Biotechnol. 2021, 105, 7935–7947. [Google Scholar] [CrossRef] [PubMed]
  82. Stagaman, K.; Kasschau, K.D.; Tanguay, R.L.; Sharpton, T.J. Experimental methods modestly impact interpretation of the effect of environmental exposures on the larval zebrafish gut microbiome. Sci. Rep. 2022, 12, 14538. [Google Scholar] [CrossRef] [PubMed]
  83. Li, D.; Miao, J.; Pan, L.; Zhou, Y.; Gao, Z.; Yang, Y.; Xu, R.; Zhang, X. Impacts of benzo(a)pyrene exposure on scallop (Chlamys farreri) gut health and gut microbiota composition. Sci. Total Environ. 2021, 799, 149471. [Google Scholar] [CrossRef] [PubMed]
  84. Murinova, S.; Dercova, K. Response mechanisms of bacterial degraders to environmental contaminants on the level of cell walls and cytoplasmic membrane. Int. J. Microbiol. 2014, 2014, 873081. [Google Scholar] [CrossRef]
  85. Wang, X.; Quinn, P.J. Lipopolysaccharide: Biosynthetic pathway and structure modification. Prog. Lipid Res. 2010, 49, 97–107. [Google Scholar] [CrossRef]
  86. Hardonniere, K.; Saunier, E.; Lemarie, A.; Fernier, M.; Gallais, I.; Helies-Toussaint, C.; Mograbi, B.; Antonio, S.; Benit, P.; Rustin, P.; et al. The environmental carcinogen benzo[a]pyrene induces a Warburg-like metabolic reprogramming dependent on NHE1 and associated with cell survival. Sci. Rep. 2016, 6, 30776. [Google Scholar] [CrossRef]
  87. Almeida-da-Silva, C.L.C.; Savio, L.E.B.; Coutinho-Silva, R.; Ojcius, D.M. The role of NOD-like receptors in innate immunity. Front. Immunol. 2023, 14, 1122586. [Google Scholar] [CrossRef]
  88. Zhou, S.; Chen, G.; Qi, M.; El-Assaad, F.; Wang, Y.; Dong, S.; Chen, L.; Yu, D.; Weaver, J.C.; Beretov, J.; et al. Gram Negative Bacterial Inflammation Ameliorated by the Plasma Protein Beta 2-Glycoprotein I. Sci. Rep. 2016, 6, 33656. [Google Scholar] [CrossRef]
  89. Defois, C.; Ratel, J.; Garrait, G.; Denis, S.; Le Goff, O.; Talvas, J.; Mosoni, P.; Engel, E.; Peyret, P. Food Chemicals Disrupt Human Gut Microbiota Activity and Impact Intestinal Homeostasis as Revealed by In Vitro Systems. Sci. Rep. 2018, 8, 11006. [Google Scholar] [CrossRef]
  90. De Preter, V.; Joossens, M.; Ballet, V.; Shkedy, Z.; Rutgeerts, P.; Vermeire, S.; Verbeke Phd, K. Metabolic profiling of the impact of oligofructose-enriched inulin in Crohn’s disease patients: A double-blinded randomized controlled trial. Clin. Transl. Gastroenterol. 2013, 4, e30. [Google Scholar] [CrossRef]
  91. d’Afflitto, M.; Upadhyaya, A.; Green, A.; Peiris, M. Association Between Sex Hormone Levels and Gut Microbiota Composition and Diversity—A Systematic Review. J. Clin. Gastroenterol. 2022, 56, 384–392. [Google Scholar] [CrossRef] [PubMed]
  92. Monteiro, R.; Teixeira, D.; Calhau, C. Estrogen signaling in metabolic inflammation. Mediat. Inflamm. 2014, 2014, 615917. [Google Scholar] [CrossRef] [PubMed]
  93. Hotamisligil, G.S. Inflammation and metabolic disorders. Nature 2006, 444, 860–867. [Google Scholar] [CrossRef] [PubMed]
  94. Mathis, D.; Shoelson, S.E. Immunometabolism: An emerging frontier. Nat. Rev. Immunol. 2011, 11, 81. [Google Scholar] [CrossRef] [PubMed]
  95. Wang, Z.; Shi, L.; Li, H.; Song, W.; Li, J.; Yuan, L. Selenium-Enriched Black Soybean Protein Prevents Benzo(a)pyrene-Induced Pyroptotic Colon Damage and Gut Dysbacteriosis. J. Agric. Food Chem. 2022, 70, 12629–12640. [Google Scholar] [CrossRef]
  96. Wu, J.C.; Tsai, M.L.; Lai, C.S.; Lo, C.Y.; Ho, C.T.; Wang, Y.J.; Pan, M.H. Polymethoxyflavones prevent benzo[a]pyrene/dextran sodium sulfate-induced colorectal carcinogenesis through modulating xenobiotic metabolism and ameliorate autophagic defect in ICR mice. Int. J. Cancer 2018, 142, 1689–1701. [Google Scholar] [CrossRef]
  97. Csak, T.; Ganz, M.; Pespisa, J.; Kodys, K.; Dolganiuc, A.; Szabo, G. Fatty acid and endotoxin activate inflammasomes in mouse hepatocytes that release danger signals to stimulate immune cells. Hepatology 2011, 54, 133–144. [Google Scholar] [CrossRef]
  98. Ganz, M.; Csak, T.; Nath, B.; Szabo, G. Lipopolysaccharide induces and activates the Nalp3 inflammasome in the liver. World J. Gastroenterol. 2011, 17, 4772–4778. [Google Scholar] [CrossRef]
  99. Xie, S.; Zhou, A.; Xu, N.; Feng, Y.; Pan, Z.; Junaid, M.; Wang, J.; Zou, J. Benzo[a]pyrene induces microbiome dysbiosis and inflammation in the intestinal tracts of western mosquitofish (Gambusia affinis) and zebrafish (Danio rerio). Fish Shellfish Immunol. 2020, 105, 24–34. [Google Scholar] [CrossRef]
  100. Mostafavi Abdolmaleky, H.; Zhou, J.R. Gut Microbiota Dysbiosis, Oxidative Stress, Inflammation, and Epigenetic Alterations in Metabolic Diseases. Antioxidants 2024, 13, 985. [Google Scholar] [CrossRef]
  101. Khan, M.F.; Wang, H. Environmental Exposures and Autoimmune Diseases: Contribution of Gut Microbiome. Front. Immunol. 2019, 10, 3094. [Google Scholar] [CrossRef] [PubMed]
  102. Sencio, V.; Machado, M.G.; Trottein, F. The lung-gut axis during viral respiratory infections: The impact of gut dysbiosis on secondary disease outcomes. Mucosal. Immunol. 2021, 14, 296–304. [Google Scholar] [CrossRef] [PubMed]
  103. Qu, L.; Cheng, Q.; Wang, Y.; Mu, H.; Zhang, Y. COPD and Gut-Lung Axis: How Microbiota and Host Inflammasome Influence COPD and Related Therapeutics. Front. Microbiol. 2022, 13, 868086. [Google Scholar] [CrossRef] [PubMed]
  104. Chunxi, L.; Haiyue, L.; Yanxia, L.; Jianbing, P.; Jin, S. The Gut Microbiota and Respiratory Diseases: New Evidence. J. Immunol. Res. 2020, 2020, 2340670. [Google Scholar] [CrossRef]
  105. Carrizales-Sanchez, A.K.; Garcia-Cayuela, T.; Hernandez-Brenes, C.; Senes-Guerrero, C. Gut microbiota associations with metabolic syndrome and relevance of its study in pediatric subjects. Gut Microbes 2021, 13, 1960135. [Google Scholar] [CrossRef]
  106. Liu, J.; Zhou, L.; Sun, L.; Ye, X.; Ma, M.; Dou, M.; Shi, L. Association Between Intestinal Prevotella copri Abundance and Glycemic Fluctuation in Patients with Brittle Diabetes. Diabetes Metab. Syndr. Obes. 2023, 16, 1613–1621. [Google Scholar] [CrossRef]
  107. Callahan, B.J.; McMurdie, P.J.; Rosen, M.J.; Han, A.W.; Johnson, A.J.; Holmes, S.P. DADA2: High-resolution sample inference from Illumina amplicon data. Nat. Methods 2016, 13, 581–583. [Google Scholar] [CrossRef]
  108. Magne, F.; Gotteland, M.; Gauthier, L.; Zazueta, A.; Pesoa, S.; Navarrete, P.; Balamurugan, R. The Firmicutes/Bacteroidetes Ratio: A Relevant Marker of Gut Dysbiosis in Obese Patients? Nutrients 2020, 12, 1474. [Google Scholar] [CrossRef]
  109. Lu, L.; Luo, T.; Zhao, Y.; Cai, C.; Fu, Z.; Jin, Y. Interaction between microplastics and microorganism as well as gut microbiota: A consideration on environmental animal and human health. Sci. Total Environ. 2019, 667, 94–100. [Google Scholar] [CrossRef]
  110. Liao, X.; Zhao, P.; Hou, L.; Adyari, B.; Xu, E.G.; Huang, Q.; Hu, A. Network analysis reveals significant joint effects of microplastics and tetracycline on the gut than the gill microbiome of marine medaka. J. Hazard. Mater. 2023, 442, 129996. [Google Scholar] [CrossRef]
  111. Zhang, Z.; Chen, W.; Chan, H.; Peng, J.; Zhu, P.; Li, J.; Jiang, X.; Zhang, Z.; Wang, Y.; Tan, Z.; et al. Polystyrene Microplastics Induce Size-Dependent Multi-Organ Damage in Mice: Insights into Gut Microbiota and Fecal Metabolites. J. Hazard. Mater. 2023, 461, 132503. [Google Scholar] [CrossRef] [PubMed]
  112. Fournier, E.; Ratel, J.; Denis, S.; Leveque, M.; Ruiz, P.; Mazal, C.; Amiard, F.; Edely, M.; Bezirard, V.; Gaultier, E.; et al. Exposure to polyethylene microplastics alters immature gut microbiome in an infant in vitro gut model. J. Hazard. Mater. 2023, 443, 130383. [Google Scholar] [CrossRef] [PubMed]
  113. Guo, X.; Lv, M.; Li, J.; Ding, J.; Wang, Y.; Fu, L.; Sun, X.; Han, X.; Chen, L. The distinct toxicity effects between commercial and realistic polystyrene microplastics on microbiome and histopathology of gut in zebrafish. J. Hazard. Mater. 2022, 434, 128874. [Google Scholar] [CrossRef] [PubMed]
  114. Caruso, G. Microplastics as vectors of contaminants. Mar. Pollut. Bull. 2019, 146, 921–924. [Google Scholar] [CrossRef]
  115. Koelmans, A.A.; Bakir, A.; Burton, G.A.; Janssen, C.R. Microplastic as a Vector for Chemicals in the Aquatic Environment: Critical Review and Model-Supported Reinterpretation of Empirical Studies. Environ. Sci. Technol. 2016, 50, 3315–3326. [Google Scholar] [CrossRef]
  116. Fu, L.; Li, J.; Wang, G.; Luan, Y.; Dai, W. Adsorption behavior of organic pollutants on microplastics. Ecotoxicol. Environ. Saf. 2021, 217, 112207. [Google Scholar] [CrossRef]
  117. Shaoyong, W.; Jin, H.; Jiang, X.; Xu, B.; Liu, Y.; Wang, Y.; Jin, M. Benzo [a] pyrene-loaded aged polystyrene microplastics promote colonic barrier injury via oxidative stress-mediated notch signalling. J. Hazard. Mater. 2023, 457, 131820. [Google Scholar] [CrossRef]
  118. Katsumiti, A.; Losada-Carrillo, M.P.; Barros, M.; Cajaraville, M.P. Polystyrene nanoplastics and microplastics can act as Trojan horse carriers of benzo(a)pyrene to mussel hemocytes in vitro. Sci. Rep. 2021, 11, 22396. [Google Scholar] [CrossRef]
  119. Zou, H.; Qu, H.; Bian, Y.; Sun, J.; Wang, T.; Ma, Y.; Yuan, Y.; Gu, J.; Bian, J.; Liu, Z. Polystyrene Microplastics Induce Oxidative Stress in Mouse Hepatocytes in Relation to Their Size. Int. J. Mol. Sci. 2023, 24, 7382. [Google Scholar] [CrossRef]
  120. Ziani, K.; Ionita-Mindrican, C.B.; Mititelu, M.; Neacsu, S.M.; Negrei, C.; Morosan, E.; Draganescu, D.; Preda, O.T. Microplastics: A Real Global Threat for Environment and Food Safety: A State of the Art Review. Nutrients 2023, 15, 617. [Google Scholar] [CrossRef]
  121. Amelia, T.S.M.; Khalik, W.M.A.W.M.; Ong, M.C.; Shao, Y.T.; Pan, H.-J.; Bhubalan, K. Marine microplastics as vectors of major ocean pollutants and its hazards to the marine ecosystem and humans. Prog. Earth Planet. Sci. 2021, 8, 12. [Google Scholar] [CrossRef]
  122. Cao, J.; Yang, Q.; Jiang, J.; Dalu, T.; Kadushkin, A.; Singh, J.; Fakhrullin, R.; Wang, F.; Cai, X.; Li, R. Coronas of micro/nano plastics: A key determinant in their risk assessments. Part. Fibre Toxicol. 2022, 19, 55. [Google Scholar] [CrossRef] [PubMed]
  123. Zhang, X.; Chen, Y.; Li, X.; Zhang, Y.; Gao, W.; Jiang, J.; Mo, A.; He, D. Size/shape-dependent migration of microplastics in agricultural soil under simulative and natural rainfall. Sci. Total Environ. 2022, 815, 152507. [Google Scholar] [CrossRef] [PubMed]
  124. Wang, Y.; Zhang, M.; Ding, G.; Shi, H.; Cong, Y.; Li, Z.; Wang, J. Polystyrene microplastics alleviate adverse effects of benzo[a]pyrene on tissues and cells of the marine mussel, Mytilus galloprovincialis. Aquat. Toxicol. 2023, 256, 106430. [Google Scholar] [CrossRef]
  125. Bebianno, M.J.; Mendes, V.M.; O’Donovan, S.; Carteny, C.C.; Keiter, S.; Manadas, B. Effects of microplastics alone and with adsorbed benzo(a)pyrene on the gills proteome of Scrobicularia plana. Sci. Total Environ. 2022, 842, 156895. [Google Scholar] [CrossRef]
  126. Romdhani, I.; De Marco, G.; Cappello, T.; Ibala, S.; Zitouni, N.; Boughattas, I.; Banni, M. Impact of environmental microplastics alone and mixed with benzo[a]pyrene on cellular and molecular responses of Mytilus galloprovincialis. J. Hazard. Mater. 2022, 435, 128952. [Google Scholar] [CrossRef]
  127. Rodrigues, A.R.; Mestre, N.C.C.; Fonseca, T.G.D.; Pedro, P.Z.; Carteny, C.C.; Cormier, B.; Keiter, S.; Bebianno, M.J. Influence of Particle Size on Ecotoxicity of Low-Density Polyethylene Microplastics, with and without Adsorbed Benzo-a-Pyrene, in Clam Scrobicularia plana. Biomolecules 2022, 12, 78. [Google Scholar] [CrossRef]
  128. von Hellfeld, R.; Zarzuelo, M.; Zaldibar, B.; Cajaraville, M.P.; Orbea, A. Accumulation, Depuration, and Biological Effects of Polystyrene Microplastic Spheres and Adsorbed Cadmium and Benzo(a)pyrene on the Mussel Mytilus galloprovincialis. Toxics 2022, 10, 18. [Google Scholar] [CrossRef]
  129. Martinez-Alvarez, I.; Le Menach, K.; Devier, M.H.; Cajaraville, M.P.; Budzinski, H.; Orbea, A. Screening of the Toxicity of Polystyrene Nano- and Microplastics Alone and in Combination with Benzo(a)pyrene in Brine Shrimp Larvae and Zebrafish Embryos. Nanomaterials 2022, 12, 941. [Google Scholar] [CrossRef]
  130. Coffin, S.; Magnuson, J.T.; Vliet, S.M.F.; Volz, D.C.; Schlenk, D. Effects of short-term exposure to environmentally-relevant concentrations of benzo(a)pyrene-sorbed polystyrene to White seabass (Atractoscion nobilis)☆. Environ. Pollut. 2020, 263, 114617. [Google Scholar] [CrossRef]
  131. Abouda, S.; Missawi, O.; Cappello, T.; Boughattas, I.; De Marco, G.; Maisano, M.; Banni, M. Toxicological impact of environmental microplastics and benzo[a]pyrene in the seaworm Hediste diversicolor under environmentally relevant exposure conditions. Environ. Pollut. 2022, 310, 119856. [Google Scholar] [CrossRef] [PubMed]
  132. Su, Y.; Qi, H.; Hou, Y.; Gao, M.; Li, J.; Cai, M.; Zhu, X.; Chen, M.; Ge, C.; Fu, D.; et al. Combined Effects of Microplastics and Benzo[a]pyrene on the Marine Diatom Chaetoceros muelleri. Front. Mar. Sci. 2022, 8, 779321. [Google Scholar] [CrossRef]
  133. Li, W.; Chen, X.; Li, M.; Cai, Z.; Gong, H.; Yan, M. Microplastics as an aquatic pollutant affect gut microbiota within aquatic animals. J. Hazard. Mater. 2022, 423, 127094. [Google Scholar] [CrossRef]
  134. Usman, S.; Razis, A.F.A.; Shaari, K.; Azmai, M.N.A.; Saad, M.Z.; Isa, N.M.; Nazarudin, M.F. Polystyrene microplastics induce gut microbiome and metabolome changes in Javanese medaka fish (Oryzias javanicus Bleeker, 1854). Toxicol. Rep. 2022, 9, 1369–1379. [Google Scholar] [CrossRef]
  135. Kang, H.; Zhang, W.; Jing, J.; Huang, D.; Zhang, L.; Wang, J.; Han, L.; Liu, Z.; Wang, Z.; Gao, A. The gut-brain axis involved in polystyrene nanoplastics-induced neurotoxicity via reprogramming the circadian rhythm-related pathways. J. Hazard. Mater. 2023, 458, 131949. [Google Scholar] [CrossRef]
  136. Grodzicki, W.; Dziendzikowska, K.; Gromadzka-Ostrowska, J.; Kruszewski, M. Nanoplastic Impact on the Gut-Brain Axis: Current Knowledge and Future Directions. Int. J. Mol. Sci. 2021, 22, 12795. [Google Scholar] [CrossRef]
  137. Yang, Q.; Dai, H.; Cheng, Y.; Wang, B.; Xu, J.; Zhang, Y.; Chen, Y.; Xu, F.; Ma, Q.; Lin, F.; et al. Oral feeding of nanoplastics affects brain function of mice by inducing macrophage IL-1 signal in the intestine. Cell Rep. 2023, 42, 112346. [Google Scholar] [CrossRef]
  138. Zhao, N.; Zhao, M.; Jin, H. Microplastic-induced gut microbiota and serum metabolic disruption in Sprague-Dawley rats. Environ. Pollut. 2023, 320, 121071. [Google Scholar] [CrossRef]
  139. Chen, Y.; Xu, J.; Chen, Y. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients 2021, 13, 2099. [Google Scholar] [CrossRef]
  140. Hou, K.; Wu, Z.X.; Chen, X.Y.; Wang, J.Q.; Zhang, D.; Xiao, C.; Zhu, D.; Koya, J.B.; Wei, L.; Li, J.; et al. Microbiota in health and diseases. Signal Transduct. Target. Ther. 2022, 7, 135. [Google Scholar] [CrossRef]
Figure 1. Publication trends in polycyclic aromatic hydrocarbon (PAH) research (to facilitate data interpretation above, the paper numbers were collected from PubMed and Scopus and represented graphically in the heatmap, stack bar, and diagram generated using GraphPad Prism 10.1.2).
Figure 1. Publication trends in polycyclic aromatic hydrocarbon (PAH) research (to facilitate data interpretation above, the paper numbers were collected from PubMed and Scopus and represented graphically in the heatmap, stack bar, and diagram generated using GraphPad Prism 10.1.2).
Toxics 12 00938 g001
Figure 2. Research history on the health risks of benzo(a)pyrene (B(a)P). Blue text indicates studies on benzo(a)pyrene’s carcinogenic mechanisms, green text represents research on the risks of simultaneous exposure to this substance and microplastics, and red text denotes studies on its effects on the gut microbiome [5,12,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42].
Figure 2. Research history on the health risks of benzo(a)pyrene (B(a)P). Blue text indicates studies on benzo(a)pyrene’s carcinogenic mechanisms, green text represents research on the risks of simultaneous exposure to this substance and microplastics, and red text denotes studies on its effects on the gut microbiome [5,12,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42].
Toxics 12 00938 g002
Figure 3. (a) A schematic illustration depicting the metabolic pathway of B(a)P (i) elucidating its enterohepatic circulation and association with cancer development involving diverse bacterial interactions and (ii) delineating its toxicity, which induces damage to bacterial cells, subsequently contributing to immune disorders and resultant diseases. (b) Illustration of (i) several types of MP/NP characteristics on combined adsorption with B(a)P and (ii) mechanism of micro-/nanoplastic (MP/NP) co-exposure with B(a)P and single exposure of MPs/NPs on gut microbiome disturbances which induces reactive oxygen species (ROS) production, increasing antioxidant activity and causing several chronic diseases in different organs. Yet the co-exposure MPs and B(a)P study effects on gut microbiota require further research, this co-exposure may also damage gut microbiome cells potentially leading to dysbiosis.
Figure 3. (a) A schematic illustration depicting the metabolic pathway of B(a)P (i) elucidating its enterohepatic circulation and association with cancer development involving diverse bacterial interactions and (ii) delineating its toxicity, which induces damage to bacterial cells, subsequently contributing to immune disorders and resultant diseases. (b) Illustration of (i) several types of MP/NP characteristics on combined adsorption with B(a)P and (ii) mechanism of micro-/nanoplastic (MP/NP) co-exposure with B(a)P and single exposure of MPs/NPs on gut microbiome disturbances which induces reactive oxygen species (ROS) production, increasing antioxidant activity and causing several chronic diseases in different organs. Yet the co-exposure MPs and B(a)P study effects on gut microbiota require further research, this co-exposure may also damage gut microbiome cells potentially leading to dysbiosis.
Toxics 12 00938 g003
Table 2. B(a)P exposure effects on alterations in the gut microbial community.
Table 2. B(a)P exposure effects on alterations in the gut microbial community.
Species TargetLife StageCo-Exposure PollutantPollutants ConcentrationExposure
Type
Exposure PeriodIntestinal ImpactsRef.
In vitro
Human microbiome--0.005, 0.05, and 0.5 mg/mLHuman fecal culture in bio-fermenter24 h↓ Microbial volatile in a dose-dependent manner
↓ Transcript level of bacterial chemotaxis toward simple carbohydrate pathways
[18]
Camel rumen and rectum--50 mg/LRumen and gut microbes culture20 daysKlebsiella sp., Ochrobactrum sp., and Bacillus sp. showed particular function as B(a)P degradation[76]
In vivo (mice)
C57BL/6 mice5-week-old (20–25 g)-10 mL/kg B.W.Oral gavage28 daysBacteroides, Parabacteroides, and Paraprevotella >27 days
↓ Lactobacillus, A. muciniphila, and Verrumicrobiaceae
[33]
BALB/c mice treated with Isoorientin5-week-old-50 mg/kg BWOral gavage42 days↑ Desulfovibrio, Acinetobacter, Odoribacter, and Veillonella in B(a)P group
Faecalibaculum and
Lactobacillus in B(a)P + ISO group
[77]
C57BL/6 mice and male Sprague Dawley (S.D.) rats8-week-old mice (22 ± 2 g) and (220–250 g) ratsCorannulene100 mg/kg BW COR or B(a)POral gavage and intraperitoneally injection3 daysBacteroidetes after I.P. injection of B(a)P and COR
Bacteroidetes after oral gavage B(a)P and COR
Actinobacteria ↓ by oral COR but ↑ by oral B(a)P
[78]
Conventional C57BL/6NTac and germ-free C57Bl/6GFTac mice7-week-old1-Nitropyrene180 mg/kg BW./day B(a)POral gavage72 hActive P450s enzyme in the liver is impacted by the presence of the gut microbiome, which is modified by PAH metabolism[79]
C57BL/6 mice (SPF) treated with Ovalbumin5–6 weeks old-50 ug/ mouse/ dayOral gavage23 daysB. virosa and N. subflava, and
B. uniformis and L. bacterium COE1 in B(a)P group
L. bacterium 3-2, L. bacterium COE1, and Prevotella sp. MGM1 in OVA group
[45]
In vivo (Aquatic animals)
Female and male Fathead minnow (Pimephales promelas)Adult (two years)-1.3, 4.0, and 12.0 mg/LWater immersion4 days↑ Alpha diversity in the female group compared to the male group
Vibrionaceae, the only abundant family in the male group
[44]
Fathead minnow (Pimephales promelas)Juvenile (2.5-month-old)-1, 10, 100, or 1000 u µg/g in food (DM)Feeding2 weeks↓ Alpha diversity, ↑ pathogenic taxa (Erysipelotrichaceae, Moraxellaceae, and Caulobacteraceae)[80]
Nile tilapia (Oreochromis niloticus)Juvenile (125.6 ± 41.4 g)-20 mg/kg B.W.Intraperitoneal injection24, 72, and 120 h post-injection↑ Fusobacteria and Bacteroidetes in <24 h
↓ Proteobacteria and Spirochaetae in >24 h
GM recovered after 72 h and was stable at 120 h post-injection
[81]
Zebrafish (Danio rerio)Embryos (9 days post fertilization)-1, 5, and 10 μMEmbryos Incubation test—with different dissection methods 9 daysGut microbiota significantly altered based on dose-dependent[82]
Scallop (Chlamys farreri)(5.7 ± 0.3 cm in length)-0, 0.4, 2 and 10 μg/LWater immersion7, 14 and 21 days↓ Alpha diversity, ↑ pathogenic bacteria Mycoplasma and Tenacibaculum. Hydrocarbon-degrading bacteria were found: Pseudomonas, Polaribacter, Amphritea, and Kordiimonas[83]
Sea cucumbers (Apostichopus japonicus)Juvenile (5.36 ± 0.14 g)-0, 0.5, 5, and 25 μg/LWater immersion14 days↑ Ratio of Bacteroidetes to Firmicutes[43]
Table 3. B(a)P and MP co-exposure effects on aquatic animals.
Table 3. B(a)P and MP co-exposure effects on aquatic animals.
Species TargetMPs TypeMPs SizeMPs Binding Affinity 1Pollutant Concentrations and Exposure MethodExposure PeriodToxicity EffectsRef.
Marine mussels
(M. galloprovincialis)
Green fluorescent polystyrene10 µm***5.5 µg/L MPs + 0.1 µg/L B(a)P in water immersion5 days↓ mRNA expression of NF- κB in gills
↑ strong affinity adsorption of B(a)P to PS-MPs
↓ The uptake and toxicity of B(a)P
[124]
Clam
(Scrobicularia plana)
Low-density polyethylene 11–13 µm-MPs with B(a)P adsorbed at one mg/L in water immersion 14 daysChanges in protein expression of the cytoskeleton, cell structure, oxidative stress, energy metabolism, and DNA binding also induce changes in glucose metabolism, RNA binding, and apoptosis[125]
Marine mussels
(M. galloprovincialis)
Environmental mixture MPs: polyethylene, polyethylene terephthalate, polypropylene, polyethylene vinyl acetate, and high-density polyethylene <100 µm-50 µg/L MP +
1 µg/L B(a)P in water immersion
1 and 3 daysInduced the apoptosis process:
↑ DNA ligase on day 1
↑ Bax, Cas-3, and P53 and on day 3
↓ Bcl-2 and DNA ligase on day 3
[126]
Clam
(Scrobicularia plana)
Low-density polyethylene 4–6 μm and 20–25 μm-1 mg/L MP + 16.64 ± 87 µg/g B(a)P in water immersion7 and 14 days4–6 μm-sized MPs resulted in more significant alterations in oxidative stress biomarkers [127]
Marine mussels
(M. galloprovincialis)
Polystyrene pristine4.5 and 45 µm***(0.05, 5, 50 µg/L MPs) + 252.3 µg/L B(a)P and Cd3 days↑ PS shows a higher affinity to B(a)P than Cd
MPs and B(a)P group induced histological alteration in digestive glands
[128]
Brine shrimp larvae and zebrafish embryosPolystyrene50 and 500 nm***0.069–6.87 mg/L PS + 0.1–10 mg/L B(a)P 24–48 h↑ malformation prevalence in the highest concentration of MPs and B(a)P groups in zebrafish; meanwhile, NPs were successful vectors to B(a)P in brine shrimp[129]
White seabassPolystyrene2.00–2.83 mm***100 g/2 L PS + 1µg/L B(a)P and single dose of 1 µg/L B(a)P and 252 µg/ B(a)P in water immersion5 daysSingle dose of 252 µg/ B(a)P group; fish exposed to polystyrene B(a)P-absorbed polystyrene show significant variations in the observed cellular or behavioral parameters compared to the control group [130]
Seaworm
(Hediste diversicolor)
Environmental mixture MPs: polyethylene, polyethylene terephthalate, polypropylene, polyethylene vinyl acetate, and high-density polyethylene >3, 3.0–1.22, 1.22–0.45 µm-1 mg/kg (sediment) of environmental MPs + 1 μg/kg (sediment) B[a]P3 and 7 days↑ cytotoxic and genotoxic damage in the co-exposure and single groups after 7 days.[131]
Marine diatom
(Chaetoceros muelleri)
Polyethylene terephthalate--200 mg/L PET + (10, 150 µg/L B(a)P) in medium culture1, 5, 15 days↑ SOD and MDA on day 1 and 5
↓ SOD and MDA on day 15
MPs and 10 µg/L B(a)P group showed higher antagonistic effects to the marine diatom.
[132]
1 Microplastics’ binding strength to pollutants, measured by sorption tests, is indicated (-, no sorption test was conducted; ***, the strong binding affinity when tested).
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

Mauliasari, I.R.; Lee, H.J.; Koo, S.Y.; Hitayezu, E.; Kieu, A.N.T.; Lee, S.-M.; Cha, K.H. Benzo(a)pyrene and Gut Microbiome Crosstalk: Health Risk Implications. Toxics 2024, 12, 938. https://doi.org/10.3390/toxics12120938

AMA Style

Mauliasari IR, Lee HJ, Koo SY, Hitayezu E, Kieu ANT, Lee S-M, Cha KH. Benzo(a)pyrene and Gut Microbiome Crosstalk: Health Risk Implications. Toxics. 2024; 12(12):938. https://doi.org/10.3390/toxics12120938

Chicago/Turabian Style

Mauliasari, Intan Rizki, Hee Ju Lee, Song Yi Koo, Emmanuel Hitayezu, Anh Nguyen Thi Kieu, Sang-Min Lee, and Kwang Hyun Cha. 2024. "Benzo(a)pyrene and Gut Microbiome Crosstalk: Health Risk Implications" Toxics 12, no. 12: 938. https://doi.org/10.3390/toxics12120938

APA Style

Mauliasari, I. R., Lee, H. J., Koo, S. Y., Hitayezu, E., Kieu, A. N. T., Lee, S.-M., & Cha, K. H. (2024). Benzo(a)pyrene and Gut Microbiome Crosstalk: Health Risk Implications. Toxics, 12(12), 938. https://doi.org/10.3390/toxics12120938

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