2,471
Views
8
CrossRef citations to date
0
Altmetric
Review

Gut microbiota: a non-target victim of pesticide-induced toxicity

ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Article: 2187578 | Received 15 Sep 2022, Accepted 01 Mar 2023, Published online: 15 Mar 2023

ABSTRACT

The human gut microbiota can be potentially disrupted due to exposure of various environmental contaminants, including pesticides. These contaminants enter into non-target species in multiple ways and cause potential health risks. The gut microbiota-derived metabolites have a significant role in maintaining the host’s health by regulating metabolic homeostasis. An imbalance in this homeostasis can result in the development of various diseases and their pathogenesis. Pesticides have hazardous effects on the host’s gut microbiota, which is evident in a few recent studies. Therefore, there is an urgent need to explore the effect of pesticide on gut microbiota-mediated metabolic changes in the host, which may provide a better understanding of pesticide-induced toxicity. The present review summarizes the pesticide-induced effects on gut microbiota, which in turn, induces changes in the release of their secondary metabolites that could lead to various host health effects.

1. Introduction

Currently, there is an increased demand for agricultural production due to a significant increase in the growing population globally. Thus, the use of pesticides and fertilizers has become an impediment to meeting the increased demand for crops with the rise in population. However, the overuse of these pesticides increases the presence of pesticide residues in agricultural products, which is a severe risk to human health and the environmentCitation1. The widespread and inevitable use of pesticides threatens non-target species, including humans. Being the top consumer in the food chain, humans are the most affected non-target species by pesticide exposure. Various studies have reported pesticide levels in both human blood and urine, which are further found to be associated with multiple diseases, including cancer, diabetes, hormonal disorders, asthma, metabolic diseases, and neurotoxicityCitation2–5. Furthermore, prolonged exposure to pesticides was also found to have teratogenic effects resulting in a reduction in birth weight, abnormal growth, diseases, and mortalityCitation6,Citation7. Most available studies focused on the association between pesticide exposure and host health effects. Still, there is a paucity of studies focused on elucidating the mechanism involving the adverse impact of pesticides on human health.

In recent years, the research attention is shifted toward the effect of pesticide exposure on the host gut microbiotaCitation8–11. The host gut microbiota is a secondary endocrine system that maintains the host’s normal biological function through hormone secretion and immune regulationCitation12,Citation13. Altered gut microbiota may result in various diseases, including obesity, colon cancer, diabetes, and other metabolic diseases. It is also evident that pesticides could promote host metabolic disorders by disrupting gut microbiota functionCitation8,Citation10,Citation14,Citation15. This review summarized the relationship between pesticide exposure, leading to alterations in the gut microbiota and their secondary metabolites release on host health effects, as well as the mechanism(s) by which different types of persistent organic pollutants induce changes in microbiota composition and function.

2. Gut microbiota

The ‘gut microbiota’ composed of bacteria, archaea, virus and eukarya, which are colonized in the Gastrointestinal (GI) tractCitation16. They live as a complex community in the digestive tracts of humans and animals, as well as insects. It is estimated that in an average human lifetime, around 60 tons of food pass through the GI tract of humans, along with an abundance of microorganisms; many environmental factors like pesticides have a massive threat to gut integrityCitation17.

2.1. Host gut microbiota and its metabolite

Around 10Citation14 microorganisms with more than 500 species are present in the gut of adult humans, contributing to multiple biochemical and physiological activities of the hostCitation18. Gut microbiota comprises bacteria, archaea, viruses, and eukaryotic microbes, most of which are anaerobic microorganisms residing in the human GI tractCitation19. In the gut of healthy adults human, Firmicutes and Bacteroidetes are the superabundant bacterial phylum of human gut microbiota, in addition to the Proteobacteria, Verrucomicrobia, Actinomycetes, Fusobacteria, and CyanobacteriaCitation18,Citation20,Citation21. Gut microbiota has a significant role in the expansion and divergence of host intestinal epithelium, educating the host immune system, providing protection against pathogens, and maintaining intestinal homeostasisCitation22. Various studies have demonstrated that gut microbiota regulates physiological function by releasing secondary metabolitesCitation23. The gut microbiota’s secondary metabolites include short-chain fatty acids (SCFAs), lipopolysaccharides (LPS), trimethylamine (TMA), branch-chained amino acids (BCAAs), bile acids (BAs), tryptophan, trimethylamine trioxide, and indolesCitation24. LPS is one of the components released into the intestinal luminal environment upon bacterial death and lysis, which disrupts the host immune system and the blood coagulation systemCitation25. Due to the oxidation of flavin-containing monooxygenase 3 (Fmo3), TMA gets converted into trimethylamine oxide (TMAO), and is a critical risk factor for cardiovascular and cerebrovascular diseasesCitation26. BAs are the key player in the body’s energy metabolism by promoting digestion and absorptionCitation27. SCFAs are a chief fuel for intestinal epithelial cells and strengthen the gut barrier and immune-modulatory function via influencing GPR41/43Citation28. Many indoles are produced by tryptophan metabolism, which maintains the intestinal barrier homeostasis and regulates the intestinal immune responseCitation28.

2.2. Regulatory mechanism of host gut microbiota

The function of gut microbiota is categorized into two aspects: 1. As a barrier in providing immunity and 2. to provide nutrition through metabolism. Additionally, gut microbiota also provides an indispensable barrier system to the body against pathogens via maintaining the steadiness of intestinal immunityCitation29. Gut microbiota impedes the host immune system’s induction, training, and proper functioning of the host immune system. Under normal circumstances, gut microbiota composition is relatively stable until disturbed by a strong external intrusion. Once the composition of gut microbiota gets altered (dysbiosis), it may lead to endangering health conditions with an increase in the colonization of pathogenic bacteriaCitation29–31. Gut microbiota induces the activation of αβ-T cells by influencing immune pathway-related genesCitation32. Further, it was also reported that gut microbiota regulates the inflammatory response via modulating TH17 cells in the small intestineCitation33–35.

In normal physiological conditions, the colon is mainly involved in water and mineral absorption. Further, the gut microbiota is vital in digesting undigestable fiber/carbohydrates to produce short-chain fatty acids, including butyrates. Due to this gut microbiota’s metabolic activity, the host’s epithelial cells obtain butyrate as an energy source required for growth and multiplicationCitation36. A study revealed that the composition of the gut microbiota of obese mice is entirely different from non-obese mice. However, there is a significant increase in the energy absorption capacity of the gut microbiota present in the host mice with obese conditionsCitation36,Citation37. The SCFAs produced through fermentation of undigestable starch by the gut microbiota have significant importance on the host’s normal physiological function by entering peripheral circulation and providing more energy through metabolizationCitation38,Citation39. SCFAs also affect the metabolism of fats, and carbohydrates, by activating the acetyl CoA synthase in the liverCitation39. In a nutshell, the gut microbiota has a crucial role in controlling the function of the gut barrier, immune system, and host metabolic activity. In , we have represented the host gut microbiota regulatory mechanism.

Figure 1. Graphical representation of the effect of pesticides and subsequent outcomes on gut microbiota. The figure represents different mechanisms that could alter gut microbiota composition and its metabolites induced via pesticide toxicity. The dysbiosis led by pesticide toxicity could adversely affect the host’s health through various known and unknown pathways.

Figure 1. Graphical representation of the effect of pesticides and subsequent outcomes on gut microbiota. The figure represents different mechanisms that could alter gut microbiota composition and its metabolites induced via pesticide toxicity. The dysbiosis led by pesticide toxicity could adversely affect the host’s health through various known and unknown pathways.

3. Role of persistent organic pollutants in the modulation of gut microbiota

Due to the lipophilic nature of persistent organic pollutants (POPs), these compounds are resistant to excrete out from the body once they enter into the body systemCitation40. Few POPs are naturally occurring, whereas most are man-made synthetic compounds for agricultural and industrial use. The accumulation of these compounds in the body system is hazardous to human health and is linked with immune dysfunction, reproductive and neurological disordersCitation41–44. In recent years, harmful effects of POPs, which include polychlorinated biphenyl (PCB), polycyclic aromatic hydrocarbon (PAH), pesticides, insecticides, herbicides, and heavy metals, are recognized due to their effect on gut microbiota modulation and are extensively studied in terms of their toxic effects in human, and mice gut microbiotaCitation40 .

3.1. Polychlorinated (PCBs)

These compounds were banned in the United States three decades ago. However, PCBs are persisted in the environment and continue to contaminate food products. The primary route of exposure to PCB in the host biological system is through ingestion. The mice exposed to PCB showed a significant impact on gastrointestinal physiology and gut microbiotaCitation45. In addition, maternal exposure to PCB in mice significantly increased the gut permeability as well as the expression of gene Toll-like receptor 4 (Tlr4) and its signaling pathway in the colon of offspringCitation45. Bacteroidetes and Firmicutes are two classes of beneficial bacteria in the human gut. Moreover, exposure to PCB increased the risk of developing obesity by altering the Firmicutes to Bacteroidetes ratio in mice and humansCitation12,Citation46. In addition, PCB exposure was associated with increased inflammation in the intestine in an age-dependent manner which is directly proportional to the abundance of proteobacteriaCitation47,Citation48. However, more studies are required to conclude the mechanism behind PCB’s role on gut microbiota and ways to minimize these effects to decrease gut permeability and inflammation.

3.2. Polyaromatic hydrocarbons (PAH)

Polyaromatic hydrocarbons (PAH) gain less attention for their associated impact on gut microbiota than PCB. However, an in vitro study has shown that PAH exposure resulted in colonic microbiota transformation via acquiring estrogenic activityCitation47. Also, microbial PAH transformation was confirmed by detecting its metabolite 1-hydroxypyrene and 7-hydroxybenzopyrene in the colonCitation49. However, more studies will be needed to determine the possible effects of PAH on the gut microbiota.

3.3. Pesticides

Pesticide is a broad term which includes herbicides, insecticides, and fungicides. These pesticides are extensively used globally to increase crop production. These compounds are believed to be toxic to target species, but their residue on food products simultaneously affects non-target species, including humans, via an endocrine-disrupting mechanismCitation50. Furthermore, recent studies have shown that the parent compound and their respective metabolites also cause adverse consequences to the environment and humans, especially the gutCitation15,Citation51. Recently, pesticide exposure and its effect on gut microbiota have gained more attention due to their increased use in crop production. Several studies concluded that pesticide exposure significantly alters the composition of gut microbiota, hence leading to a change in the metabolic profile of the hostCitation1,Citation52.

3.3.1. Organochlorine pesticides (OCPs)

Despite the ban on the usage of OCPs during the last three decades in several countries, their levels are continuously detected in the human body, which is a significant threat to human healthCitation53,Citation54. Among all the OCPs, dichlorodiphenyltrichloroethane (DDT) and hexachlorocyclohexane are major compounds that can potentially metabolize to b-hexachlorocyclohexane and p’p’ dechlorodiphenyldichloro-ethylene (p’p’DDE), which are major breakout detrimental products in the environment. A recent finding has shown that serum OCP level in the general population is positively correlated with gut methanobacteriales and obesityCitation55,Citation56. Similarly, another study also demonstrated that the chronic exposure (8 weeks) of mice with β-HCH and DDT resulted in an alteration of the relative abundance and composition of gut microbiota by increasing Bacteroidetes and reducing Proteobacteria, Deferribacteres, and Cyanobacteria with increased quantity of the Lactobacillus strain with bile salt hydrolase activity. Significantly these alterations influenced the hepatic and bile acid hydrophobicityCitation27.

Further, exposure of mice with β-HCH and DDT led to activation of de novo bile acid synthesis and inhibited the ileal bile acid reabsorption in mice. Findings from these studies are alarming and draw attention to the relationship of OCPs with the alterations in gut microbiota that need to be explored to illustrate further this relationship’s role in the pathogenesis of the diseaseCitation27,Citation57.

3.3.2. Organophosphate pesticides (OPPs)

This category of insecticides are derived from amides and phosphoric acid. Due to their biodegradable nature, OPPs are the most frequently and commonly used insecticide worldwide. The residues of these compounds are found on plant surfaces, air, water, and soil, from which these compounds get leached out to the human system. OPPs residues in human plasma samples are found to be positively associated with diabetes. The gut microbial-mediated degradation of OPP alters the esterase activities and acetate, which ultimately induces gluconeogenesis and glucose intoleranceCitation58. Another study supports the findings and demonstrates that chronic exposure (180 days) of mice to OPPs induces glucose intolerance via the breakdown of OPP to acetic acid through the mechanism of gluconeogenesis by gut microbiotaCitation58.

The most extensively studied and widely used OPP is chlorpyrifos (CPF)Citation59. Various in vitro and in-vivo studies revealed the ill effects of CPFCitation60–62. The in-vitro model for the intestinal environment is the SHIME®, where the CPF exposure is found to be associated with decreased number of Bifidobacterium and Lactobacillus (beneficial bacteria; considered to be probiotics), accompanied by an increased number of Enterococcus and Bacteroides, which ultimately leads to the change in pH and stimulation of SCFAsCitation63–65. Similarly, the effects of CPF on the Caco-2/TC7 cells were studied in vitro, and CPF could alter the mucosal barrier and cause inflammation through the divulging of IL-8. Although the above-mentioned in vitro models provide significant findings, there is a lack of microbiota and host crosstalkCitation64.

Another study demonstrated that long-term exposure to CPF in mice resulted in increased gut absorbency and inhibited the expression of tight junction proteins, like claudin-1, ZO-1, and occludin in the epithelial lining of ileum and colon. Further, mice gut microbiota showed an increment in the Proteobacteria phylum and decreased Bacteroidetes phylum upon exposure to chlorpyrifosCitation15.

Exposure to adult zebrafish with CPF resulted in alteration in the gut microbiota and metabolismCitation66. It was observed that CPF alters the gut microbiota in rats by changing the abundance of bacteria associated with diabetes and obesityCitation67. Whereas treatment of mice with a 1 mg/kg body weight dose of CPF for 30 days resulted in alterations in the gut microbiota with changes in urine metabolites, amino acids, metabolite, SCFAs, and bile acids elicited the intestinal inflammation and abnormal permeability of the intestineCitation52. Further, treatment of pregnant rats until weaning with CPF revealed gut microbial dysbiosis in puppies, which can further result in impairment in the mucosal barrier (mucin 2) and an increment in bacterial translocation. Additionally, perinatal exposure of pups with CPF has a potential effect on the delayed maturation of intestines in pups and alters the development of the immune systemCitation63,Citation68. Finally, CPF exposure decreases hepatic glucose and lipid metabolism via increased oxidative stress and microbiota dysbiosis in animal modelsCitation66.

Diazinon is another OPP that raised public health concerns due to its extensive usage in agriculture. Its level is significantly traceable in drinking water, the primary route/source for human exposure to DiazinonCitation69–71. Treatment of either sex mice with a four-ppm dose diazinon for 13 weeks caused gut microbiota dysbiosis, impaired energy metabolism, and activated multitudinous stress response pathways in male mice compared to female miceCitation72,Citation73. Exposure of rats for 14 days to diazinon led to induced histological changes and elevated the Bacteroidetes, Firmicutes, and Fusobacteria phyla in the gut of ratsCitation74. Malathion is one of the OPP known to alter gut metabolite (decrease taurine and glycine) and is also associated with increased inflammationCitation75. All the above findings provide new insight into the relationship between gut microbiota and disease pathogenesis due to diazinon exposure.

3.3.3. Herbicides

Herbicides are widely used as weed killers, but their residue on food grains like Maize/Soya bean potentially affects non-target species like humans causing harmful health effects. Likewise, traces of insecticides and herbicides are also detected in grains, fruits, and vegetables used for human consumption. Other significant sources of human exposure to these compounds through drinking water, air, and soil. Recently, several studies were directed to assess the impact of these compounds on gut microbiota and disease occurrence in various experimental modelsCitation76,Citation77.

3.3.3.1. Glyphosate

Glyphosate-based compounds are extensively used as an herbicide in agriculture for controlling weedsCitation78,Citation79. Recent studies have illustrated that the effect of glyphosate on the gut environment was limited and is directly proportional to sufficient amounts of aromatic aminoCitation80,Citation81. However, the detrimental effects of glyphosate were observed on the gut of individuals with malnutrition or consuming low protein diets. A recent study revealed a significant reduction of bacterial density upon the exposure of Hawaiian green turtles to glyphosate ≥ 2.2 × 10-4gL-1. Moreover, the abundance of Pantoea, Proteus, Shigella, and Staphylococcus has significantly reduced in a dose-dependent manner with glyphosate, indicating that glyphosate may have an adverse effect on gut microbiota and eventually on overall healthCitation82. Exposure of rats to glyphosate resulted in an alteration in the villi’s morphology in the duodenum and jejunum of rats.

Furthermore, glyphosate exposure suppressed the antioxidant mechanism by inducing inflammatory responses via increasing mRNA expression of IL-1β, IL-6, TNF-α, caspase-3, Mapk3, and NF-κB. Exposure of rats to glyphosate significantly decreased the abundance of Firmicutes compared to other phylaCitation82. In contrast, another study demonstrated the deleterious effects of higher concentrations of glyphosate on immature bees through alteration in the richness of gut microbiota by shifting species diversityCitation83. Most importantly, environmental exposure of human beings with glyphosate significantly affected the gut-brain barrier in humans by elevating the abundance of clostridium bacteriaCitation84.

3.3.3.2. Pentachlorophenol (PCP)

Pentachlorophenol (PCP) is extensively used in paddy fields resulting in significant PCP contamination of the aquatic environmentCitation85. Interestingly, a study in goldfish demonstrated the change in the gut microbial community due to the toxic effects of PCP. In addition, PCP exposure to fish for 28 days revealed that the predisposition of PCP in the liver and intestine leads to growth inhibition, increased oxidative stress, and histopathological damage. Further, it also resulted in an altered ratio of Bacteroidetes/Firmicutes in the fish gut, causing the microbial community shiftCitation77. These findings increased interest in PCP-induced toxicity on gut microbiota dysbiosis and host health. Further, antibiotics significantly elevated the triazine-induced risks via gut microbiota dysbiosisCitation86, supporting possible interaction between antibiotics, herbicide, and triazine. However, additional studies will be needed to establish the association between gut microbiota and human disease risk due to herbicide toxicity. Based on the available literature, the apparent effect of different pesticides on gut microbiota is collectively represented in .

Table 1. Impact of pesticides with the subsequent outcome on gut microbiota.

3.3.4. Fungicides

This class of compounds is used to target and kill fungi and their spores. These biocidal compounds are generally fortified in post-harvested crops like fruits/vegetables and grains, causing significant contamination of agricultural products; consumption of this contaminated agricultural produce significantly resulted in health risks in humans and other animals, as discussed below.Citation87

3.3.4.1. Carbendazim (CBZ)

CBZ (methyl 2-benzimidazole carbamate) is a fungicide that inhibits fungal infections during agriculture storage and industrial processingCitation88. However, CBZ potentiates disease pathogenesis by restricting liver oxidative stress, causing reproductive toxicity and endocrine disruptionCitation89,Citation90. Oral high-dose administration of CBZ in mice for 28 days resulted in hepatic lipid metabolic disorder due to its accumulation in liver and fat tissue, eventually leading to inflammationCitation91. Further, CBZ accumulates in the GI tract, where it interacts with gut microbiota leading to dysbiosis due to decreased population of Bacteroides and an increased abundance of Firmicutes, Proteobacteria, and ActinobacteriaCitation9. Further, chronic exposure of mice with CBZ for 14 weeks resulted in dysbiosis of gut microbiota leading to dyslipidemia and facilitated the intestinal absorption of triglycerides leading to increased inflammatory responseCitation92. Therefore, CBZ exposure potentially alters the gut microbiota and affects overall health by disrupting host hepatic lipid metabolism and inflammatory response in the host.

3.3.4.2. Imazalil (IMZ)

Like other fungicides, IMZ is a well-known compound used to protect post-harvested crop produce and is often detected in soil, water, fruits, and vegetablesCitation93–95. Exposure of zebrafish with IMZ at the early development stage potentially induced developmental toxicity and locomotor behavior abnormalitiesCitation96. Further, exposure of zebrafish with IMZ for 21 days resulted in dysbiosis and altered hepatic metabolismCitation8. Additionally, exposure of mice with IMZ for 28 days (100 mg/kg body wt/day) potentially initiated mouse colonic inflammation and gut microbiota dysbiosis via decreasing abundance of Firmicutes and increasing Proteobacteria, Actinobacteria, and Bacteroidetes. Simultaneously, IMZ exposure significantly reduced the Lactobacillus population and Bifidobacterium, whereas elevated Deltaproteobacteria and DesulfovibrioCitation92. Chronic exposure of mice with IMZ at a low dose altered the intestinal barrier function in mice. These results also revealed that IMZ exposure is associated with reduced mucosal release and downregulates the expression of cystic fibrosis transmembrane conductance regulator in the ileum and colon of the miceCitation97.

3.3.4.3. Propamocarb (PM)

PM in the environment is another health threat to the human population. It is known as carbamate fungicide, which controls diseases associated with the OomycetesCitation98. The residue of PM was detected in humans and shown to have detrimental effects on healthCitation99. Hepatic lipid metabolism was perturbed in zebrafish upon high-dose exposure to PM for a short durationCitation11. Previous studies have shown that PM exposure resulted in perturbation of the microbial metabolite due to gut microbiota alterations. Exposure to PM at a dose of 300 mg/L for four weeks led to dysbiosis, which resulted in altered fecal metabolites, TMA, SCFAs, and succinates, leading to detrimental effects on the host’s healthCitation100. TMA is the key molecule involved in the development of atherosclerosis, which is significantly elevated in the feces upon exposure to PM.

Further, the PM also alters the cardiac NO/NOS pathway and upregulates NF-κB at the transcriptional level, ultimately increasing the risk of cardiovascular disease through induction of the enterohepatic metabolismCitation99,Citation100. Moreover, the composition of gut microbiota and fecal metabolites involved in energy metabolism in mice was significantly altered due to long-term exposure to a low dose of PM. Nevertheless, gut microbiota is reported to metabolize dietary choline to TMA, which is further oxidized to TMAO in the liver by flavin monooxygenase-3 and the farnesoid receptor. The increased level of TMAO is a potent biomarker for the risk assessment for the development of cardiac diseasesCitation101–103.

3.3.4.4. Epoxiconazole

This category of fungicides belongs to the azole group and use to intercept the diseases like leaf blotch and rust in wheatCitation104,Citation105. Therefore, consuming contaminated wheat products was a potential source of exposure to the human population. Furthermore, the previous study indicates that exposure of female rats to different doses of epoxiconazole (4 mg/Kg/day and 100 mg/Kg/day) for 90 days potentially modified the composition of the gut microbiota in rats by decreasing the level of Firmicutes and increasing the abundance of Bacteroidetes and Proteobacteria. This alteration in the abundance of Bacteroidaceae (increase/decrease) is associated with pouchitis and Crohn’s disease. Similarly, the Lactobacillaceae known to protect against liver damage is found to be influenced due to the exposure of epoxiconazoleCitation106–108. All these studies have shown the potential effect of epoxiconazole in altering gut microbiota and inducing hepatic toxicity, suggesting that early monitoring could be a better preventive measure to avoid potential health risks to the hostCitation109. To summarize, fungicides are potent compounds affecting human health by modulating gut microbiota. However, further mechanistic-based studies would be needed to fulfill the significant gap and the scope of research to explore fungicides direct impact on gut microbiota.

3.3.5. Insecticides

Insecticides are potentially used to increase crop production; however, their chronic application on crops leads to their residue/remnants on agricultural commodities resulting in harmful effects on the health of humans and other animalsCitation110,Citation111. As evident from the available literature, these compounds, including imidacloprid and thiamethoxam, are toxic to the environment and accumulate in the food chain, which is one of the significant concerns to human health. Their levels are continuously detected in the human body fluids vary significantly, indicating that these compounds enter the human body leading to a predisposition to different diseasesCitation112–114. The gut is one of the significant non-target organs majorly affected due to the exposure of insecticides directly or indirectly through contaminated food and water, which gains the researcher’s attention to identify the toxic effect of insecticides on gut microbiota and their consequences on human health.

4. Role of pesticide exposure in modulating host health via gut microbiota

The harmful effects of pesticide contamination on non-target species have increasingly gained more attention in the last few years. Surprisingly, few researchers have identified the function of gut microbiota in the detoxifying pesticide effects to maintain the host metabolic homeostasisCitation1,Citation83,Citation115. Dysregulation of gut microbiota composition is found to be associated with metabolic disorders, which can further lead to diseases like diabetes, colon cancer, etc.Citation116,Citation117. Various studies have unraveled the mechanism of pesticide toxicity on non-target species. The usage of various organochlorine pesticides, including β-HCH and p’p’ DDE were banned three decades back. However, their residual levels are still detected in the environment at a significant levelCitation53. Alteration in metabolites, diversity changes, and energy interference are the three indicators of microbiota disturbance and may impact host health via increasing disease risk. Therefore, the toxicity assessment of gut microbiota can potentially bridge environmental exposure and human health/diseases. Thus, this section discusses the connection between gut microbiota toxicity and human health. Pesticides and other xenobiotics are known to influence the physiologic function of the gut microbiota and are responsible for the pathogenesis of various diseases (). Consistent modulation in the gut microbiota due to exposure to a particular chemical could be a potential biomarker to assess its toxicity. SCFAs, one of the metabolites of gut microbiota, acts as a signaling molecule that binds to cellular receptors, including G-protein coupled receptor (GPCRs) and transmembrane G protein-coupled receptor 5 (TGR5). Farnesoid X receptor (FXR)Citation118 activates the signaling cascade, contributing to regulating metabolic activities through the gut-brain barrier. SCFA and bile acids affect insulin secretion and glucose homeostasis, possibly leading to the risk of developing diabetesCitation119,Citation120. The tryptophan metabolites can contribute to the modulation of intestinal immune cells and barrier functions via activating aryl hydrocarbon receptor (AHR), which is involved in the inflammatory bowel disease (IBD) pathogenesis Citation121–123. Gut bacteria convert choline and L-carnitine into trimethylamine, which is metabolized in the liver in triethylamine N-oxide (TMAO), which is significantly associated with cardiovascular disease riskCitation124,Citation125. Fermented products of proteins like polyamines and N-nitroso compounds derived by gut bacteria deploy carcinogenic effects and promote colorectal malignancyCitation125,Citation126. Environmental chemicals like diazinon and nicotine perturb bacterial metabolites that are neurotransmitters (serotonin, gamma-aminobutyric acid), resulting in neurotoxic effectsCitation127. Exposure of mice to various pesticides, especially organochlorine compounds β-HCH and DDE, could hinder the constitution of gut microbiota leading to further disturbance in the metabolism of bile acids and simultaneously decreasing the β–Muricholic acid (β-MCA), which is an important component that regulates the metabolism of bile, lipid, and glucoseCitation57. There is an alleviation in high-fat diet-induced obesity in mice upon exposure to endosulfan sulfate, resulting in alteration in gut microbiota, lipid metabolism, and glucose homeostasisCitation128. Apart from organochlorine pesticides, chlorpyrifos, an organophosphate pesticide, has been shown to possess significant toxic effects on the gut microbiota. Chronic exposure to chlorpyrifos may lead to inflammation in the intestine via influencing gut microbiota, disturbing the urine and liver metabolism, and increasing the mucous volume in the intestine, which leads to the predisposition of oxidative stressCitation52,Citation66. Other pesticides like nitenpyram and imazalil are known to impact gut microbiota negatively. Prenatal exposure of mice with nitenpyram resulted in dysregulation of gut microbiota and fecal metabolites like purines, BCAAs, and TCA cycle components leading to elevated consumption of the host energyCitation128. To assess the toxic effect of imazalil, the zebrafish model was used, and it was found to alter the gut microbiota and influence liver metabolismCitation8. Various pathways, including the TCA cycle, amino acid/lipid/nucleotide, and glucose metabolism, were also altered after exposure to imazalilCitation8. Similarly, short-term exposure to pro-paraben and propamocarb resulted in adverse effects on the gut microbiota and liver metabolic profileCitation129. It was observed that there are approximately 20 fecal metabolites were affected due to the exposure to the propamocarbCitation99. Previous studies also revealed the impact of exposure to penconazole on gut microbiota and serum metabolic profile in miceCitation130. Exposure to pesticides can significantly impact the gut microbiota, leading to an imbalance of microbiota composition, which may further cause metabolic diseases in the host. However, in the future, more studies will be required for a further and in-depth understanding of mechanisms and their effect on the host.

Table 2. Gut microbiota toxicity associated with specific pesticide and heavy metal exposure.

5. Other potential environmental contaminants

Heavy metals are one of the extensively studied environmental contaminants, but their role in gut toxicity is unidentified. The gut bacterial flora is vital in the biotransformation of heavy metals via promoting or attenuating their toxic effects. For example, arsenic exposure is one of the major concerns to human health through the ingestion of contaminated water. A study has shown that human gut microbiota can convert inorganic arsenic into an organic form that is less toxic to human healthCitation131. Exposure of mice to arsenic, cadmium, cobalt, chromium, and nickel in drinking water resulted in alterations in the gut metabolic profileCitation132,Citation133. In agreement with this finding, another study also showed the alteration in gut microbiota after exposure to a 100 ppb dose of arsenic in miceCitation134, which provides a new horizon to evaluate the pathways behind arsenic-induced toxic effects in humans. Further, the arsenic exposure in the Bangladesh population significantly modulated the gut microbiota by promoting an increased abundance of Citrobacter, which is responsible for various health concerns in humans, including urinary tract infections, respiratory diseases, inflamed gastrointestinal tract, cardiovascular disease, specifically atherosclerosis through TMAO, etc.Citation135–137. Previous studies have shown that exposure to heavy metals like arsenic, cadmium, lead, and manganese may contribute toward the inflammatory events in the gut via a decrease in indole lactic acid, daidzein, and glycocholic acid, vitamin E leading to an increase in LPSCitation138–140 level in the lumen of the gut. Gut microbiota also performs the demethylation of mercury and its conversion into methylmercury, a more toxic form of mercuryCitation141. Mercury is another heavy metal extensively studied for its effect on the microbiota in several experimental models, including mice, rats, chickens, fish, and humans. Exposure to methylmercury antedate damages the GI tract and alters the gut microbiotaCitation142,Citation143. However, there are studies conducted to examine the effects of heavy metals on gut microbiota composition (). Still, there is a paucity of literature that explores the mechanism behind the heavy metal-induced toxicity of gut microbiota. In the future, there is an urgent need to conduct more mechanistic studies to explore heavy metals’ toxic effects on gut microbiota and metabolic alteration.

6. Future perspectives and conclusion

In recent years, due to the extensive usage of pesticides globally, their harmful effect on human health has gained significant attention. To avoid the detrimental impact of pesticides on human health, biopesticides could be a promising replacement. This could include microorganisms that target pathogens or RNA-based biopesticides. The use of the synthetic biology approach may offer a better future for agriculture with minimal harm to human health. All the available studies have correlated to pesticide exposure leading to alterations in host gut microbiota composition and metabolic profiles. However, gut microbiota toxicity has yet to be explored so far due to a lack of literature on mechanistic-based studies.

Further, the available studies have yet to provide a concrete relationship between a particular pesticide and microbiota or metabolite to assess its toxicity. Therefore, there is vast space to explore the mechanism behind the toxicity induced by pesticide exposure to gut microbiota and associated metabolites. In the future, deeper sequencing techniques should be adopted, like shotgun sequencing, as well as 16s RNA will be required to identify the strain and species, which are altered explicitly due to pesticide exposure in the gut.

To conclude, pesticide exposure may induce toxicity to gut microbiota by modulating gut microbiota composition and their functional changes (summarized in ). Due to these changes, gut microbiota can be a potential biomarker to assess pesticide toxicity. Furthermore, pesticide exposure is linked to various human diseases due to diversity loss, alteration in metabolic profiles, and energy metabolism. Although there is multiple evidence for the effect of environmental exposure on the gut microbiota, it is propitious to evaluate the mechanism of such disturbance through the lens of toxicology. This review emphasized pesticide toxicity on gut microbiota and xenobiotic-induced damage to human health and disease. Further development of biomarkers development by assessment and modification in gut microbiota composition. Thus, an overall in-depth evaluation of xenobiotic-induced toxicity on the gut microbiota will provide insight into environmental exposure and human gut health, which may facilitate the development of biomarkers for therapeutic interventions.

Figure 2. Effect of the predisposition of different environmental contaminants (pesticides and heavy metals) on the functional changes of gut microbiota of the host. The functional modification in gut microbiota leads to diversity loss and alteration in functional metabolites. This subsequently results in the pathogenesis of various diseases, including colorectal cancer, via invasion of IBD pathogens and accumulation of non-digestible carbohydrates.

Figure 2. Effect of the predisposition of different environmental contaminants (pesticides and heavy metals) on the functional changes of gut microbiota of the host. The functional modification in gut microbiota leads to diversity loss and alteration in functional metabolites. This subsequently results in the pathogenesis of various diseases, including colorectal cancer, via invasion of IBD pathogens and accumulation of non-digestible carbohydrates.

Abbreviations

PCP=

Pentachlorophenol

CPF=

chlorpyrifos

HCH=

b-hexachlorohexane

DDT=

p’p’ dichlorodiphenyltrichloroethane

p’p’DDE=

p’p’ dechlorodiphenyldichloro-ethylene

HCH=

hexachlorocyclohexane

OCPs=

Organochlorines pesticides

Tlr4=

Toll-like receptor 4

PAH=

polycyclic aromatic hydrocarbon

PCB=

polychlorinated biphenyl

POPs=

persistent organic pollutants

TMAO=

trimethylamine oxide

Fmo3=

flavin-containing monooxygenase 3

LPS=

lipopolysaccharides

TMA=

trimethylamine

SCFAs=

Short-chain fatty acids

BA=

bile acids

PCP=

Pentachlorophenol

CBZ=

methyl 2-benzimidazolecarbamate

IMZ=

Imazalil

CFTR=

cystic fibrosis transmembrane conductance regulator

PM=

Propamocarb

FXR=

farnesoid X receptor

GPCRs=

G-protein coupled receptors

AHR=

aryl hydrocarbon receptor

IBD=

inflammatory bowel disease

β-MCA=

β – Muricholic acid

Credit authorship contribution statement

T.S. Writing – original draft preparation, review, and editing, N.S.N. Writing, review, editing, and visualization. RP. Review and editing, S.K.B and S.R. Conceptualization review and editing, all authors have read and agreed to the published version of the manuscript.

Acknowledgments

Figures are created with BioRender.com.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

The authors obtained funding support for this manuscript, in part, from the National Institutes of Health [R01 CA247763].

References

  • Meng Z, Liu L, Yan S, Sun W, Jia M, Tian S, Huang S, Zhou Z, Zhu W. Gut microbiota: a key factor in the host health effects induced by pesticide exposure? J Agric Food Chem. 2020;68(39):10517–19. doi:10.1021/acs.jafc.0c04678.
  • Lasram MM, Annabi AB, El Elj N, Selmi S, Kamoun A, El-Fazaa S, Gharbi N. Metabolic disorders of acute exposure to malathion in adult Wistar rats. J Hazard Mater. 2009;163(2–3):1052–1055. doi:10.1016/j.jhazmat.2008.07.059.
  • Lasram MM, Bouzid K, Douib IB, Annabi A, El Elj N, El Fazaa S, Abdelmoula J, Gharbi N. Lipid metabolism disturbances contribute to insulin resistance and decrease insulin sensitivity by malathion exposure in Wistar rat. Drug Chem Toxicol. 2015;38(2):227–234. doi:10.3109/01480545.2014.933348.
  • Ogutcu A, Suludere Z, Kalender Y. Dichlorvos-induced hepatotoxicity in rats and the protective effects of vitamins C and E. Environ Toxicol Pharmacol. 2008;26(3):355–361. doi:10.1016/j.etap.2008.07.005.
  • Zhang W, Lu Y, Huang L, Cheng C, Di S, Chen L, Zhou Z, Diao J. 2018. Comparison of triadimefon and its metabolite on acute toxicity and chronic effects during the early development of Rana nigromaculata tadpoles. Ecotoxicol Environ Saf. 156:247–254. doi:10.1016/j.ecoenv.2018.03.009.
  • Baldi I, Gruber A, Rondeau V, Lebailly P, Brochard P, Fabrigoule C. Neurobehavioral effects of long-term exposure to pesticides: results from the 4-year follow-up of the PHYTONER study. Occup Environ Med. 2011;68(2):108–115. doi:10.1136/oem.2009.047811.
  • Sharon M, Bhawana M, Anita S, Gothecha VK. A short review on how pesticides affect human health. Int J Ayurvedic Herb Med. 2012;2:935–946.
  • Jin C, Luo T, Zhu Z, Pan Z, Yang J, Wang W, Fu Z, Jin Y. 2017. Imazalil exposure induces gut microbiota dysbiosis and hepatic metabolism disorder in zebrafish. Comp Biochem Physiol C Toxicol Pharmacol. 202:85–93. doi:10.1016/j.cbpc.2017.08.007.
  • Jin Y, Zeng Z, Wu Y, Zhang S, Fu Z. Oral exposure of mice to carbendazim induces hepatic lipid metabolism disorder and gut microbiota dysbiosis. Toxicol Sci. 2015;147(1):116–126. doi:10.1093/toxsci/kfv115.
  • Liang Y, Liu D, Zhan J, Luo M, Han J, Wang P, Zhou Z. 2020. New insight into the mechanism of POP-induced obesity: evidence from DDE-altered microbiota. Chemosphere. 244:125123. doi:10.1016/j.chemosphere.2019.125123.
  • Zhang R, Pan Z, Wang X, Shen M, Zhou J, Fu Z, Jin Y. Short-term propamocarb exposure induces hepatic metabolism disorder associated with gut microbiota dysbiosis in adult male zebrafish. Acta Biochim Biophys Sin (Shanghai). 2019;51(1):88–96. doi:10.1093/abbs/gmy153.
  • Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444(7122):1022–1023. doi:10.1038/4441022a.
  • Flint HJ, Scott KP, Louis P, Duncan SH. The role of the gut microbiota in nutrition and health. Nat Rev Gastroenterol Hepatol. 2012;9(10):577–589. doi:10.1038/nrgastro.2012.156.
  • Jin C, Xia J, Wu S, Tu W, Pan Z, Fu Z, Wang Y, Jin Y. Insights into a possible influence on gut microbiota and intestinal barrier function during chronic exposure of mice to imazalil. Toxicol Sci. 2018;162(1):113–123. doi:10.1093/toxsci/kfx227.
  • Liang Y, Zhan J, Liu D, Luo M, Han J, Liu X, Liu C, Cheng Z, Zhou Z, Wang P. Organophosphorus pesticide chlorpyrifos intake promotes obesity and insulin resistance through impacting gut and gut microbiota. Microbiome. 2019;7(1):19. doi:10.1186/s40168-019-0635-4.
  • Backhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host-bacterial mutualism in the human intestine. Science. 2005;307(5717):1915–1920. doi:10.1126/science.1104816.
  • Thursby E, Juge N. Introduction to the human gut microbiota. Biochem J. 2017;474(11):1823–1836. doi:10.1042/BCJ20160510.
  • Human Microbiome Project C. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486(7402):207–214. doi:10.1038/nature11234.
  • Clemente JC, Ursell LK, Parfrey LW, Knight R. The impact of the gut microbiota on human health: an integrative view. Cell. 2012;148(6):1258–1270. doi:10.1016/j.cell.2012.01.035.
  • Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59–65. doi:10.1038/nature08821.
  • Pothuraju R, Chaudhary S, Rachagani S, Kaur S, Roy HK, Bouvet M, Batra SK. Mucins, gut microbiota, and postbiotics role in colorectal cancer. Gut Microbes. 2021;13(1):1974795. doi:10.1080/19490976.2021.1974795.
  • Smith K, McCoy KD, Macpherson AJ. Use of axenic animals in studying the adaptation of mammals to their commensal intestinal microbiota. Semin Immunol. 2007;19(2):59–69. doi:10.1016/j.smim.2006.10.002.
  • Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, Pettersson S. Host-gut microbiota metabolic interactions. Science. 2012;336(6086):1262–1267. doi:10.1126/science.1223813.
  • Tang WH, Kitai T, Hazen SL. Gut microbiota in cardiovascular health and disease. Circ Res. 2017;120(7):1183–1196. doi:10.1161/CIRCRESAHA.117.309715.
  • Carpino G, Del Ben M, Pastori D, Carnevale R, Baratta F, Overi D, Francis H, Cardinale V, Onori P, Safarikia S, et al. Increased liver localization of lipopolysaccharides in human and experimental NAFLD. Hepatology. 2020;72(2):470–485. doi:10.1002/hep.31056.
  • Roberts AB, Gu X, Buffa JA, Hurd AG, Wang Z, Zhu W, Gupta N, Skye SM, Cody DB, Levison BS, et al. Development of a gut microbe–targeted nonlethal therapeutic to inhibit thrombosis potential. Nat Med. 2018;24(9):1407–1417. doi:10.1038/s41591-018-0128-1.
  • Wahlstrom A, Sayin SI, Marschall HU, Backhed F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab. 2016;24(1):41–50. doi:10.1016/j.cmet.2016.05.005.
  • Lavelle A, Sokol H. Gut microbiota-derived metabolites as key actors in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol. 2020;17(4):223–237. doi:10.1038/s41575-019-0258-z.
  • Heyman M, Corthier G, Lucas F, Meslin JC, Desjeux JF. Evolution of the caecal epithelial barrier during Clostridium difficile infection in the mouse. Gut. 1989;30(8):1087–1093. doi:10.1136/gut.30.8.1087.
  • Sekirov I, Tam NM, Jogova M, Robertson ML, Li Y, Lupp C, Finlay BB. Antibiotic-induced perturbations of the intestinal microbiota alter host susceptibility to enteric infection. Infect Immun. 2008;76(10):4726–4736. doi:10.1128/IAI.00319-08.
  • Ferreira RB, Gill N, Willing BP, Antunes LC, Russell SL, Croxen MA, Finlay BB. The intestinal microbiota plays a role in Salmonella-induced colitis independent of pathogen colonization. PLoS One. 2011;6(5):e20338. doi:10.1371/journal.pone.0020338.
  • Shanahan F. Nutrient tasting and signaling mechanisms in the gut V. Mechanisms of immunologic sensation of intestinal contents. Am J Physiol Gastrointest Liver Physiol. 2000;278(2):G191–6. doi:10.1152/ajpgi.2000.278.2.G191.
  • Alam M, Midtvedt T, Uribe A. Differential cell kinetics in the ileum and colon of germfree rats. Scand J Gastroenterol. 1994;29(5):445–451. doi:10.3109/00365529409096836.
  • Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol. 2009;9(5):313–323. doi:10.1038/nri2515.
  • Podolsky DK. The current future understanding of inflammatory bowel disease. Best Pract Res Clin Gastroenterol. 2002;16(6):933–943. doi:10.1053/bega.2002.0354.
  • Backhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF, Gordon JI. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A. 2004;101(44):15718–15723. doi:10.1073/pnas.0407076101.
  • Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444(7122):1027–1031. doi:10.1038/nature05414.
  • Koh A, De Vadder F, Kovatcheva-Datchary P, Backhed F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell. 2016;165(6):1332–1345. doi:10.1016/j.cell.2016.05.041.
  • Wong JM, de Souza R, Kendall CW, Emam A, Jenkins DJ. Colonic health: fermentation and short chain fatty acids. J Clin Gastroenterol. 2006;40(3):235–243. doi:10.1097/00004836-200603000-00015.
  • Chiu K, Warner G, Nowak RA, Flaws JA, Mei W. The impact of environmental chemicals on the gut microbiome. Toxicol Sci. 2020;176(2):253–284. doi:10.1093/toxsci/kfaa065.
  • Li QQ, Loganath A, Chong YS, Tan J, Obbard JP. Persistent organic pollutants and adverse health effects in humans. J Toxicol Environ Health A. 2006;69(21):1987–2005. doi:10.1080/15287390600751447.
  • Wang S, Huang J, Yang Y, Hui Y, Ge Y, Larssen T, Yu G, Deng S, Wang B, Harman C. First report of a Chinese PFOS alternative overlooked for 30 years: its toxicity, persistence, and presence in the environment. Environ Sci Technol. 2013;47(18):10163–10170. doi:10.1021/es401525n.
  • Kandel Gambarte PC, Wolansky MJ. 2022. The gut microbiota as a biomarker for realistic exposures to pesticides: a critical consideration. Neurotoxicol Teratol. 91:107074. doi:10.1016/j.ntt.2022.107074.
  • Paik D, Yao L, Zhang Y, Bae S, D’agostino GD, Zhang M, Kim E, Franzosa EA, Avila-Pacheco J, Bisanz JE, et al. Human gut bacteria produce ΤΗ17-modulating bile acid metabolites. Nature. 2022;603(7903):907–912. doi:10.1038/s41586-022-04480-z.
  • Rude KM, Pusceddu MM, Keogh CE, Sladek JA, Rabasa G, Miller EN, Sethi S, Keil KP, Pessah IN, Lein PJ, et al. Developmental exposure to polychlorinated biphenyls (PCBs) in the maternal diet causes host-microbe defects in weanling offspring mice. Environ Pollut. 2019;253:708–721. doi:10.1016/j.envpol.2019.07.066.
  • Ley RE, Backhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI. Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A. 2005;102(31):11070–11075. doi:10.1073/pnas.0504978102.
  • Petriello MC, Hoffman JB, Vsevolozhskaya O, Morris AJ, Hennig B. 2018. Dioxin-like PCB 126 increases intestinal inflammation and disrupts gut microbiota and metabolic homeostasis. Environ Pollut. 242:1022–1032. doi:10.1016/j.envpol.2018.07.039.
  • Choi JJ, Eum SY, Rampersaud E, Daunert S, Abreu MT, Toborek M. Exercise attenuates PCB-induced changes in the mouse gut microbiome. Environ Health Perspect. 2013;121(6):725–730. doi:10.1289/ehp.1306534.
  • Van de Wiele T, Vanhaecke L, Boeckaert C, Peru K, Headley J, Verstraete W, Siciliano S. Human colon microbiota transform polycyclic aromatic hydrocarbons to estrogenic metabolites. Environ Health Perspect. 2005;113(1):6–10. doi:10.1289/ehp.7259.
  • Mnif W, Hassine AI, Bouaziz A, Bartegi A, Thomas O, Roig B. Effect of endocrine disruptor pesticides: a review. Int J Environ Res Public Health. 2011;8(6):2265–2303. doi:10.3390/ijerph8062265.
  • Liu Y, Yao Y, Li H, Qiao F, Wu J, Du ZY, Zhang M. Influence of endogenous and exogenous estrogenic endocrine on intestinal microbiota in zebrafish. PLoS One. 2016;11(10):e0163895. doi:10.1371/journal.pone.0163895.
  • Zhao Y, Zhang Y, Wang G, Han R, Xie X. 2016. Effects of chlorpyrifos on the gut microbiome and urine metabolome in mouse (Mus musculus). Chemosphere. 153:287–293. doi:10.1016/j.chemosphere.2016.03.055.
  • Maisano M, Cappello T, Oliva S, Natalotto A, Giannetto A, Parrino V, Battaglia P, Romeo T, Salvo A, Spanò N, et al. PCB and OCP accumulation and evidence of hepatic alteration in the Atlantic bluefin tuna, T. thynnus, from the Mediterranean Sea. Mar Environ Res. 2016;121:40–48. doi:10.1016/j.marenvres.2016.03.003.
  • Muller MHB, Polder A, Brynildsrud OB, Karimi M, Lie E, Manyilizu WB, Mdegela RH, Mokiti F, Murtadha M, Nonga HE, et al. Organochlorine pesticides (OCPs) and polychlorinated biphenyls (PCBs) in human breast milk and associated health risks to nursing infants in Northern Tanzania. Environ Res. 2017;154:425–434. doi:10.1016/j.envres.2017.01.031.
  • Lee HS, Lee JC, Lee IK, Moon HB, Chang YS, Jacobs DR Jr., Lee D-H. Associations among organochlorine pesticides, Methanobacteriales, and obesity in Korean women. PLoS One. 2011;6(11):e27773. doi:10.1371/journal.pone.0027773.
  • Salihovic S, Ganna A, Fall T, Broeckling CD, Prenni JE, van Bavel B, Lind PM, Ingelsson E, Lind L. 2016. The metabolic fingerprint of p,p′-DDE and HCB exposure in humans. Environ Int. 88:60–66. doi:10.1016/j.envint.2015.12.015.
  • Liu Q, Shao W, Zhang C, Xu C, Wang Q, Liu H, Sun H, Jiang Z, Gu A. 2017. Organochloride pesticides modulated gut microbiota and influenced bile acid metabolism in mice. Environ Pollut. 226:268–276. doi:10.1016/j.envpol.2017.03.068.
  • Velmurugan G, Ramprasath T, Swaminathan K, Mithieux G, Rajendhran J, Dhivakar M, Parthasarathy A, Babu DDV, Thumburaj LJ, Freddy AJ, et al. Gut microbial degradation of organophosphate insecticides-induces glucose intolerance via gluconeogenesis. Genome Biol. 2017;18(1):8. doi:10.1186/s13059-016-1134-6.
  • Giesy JP, Solomon KR, Cutler GC, Giddings JM, Mackay D, Moore DR, Purdy J, Williams WM. Ecological risk assessment of the uses of the organophosphorus insecticide chlorpyrifos, in the United States. Rev Environ Contam Toxicol. 2014;231:1–11 doi:10.1007/978-3-319-03865-0_1.
  • Xia X, Zheng D, Zhong H, Qin B, Gurr GM, Vasseur L, Lin H, Bai J, He W, You M, et al. DNA sequencing reveals the midgut microbiota of diamondback moth, Plutella xylostella (L.) and a possible relationship with insecticide resistance. PLoS One. 2013;8(7):e68852. doi:10.1371/journal.pone.0068852.
  • Xia X, Sun B, Gurr GM, Vasseur L, Xue M, You M. 2018. Gut microbiota mediate insecticide resistance in the diamondback moth, Plutella xylostella (L.). Front Microbiol. 9:25. doi:10.3389/fmicb.2018.00025.
  • Reygner J, Joly Condette C, Bruneau A, Delanaud S, Rhazi L, Depeint F, Abdennebi-Najar L, Bach V, Mayeur C, Khorsi-Cauet H. Changes in composition and function of human intestinal microbiota exposed to chlorpyrifos in oil as assessed by the SHIME® model. Int J Environ Res Public Health. 2016;13(11):1088. doi:10.3390/ijerph13111088.
  • Joly C, Gay-Queheillard J, Leke A, Chardon K, Delanaud S, Bach V, Khorsi-Cauet H. Impact of chronic exposure to low doses of chlorpyrifos on the intestinal microbiota in the Simulator of the Human Intestinal Microbial Ecosystem (SHIME®) and in the rat. Environ Sci Pollut Res Int. 2013;20(5):2726–2734. doi:10.1007/s11356-012-1283-4.
  • Requile M, Gonzalez Alvarez DO, Delanaud S, Rhazi L, Bach V, Depeint F, Khorsi-Cauet H. Use of a combination of in vitro models to investigate the impact of chlorpyrifos and inulin on the intestinal microbiota and the permeability of the intestinal mucosa. Environ Sci Pollut Res Int. 2018;25(23):22529–22540. doi:10.1007/s11356-018-2332-4.
  • Reygner J, Lichtenberger L, Elmhiri G, Dou S, Bahi-Jaber N, Rhazi L, Depeint F, Bach V, Khorsi-Cauet H, Abdennebi-Najar L, et al. Inulin supplementation lowered the metabolic defects of prolonged exposure to chlorpyrifos from gestation to young adult stage in offspring rats. PLoS One. 2016;11(10):e0164614. doi:10.1371/journal.pone.0164614.
  • Wang X, Shen M, Zhou J, Jin Y. 2019. Chlorpyrifos disturbs hepatic metabolism associated with oxidative stress and gut microbiota dysbiosis in adult zebrafish. Comp Biochem Physiol C Toxicol Pharmacol. 216:19–28. doi:10.1016/j.cbpc.2018.11.010.
  • Fang B, Li JW, Zhang M, Ren FZ, Pang GF. 2018. Chronic chlorpyrifos exposure elicits diet-specific effects on metabolism and the gut microbiome in rats. Food Chem Toxicol. 111:144–152. doi:10.1016/j.fct.2017.11.001.
  • Joly Condette C, Bach V, Mayeur C, Gay-Queheillard J, Khorsi-Cauet H. Chlorpyrifos exposure during perinatal period affects intestinal microbiota associated with delay of maturation of digestive tract in rats. J Pediatr Gastroenterol Nutr. 2015;61(1):30–40. doi:10.1097/MPG.0000000000000734.
  • Manthripragada AD, Costello S, Cockburn MG, Bronstein JM, Ritz B. Paraoxonase 1, agricultural organophosphate exposure, and Parkinson disease. Epidemiology. 2010;21(1):87–94. doi:10.1097/EDE.0b013e3181c15ec6.
  • Shrestha S, Parks CG, Goldner WS, Kamel F, Umbach DM, Ward MH, Lerro CC, Koutros S, Hofmann JN, Beane Freeman LE, et al. Incident thyroid disease in female spouses of private pesticide applicators. Environ Int. 2018;118:282–292. doi:10.1016/j.envint.2018.05.041.
  • Aggarwal V, Deng X, Tuli A, Goh KS. Diazinon-chemistry and environmental fate: a California perspective. Rev Environ Contam Toxicol. 2013;223:107–140.
  • Gao B, Bian X, Chi L, Tu P, Ru H, Lu K. Editor’s highlight: organophosphate diazinon altered quorum sensing, cell motility, stress response, and carbohydrate metabolism of gut microbiome. Toxicol Sci. 2017;157(2):354–364. doi:10.1093/toxsci/kfx053.
  • Gao B, Bian X, Mahbub R, Lu K. Sex-specific effects of organophosphate diazinon on the gut microbiome and its metabolic functions. Environ Health Perspect. 2017;125(2):198–206. doi:10.1289/EHP202.
  • Tang J, Wang W, Jiang Y, Chu W. 2021. Diazinon exposure produces histological damage, oxidative stress, immune disorders and gut microbiota dysbiosis in crucian carp (Carassius auratus gibelio). Environ Pollut. 269:116129. doi:10.1016/j.envpol.2020.116129.
  • Gao B, Chi L, Tu P, Bian X, Thomas J, Ru H, Lu K. 2018. The organophosphate malathion disturbs gut microbiome development and the quorum-sensing system. Toxicol Lett. 283:52–57. doi:10.1016/j.toxlet.2017.10.023.
  • Aitbali Y, Ba-M’hamed S, Elhidar N, Nafis A, Soraa N, Bennis M. 2018. Glyphosate based- herbicide exposure affects gut microbiota, anxiety and depression-like behaviors in mice. Neurotoxicol Teratol. 67:44–49. doi:10.1016/j.ntt.2018.04.002.
  • Kan H, Zhao F, Zhang XX, Ren H, Gao S. Correlations of gut microbial community shift with hepatic damage and growth inhibition of Carassius auratus induced by pentachlorophenol exposure. Environ Sci Technol. 2015;49(19):11894–11902. doi:10.1021/acs.est.5b02990.
  • Shehata AA, Schrodl W, Aldin AA, Hafez HM, Kruger M. The effect of glyphosate on potential pathogens and beneficial members of poultry microbiota in vitro. Curr Microbiol. 2013;66(4):350–358. doi:10.1007/s00284-012-0277-2.
  • Bohn T, Cuhra M, Traavik T, Sanden M, Fagan J, Primicerio R. 2014. Compositional differences in soybeans on the market: glyphosate accumulates in Roundup Ready GM soybeans. Food Chem. 153:207–215. doi:10.1016/j.foodchem.2013.12.054.
  • Lozano VL, Defarge N, Rocque LM, Mesnage R, Hennequin D, Cassier R, de Vendômois JS, Panoff J-M, Séralini G-E, Amiel C. 2018. Sex-dependent impact of Roundup on the rat gut microbiome. Toxicol Rep. 5:96–107. doi:10.1016/j.toxrep.2017.12.005.
  • Nielsen LN, Roager HM, Casas ME, Frandsen HL, Gosewinkel U, Bester K, Licht TR, Hendriksen NB, Bahl MI. 2018. Glyphosate has limited short-term effects on commensal bacterial community composition in the gut environment due to sufficient aromatic amino acid levels. Environ Pollut. 233:364–376. doi:10.1016/j.envpol.2017.10.016.
  • Kittle RP, McDermid KJ, Muehlstein L, Balazs GH. 2018. Effects of glyphosate herbicide on the gastrointestinal microflora of Hawaiian green turtles (Chelonia mydas) Linnaeus. Mar Pollut Bull. 127:170–174. doi:10.1016/j.marpolbul.2017.11.030.
  • Dai ZL, Wu G, Zhu WY. Amino acid metabolism in intestinal bacteria: links between gut ecology and host health. Front Biosci (Landmark Ed). 2011;16(1):1768–1786. doi:10.2741/3820.
  • Argou-Cardozo I, Zeidan-Chulia F. Clostridium bacteria and autism spectrum conditions: a systematic review and hypothetical contribution of environmental glyphosate levels. Med Sci (Basel). 2018;6(2):6. doi:10.3390/medsci6020029.
  • Zhang M, Yin D, Kong F. The changes of serum testosterone level and hepatic microsome enzyme activity of crucian carp (Carassius carassius) exposed to a sublethal dosage of pentachlorophenol. Ecotoxicol Environ Saf. 2008;71(2):384–389. doi:10.1016/j.ecoenv.2007.10.014.
  • Zhan J, Liang Y, Liu D, Ma X, Li P, Liu C, Liu X, Wang P, Zhou Z. Antibiotics may increase triazine herbicide exposure risk via disturbing gut microbiota. Microbiome. 2018;6(1):224. doi:10.1186/s40168-018-0602-5.
  • Pico Y, El-Sheikh MA, Alfarhan AH, Barcelo D. 2018. Target vs non-target analysis to determine pesticide residues in fruits from Saudi Arabia and influence in potential risk associated with exposure. Food Chem Toxicol. 111:53–63. doi:10.1016/j.fct.2017.10.060.
  • Veerappan M, Hwang I, Pandurangan M. Effect of cypermethrin, carbendazim and their combination on male albino rat serum. Int J Exp Pathol. 2012;93(5):361–369. doi:10.1111/j.1365-2613.2012.00828.x.
  • Yu G, Liu Y, Xie L, Wang X. Involvement of Sertoli cells in spermatogenic failure induced by carbendazim. Environ Toxicol Pharmacol. 2009;27(2):287–292. doi:10.1016/j.etap.2008.11.006.
  • Jiang J, Wu S, Wu C, An X, Cai L, Zhao X. Embryonic exposure to carbendazim induces the transcription of genes related to apoptosis, immunotoxicity and endocrine disruption in zebrafish (Danio rerio). Fish Shellfish Immunol. 2014;41(2):493–500. doi:10.1016/j.fsi.2014.09.037.
  • Gao B, Chi L, Tu P, Gao N, Lu K. The carbamate aldicarb altered the gut microbiome, metabolome, and lipidome of C57BL/6J mice. Chem Res Toxicol. 2019;32(1):67–79. doi:10.1021/acs.chemrestox.8b00179.
  • Jin C, Zeng Z, Wang C, Luo T, Wang S, Zhou J, Ni Y, Fu Z, Jin Y. Insights into a possible mechanism underlying the connection of carbendazim-induced lipid metabolism disorder and gut microbiota dysbiosis in mice. Toxicol Sci. 2018;166(2):382–393. doi:10.1093/toxsci/kfy205.
  • Zega G, De Bernardi F, Groppelli S, Pennati R. Effects of the azole fungicide Imazalil on the development of the ascidian Ciona intestinalis (Chordata, Tunicata): morphological and molecular characterization of the induced phenotype. Aquat Toxicol. 2009;91(3):255–261. doi:10.1016/j.aquatox.2008.11.015.
  • Faniband MH, Littorin M, Ekman E, Jonsson BA, Lindh CH. Lc–MS-MS analysis of urinary biomarkers of imazalil following experimental exposures. J Anal Toxicol. 2015;39(9):691–697. doi:10.1093/jat/bkv100.
  • Masia A, Vasquez K, Campo J, Pico Y. 2015. Assessment of two extraction methods to determine pesticides in soils, sediments and sludges. Application to the Turia river basin. J Chromatogr A. 1378:19–31. doi:10.1016/j.chroma.2014.11.079.
  • Jin C, Zeng Z, Fu Z, Jin Y. 2016. Oral imazalil exposure induces gut microbiota dysbiosis and colonic inflammation in mice. Chemosphere. 160:349–358. doi:10.1016/j.chemosphere.2016.06.105.
  • Jin Y, Zhu Z, Wang Y, Yang E, Feng X, Fu Z. 2016. The fungicide imazalil induces developmental abnormalities and alters locomotor activity during early developmental stages in zebrafish. Chemosphere. 153:455–461. doi:10.1016/j.chemosphere.2016.03.085.
  • Liu C, Qin Z, Zhou X, Xin M, Wang C, Liu D, Li S. Expression and functional analysis of the Propamocarb-related gene CsDIR16 in cucumbers. BMC Plant Biol. 2018;18(1):16. doi:10.1186/s12870-018-1236-2.
  • Wu S, Jin C, Wang Y, Fu Z, Jin Y. 2018. Exposure to the fungicide propamocarb causes gut microbiota dysbiosis and metabolic disorder in mice. Environ Pollut. 237:775–783. doi:10.1016/j.envpol.2017.10.129.
  • Wu S, Luo T, Wang S, Zhou J, Ni Y, Fu Z, Jin Y. 2018. Chronic exposure to fungicide propamocarb induces bile acid metabolic disorder and increases trimethylamine in C57BL/6J mice. Sci Total Environ. 642:341–348. doi:10.1016/j.scitotenv.2018.06.084.
  • Bennett BJ, de Aguiar Vallim TQ, Wang Z, Shih DM, Meng Y, Gregory J, Allayee H, Lee R, Graham M, Crooke R, et al. Trimethylamine-N-oxide, a metabolite associated with atherosclerosis, exhibits complex genetic and dietary regulation. Cell Metab. 2013;17(1):49–60. doi:10.1016/j.cmet.2012.12.011.
  • Wang Z, Roberts AB, Buffa JA, Levison BS, Zhu W, Org E, Gu X, Huang Y, Zamanian-Daryoush M, Culley M, et al. Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell. 2015;163(7):1585–1595. doi:10.1016/j.cell.2015.11.055.
  • Zhu W, Gregory JC, Org E, Buffa JA, Gupta N, Wang Z, Li L, Fu X, Wu Y, Mehrabian M, et al. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell. 2016;165(1):111–124. doi:10.1016/j.cell.2016.02.011.
  • Paveley ND, Sylvester-Bradley R, Scott RK, Craigon J, Day W. Steps in predicting the relationship of yield on fungicide dose. Phytopathology®. 2001;91(7):708–716. doi:10.1094/PHYTO.2001.91.7.708.
  • Zarn JA, Bruschweiler BJ, Schlatter JR. Azole fungicides affect mammalian steroidogenesis by inhibiting sterol 14 alpha-demethylase and aromatase. Environ Health Perspect. 2003;111(3):255–261. doi:10.1289/ehp.5785.
  • Loguercio C, Federico A, Tuccillo C, Terracciano F, D’auria MV, De Simone C, Blanco CDV. Beneficial effects of a probiotic VSL#3 on parameters of liver dysfunction in chronic liver diseases. J Clin Gastroenterol. 2005;39(6):540–543. doi:10.1097/01.mcg.0000165671.25272.0f.
  • Iwaya A, Iiai T, Okamoto H, Ajioka Y, Yamamoto T, Asahara T, Nomoto K, Hatakeyama K. Change in the bacterial flora of pouchitis. Hepatogastroenterology. 2006;53:55–59.
  • Swidsinski A, Weber J, Loening-Baucke V, Hale LP, Lochs H. Spatial organization and composition of the mucosal flora in patients with inflammatory bowel disease. J Clin Microbiol. 2005;43(7):3380–3389. doi:10.1128/JCM.43.7.3380-3389.2005.
  • Xu C, Liu Q, Huan F, Qu J, Liu W, Gu A, Wang Y, Jiang Z. Changes in gut microbiota may be early signs of liver toxicity induced by epoxiconazole in rats. Chemotherapy. 2014;60(2):135–142. doi:10.1159/000371837.
  • Chaimanee V, Evans JD, Chen Y, Jackson C, Pettis JS. 2016. Sperm viability and gene expression in honey bee queens (Apis mellifera) following exposure to the neonicotinoid insecticide imidacloprid and the organophosphate acaricide coumaphos. J Insect Physiol. 89:1–8. doi:10.1016/j.jinsphys.2016.03.004.
  • Shi TF, Wang YF, Liu F, Qi L, Yu LS. Influence of the neonicotinoid insecticide thiamethoxam on miRNA expression in the honey bee (Hymenoptera: Apidae). J Insect Sci. 2017;17(5). doi:10.1093/jisesa/iex074.
  • Taira K, Fujioka K, Aoyama Y. Qualitative profiling and quantification of neonicotinoid metabolites in human urine by liquid chromatography coupled with mass spectrometry. PLoS One. 2013;8(11):e80332. doi:10.1371/journal.pone.0080332.
  • Du J, Gridneva Z, Gay MCL, Trengove RD, Hartmann PE, Geddes DT. 2017. Pesticides in human milk of Western Australian women and their influence on infant growth outcomes: a cross-sectional study. Chemosphere. 167:247–254. doi:10.1016/j.chemosphere.2016.10.005.
  • Tang W, Wang D, Wang J, Wu Z, Li L, Huang M, Xu S, Yan D. 2018. Pyrethroid pesticide residues in the global environment: an overview. Chemosphere. 191:990–1007. doi:10.1016/j.chemosphere.2017.10.115.
  • Yuan X, Pan Z, Jin C, Ni Y, Fu Z, Jin Y. 2019. Gut microbiota: an underestimated and unintended recipient for pesticide-induced toxicity. Chemosphere. 227:425–434. doi:10.1016/j.chemosphere.2019.04.088.
  • Laurans L, Venteclef N, Haddad Y, Chajadine M, Alzaid F, Metghalchi S, Sovran B, Denis RGP, Dairou J, Cardellini M, et al. Genetic deficiency of indoleamine 2,3-dioxygenase promotes gut microbiota-mediated metabolic health. Nat Med. 2018;24(8):1113–1120. doi:10.1038/s41591-018-0060-4.
  • O’mahony SM, Clarke G, Borre YE, Dinan TG, Cryan JF. 2015. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav Brain Res. 277:32–48. doi:10.1016/j.bbr.2014.07.027.
  • Tremaroli V, Backhed F. Functional interactions between the gut microbiota and host metabolism. Nature. 2012;489(7415):242–249. doi:10.1038/nature11552.
  • Tolhurst G, Heffron H, Lam YS, Parker HE, Habib AM, Diakogiannaki E, Cameron J, Grosse J, Reimann F, Gribble FM. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-Protein–coupled receptor FFAR2. Diabetes. 2012;61(2):364–371. doi:10.2337/db11-1019.
  • Thomas C, Gioiello A, Noriega L, Strehle A, Oury J, Rizzo G, Macchiarulo A, Yamamoto H, Mataki C, Pruzanski M, et al. TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab. 2009;10(3):167–177. doi:10.1016/j.cmet.2009.08.001.
  • Zelante T, Iannitti RG, Cunha C, De Luca A, Giovannini G, Pieraccini G, Zecchi R, D’angelo C, Massi-Benedetti C, Fallarino F, et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity. 2013;39(2):372–385. doi:10.1016/j.immuni.2013.08.003.
  • Venkatesh M, Mukherjee S, Wang H, Li H, Sun K, Benechet AP, Qiu Z, Maher L, Redinbo M, Phillips R, et al. Symbiotic bacterial metabolites regulate gastrointestinal barrier function via the xenobiotic sensor PXR and Toll-like receptor 4. Immunity. 2014;41(2):296–310. doi:10.1016/j.immuni.2014.06.014.
  • Lamas B, Richard ML, Leducq V, Pham HP, Michel ML, Da Costa G, Bridonneau C, Jegou S, Hoffmann TW, Natividad JM, et al. CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat Med. 2016;22(6):598–605. doi:10.1038/nm.4102.
  • Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt EB, Fu X, Chung Y-M, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472(7341):57–63. doi:10.1038/nature09922.
  • Louis P, Hold GL, Flint HJ. The gut microbiota, bacterial metabolites and colorectal cancer. Nat Rev Microbiol. 2014;12(10):661–672. doi:10.1038/nrmicro3344.
  • Sharon G, Garg N, Debelius J, Knight R, Dorrestein PC, Mazmanian SK. Specialized metabolites from the microbiome in health and disease. Cell Metab. 2014;20(5):719–730. doi:10.1016/j.cmet.2014.10.016.
  • Biesalski HK. Nutrition meets the microbiome: micronutrients and the microbiota. Ann N Y Acad Sci. 2016;1372(1):53–64. doi:10.1111/nyas.13145.
  • Yan S, Tian S, Meng Z, Yan J, Jia M, Li R, Zhou Z, Zhu W. 2020. Imbalance of gut microbiota and fecal metabolites in offspring female mice induced by nitenpyram exposure during pregnancy. Chemosphere. 260:127506. doi:10.1016/j.chemosphere.2020.127506.
  • Zhang H, Meng G, Mao F, Li W, He Y, Gin KY, Ong CN. 2019. Use of an integrated metabolomics platform for mechanistic investigations of three commonly used algaecides on cyanobacterium, Microcystis aeruginosa. J Hazard Mater. 367:120–127. doi:10.1016/j.jhazmat.2018.12.069.
  • Meng Z, Liu L, Jia M, Li R, Yan S, Tian S, Sun W, Zhou Z, Zhu W. Impacts of penconazole and its enantiomers exposure on gut microbiota and metabolic profiles in mice. J Agric Food Chem. 2019;67(30):8303–8311. doi:10.1021/acs.jafc.9b02856.
  • de Wiele T V, Gallawa CM, Kubachka KM, Creed JT, Basta N, Dayton EA, Whitacre S, Laing GD, Bradham K. Arsenic metabolism by human gut microbiota upon in vitro digestion of contaminated soils. Environ Health Perspect. 2010;118(7):1004–1009. doi:10.1289/ehp.0901794.
  • Lu K, Abo RP, Schlieper KA, Graffam ME, Levine S, Wishnok JS, Swenberg JA, Tannenbaum SR, Fox JG. Arsenic exposure perturbs the gut microbiome and its metabolic profile in mice: an integrated metagenomics and metabolomics analysis. Environ Health Perspect. 2014;122(3):284–291. doi:10.1289/ehp.1307429.
  • Richardson JB, Dancy BCR, Horton CL, Lee YS, Madejczyk MS, Xu ZZ, Ackermann G, Humphrey G, Palacios G, Knight R, et al. Exposure to toxic metals triggers unique responses from the rat gut microbiota. Sci Rep. 2018;8(1):6578. doi:10.1038/s41598-018-24931-w.
  • Chi L, Bian X, Gao B, Tu P, Ru H, Lu K. The effects of an environmentally relevant level of arsenic on the gut microbiome and its functional metagenome. Toxicol Sci. 2017;160(2):193–204. doi:10.1093/toxsci/kfx174.
  • Wu F, Yang L, Islam MT, Jasmine F, Kibriya MG, Nahar J, Barmon B, Parvez F, Sarwar G, Ahmed A, et al. The role of gut microbiome and its interaction with arsenic exposure in carotid intima-media thickness in a Bangladesh population. Environ Int. 2019;123:104–113. doi:10.1016/j.envint.2018.11.049.
  • Jonsson AL, Backhed F. Role of gut microbiota in atherosclerosis. Nat Rev Cardiol. 2017;14(2):79–87. doi:10.1038/nrcardio.2016.183.
  • Lindskog Jonsson A, Caesar R, Akrami R, Reinhardt C, Fak Hallenius F, Boren J, Bäckhed F. Impact of gut microbiota and diet on the development of atherosclerosis in apoe −/− mice. Arterioscler Thromb Vasc Biol. 2018;38(10):2318–2326. doi:10.1161/ATVBAHA.118.311233.
  • Zhang S, Jin Y, Zeng Z, Liu Z, Fu Z. Subchronic exposure of mice to cadmium perturbs their hepatic energy metabolism and gut microbiome. Chem Res Toxicol. 2015;28(10):2000–2009. doi:10.1021/acs.chemrestox.5b00237.
  • Chi L, Gao B, Bian X, Tu P, Ru H, Lu K. 2017. Manganese-induced sex-specific gut microbiome perturbations in C57BL/6 mice. Toxicol Appl Pharmacol. 331:142–153. doi:10.1016/j.taap.2017.06.008.
  • Gao B, Chi L, Mahbub R, Bian X, Tu P, Ru H, Lu K. Multi-omics reveals that lead exposure disturbs gut microbiome development, key metabolites, and metabolic pathways. Chem Res Toxicol. 2017;30(4):996–1005. doi:10.1021/acs.chemrestox.6b00401.
  • Liebert CA, Wireman J, Smith T, Summers AO. Phylogeny of mercury resistance (mer) operons of gram-negative bacteria isolated from the fecal flora of primates. Appl Environ Microbiol. 1997;63(3):1066–1076. doi:10.1128/aem.63.3.1066-1076.1997.
  • Bridges KN, Zhang Y, Curran TE, Magnuson JT, Venables BJ, Durrer KE, Allen MS, Roberts AP. Alterations to the intestinal microbiome and metabolome of Pimephales promelas and Mus musculus following exposure to dietary methylmercury. Environ Sci Technol. 2018;52(15):8774–8784. doi:10.1021/acs.est.8b01150.
  • Lin X, Zhao J, Zhang W, He L, Wang L, Chang D, Cui L, Gao Y, Li B, Chen C, et al. Acute oral methylmercury exposure perturbs the gut microbiome and alters gut-brain axis related metabolites in rats. Ecotoxicol Environ Saf. 2020;190:110130. doi:10.1016/j.ecoenv.2019.110130.