Next Article in Journal
Role of Redox-Induced Protein Modifications in Spermatozoa in Health and Disease
Previous Article in Journal
The DJ-1-Binding Compound Exerts a Protective Effect in Both In Vitro and In Vivo Models of Sepsis-Induced Acute Kidney Injury
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Impact of Diquat on the Intestinal Health and the Composition and Function of the Gut Microbiome

Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin 300052, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Antioxidants 2025, 14(6), 721; https://doi.org/10.3390/antiox14060721
Submission received: 27 March 2025 / Revised: 7 June 2025 / Accepted: 10 June 2025 / Published: 12 June 2025

Abstract

Diquat (DQ) is extensively utilized as a herbicide in farming, and its intake can result in serious systemic toxicity due to its induction of oxidative stress (OS) and disruption of intestinal homeostasis. The gastrointestinal tract is one of the first systems exposed to DQ, and damage to this system can influence the general health of the host. Our review summarizes the toxic effects of DQ on the intestinal barrier integrity, gut microbiome, and microbial metabolites (e.g., short-chain fatty acids [SCFAs], bile acids). By elucidating the mechanisms linking DQ-induced OS to gut dysbiosis, mitochondrial dysfunction, and inflammation, our work provides critical insights into novel therapeutic strategies, including probiotics, antioxidants (e.g., hydroxytyrosol, curcumin), and selenium nanoparticles. These findings address a pressing gap in understanding environmental toxin-related gut pathology and offer potential interventions to mitigate systemic oxidative damage.

1. Introduction

Pesticides are the primary reason for toxicosis-associated accidental deaths in China, which is the leading producer of pesticides, with the United States (US) and Argentina following. As paraquat (PQ) ingestion has a particularly high mortality, its sale and use have already been limited in many countries. Consequently, diquat (DQ) has become a widely used substitute for PQ [1]. DQ was first produced in 1955 by the Imperial Chemical Industries (ICI), a British company, which afterwards realized its value as a herbicide and introduced it to the market in 1958 [2,3,4]. DQ possesses a high binding appetency to soil and other organic substances and exhibits a high solubility in water [5]. Hence, DQ enters aquatic ecosystems easily through direct discharge from factories and agriculture, and does harm to humans, ducks, fish and other aquatic animals [6]. Its exposure approaches include the digestive tract, the respiratory tract, the eyes, and the skin [3,7], resulting in dysfunction of many organs such as the kidneys, brain, heart, and liver [1,8]. In recent years, the increasing cases of DQ toxicoses have attracted global attention, and DQ was classified as a potential carcinogen by the US Environmental Protection Agency (EPA) [9]. Until now, there is no specific antidote for DQ poisoning [10]. Existing studies show that DQ poisoning may be related to oxidative stress (OS), inflammatory response, and the induction of apoptosis [11,12]. Nevertheless, the specific mechanisms of DQ-induced injury are largely unclear.
As everyone knows, the main approach of pesticide exposure is the digestive tract [13,14]. Recently, the research emphasis has turned to how pesticide exposure affects the host intestinal flora, and evidence supports the possibility that DQ could disrupt the structure and function of the gut microbiome, resulting in host metabolic disorders [15,16]. The human gut, being the largest barrier organ by surface area, has a complicated barrier structure that serves as a selectively permeable shield, responsible not only for nutrient absorption but also for protection against detrimental external factors [17,18]. It is composed of four major layers: the microbiome in the intestinal lumen, responsible for decolonizing pathogenic bacteria from the gut (a biological barrier); the unstirred water layer, comprising digestive fluid and other substances released by the intestinal epithelial cells (IECs) to inhibit bacterial attachment (a chemical barrier); the mucosal layer (including antimicrobial substances generated by IECs and Paneth cells); and lined IECs (a mechanical barrier) that act as the main constituent of the intestinal wall linked through tight junctions (TJs) [17,19,20,21]. TJs (claudin, ZO-1 and occludin) are intercellular adhesion complexes that serve as gatekeepers of the paracellular space [22].
The gut microbiota, a varied ecosystem, lives within the digestive tract and contains at least 1013–1014 microbial cells and more than 2000 diverse species, involving bacteria, viruses, fungi and parasites [23,24]. The primary function of gut microbiota is to regulate the host metabolism as a ‘central metabolic organ’ and defend against harmful stimuli and pathogens [25]. The gut microbiota is crucial for preserving the integrity of IECs and beneficial for maintaining the host’s defense system homeostasis through generating antimicrobial compounds, facilitating mucus secretion, and inhibiting the colonization of pathogens [26]. A dynamic but relatively stable gut microbiota contributes to human health, while dysbiosis may lead to inflammatory responses and various diseases such as colon cancers, obesity, and diabetes [27,28]. Figuring out the relationship between DQ and the functionality of the intestinal microbiota can provide a better understanding of DQ-induced toxicity and new therapeutic strategies in DQ-related damage.
This review summarizes the potential detrimental effect that DQ has on the intestine, with special attention paid to the intestinal microbiota. Firstly, we expound on impact of DQ’s inherent toxicity on the gut. Next, we discuss its effect on the intestinal microbiome and its metabolites.

2. Toxic Effects of DQ on the Intestine and the Gut Barrier

DQ crosses the cell membrane primarily through diffusion and, to a lesser degree, by active transport through cation pumps [3]. DQ presents a direct toxic effect on the IECs and firstly makes contact with the intestinal mucosa, resulting in extensive damage. Meanwhile, the release of a large number of inflammatory factors can directly or indirectly lead to intestinal damage, impairing the intestinal barrier, consequently accelerating DQ absorption. The TJ between IECs is one of the essential structural bases for preserving the integrity of the gut barrier [29]. Accumulating evidence has demonstrated that OS destroys the TJs in many ways, leading to intestinal epithelial barrier dysfunction [30,31]. According to research by Tian et al. (2023), OS induced by DQ results in morphological abnormalities and functional damage in the small intestine [32].

2.1. Oxidative Stress and Inflammation

A survey of existing studies on pesticides showed that all pesticides can cause OS [33], which involves an imbalance between the production of reactive oxygen species (ROS) and the capability of the defense system to eliminate the superfluous ROS [34,35,36]. Normally, there is an actional equilibrium between the oxidants and antioxidants in biological systems [37]. Excessive ROS can damage cellular proteins, DNA, and lipids, causing fatal cellular injury [38]. The gut sits at the interface between the organism and its lumen environment [39], serving as a barrier against invading intestinal pathogens, and thus is more susceptible to oxidative injury [40,41,42,43,44]. The gut barrier consists of monolayer cells and is the main site of OS response [45,46]. OS mainly injures intestinal health by inducing cell injury, inflammation and gut dysfunction [47,48]. Some studies have demonstrated that the overproduction of ROS in the intestine can disrupt the epithelium function, increase the permeability of the gut, damage IECs and disturb nutrient absorption [44,49,50]. For example, studies have reported that some oxidants augment the permeability of the gut barrier through disturbing TJs, accompanied by the disruption of the nuclear factor kappa-B (NF-κB) signal pathway and inflammation [46,51]. It has been reported that OS interacts with inflammation, ROS contributes to inflammation, and in turn, inflammation contributes to the production of ROS [52,53,54,55]. Studies have found that OS can activate the NF-κB signal pathway, which in turn facilitates the release of proinflammatory cytokines [56,57]. In addition, extensive studies have shown that OS can disrupt the natural structure of the small gut, involving increasing crypt depth (CD) and decreasing villus height (VH) and the ratio of VH to CD (V/C), which can be regarded as an indicator of the absorption ability [58,59,60]. Integrating these results, we speculate that therapy to reduce oxidative injury and inflammatory response may be beneficial in maintaining the function of the epithelial barrier, as well as intestinal and host health.
Moreover, OS is regarded as a key factor in disturbing the balance of the intestinal microbiome and notably reducing the microbiota diversity [61,62]. Recently, OS has been reported to influence the structure as well as the functionality of the gut microbiota [63] and change the metabolites [64]. Furthermore, the metabolites of the microbiome are also crucial for intestinal microbiome-mediated redox adjustment [65,66]. Some metabolites, including short-chain fatty acids (SCFAs), polyphenolic and tryptophan derivatives, lipopolysaccharides (LPS), and trimethylamine-N-oxide (TMAO), were found to modulate OS [67,68,69,70]. Research has found that ingesting probiotics alone or dietary supplementation with probiotics could alleviate OS and change the activities of pivotal antioxidant enzymes [71]. These findings can offer novel insights and strategies for the therapy of intestinal OS illnesses.

2.2. The Role of OS and Inflammation in DQ-Induced Intestinal Injury

DQ can produce superoxide anion radicals by the molecular oxygen, which breaks the redox equilibrium of the gut, leading to OS and inflammation [72,73,74,75]. The ability to induce OS is greater than that of other herbicides due to DQ’s high value of redox potential [76]. Extensive studies have shown that OS ruins the structure of the gut and results in apoptosis in enterocytes [77,78]. It was also reported that OS can induce essential mitochondrial channel opening and activate the apoptosis pathway, which depends on mitochondria, causing cell death [79,80].
The main antioxidant defense machineries for humans consist of antioxidants and antioxidant enzymes, involving superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-Px) [81]. DQ was reported to elevate the content of malondialdehyde (MDA) and reduce CAT, SOD and GSH-Px in jejunal mucosa [82,83,84,85]. In addition, studies have found that SOD can effectively boost antioxidant ability, maintain mucosal barrier integrity, and inhibit pro-inflammatory responses [86,87]. Li et al. obtained a novel SOD gene from the Hydrogenobacter thermophilus strain (Ht), and their study demonstrated that HtSOD mitigates OS and intestinal injury by decreasing total ROS in DQ-treated mice [42].
Extensive evidence has revealed that the nuclear factor erythroid 2-related factor 2 (Nrf2) signal pathway and mitophagy can mitigate OS. It was reported that Nrf2 primarily adjusts the transcription of antioxidant genes, including CAT, SOD, and heme oxygenase-1 (HO-1), by regulating the antioxidant defense system [88,89,90]. Mechanistic research in IPEC-J2 cells exposed to DQ demonstrated that hydroxytyrosol (HT) alleviated intestinal oxidative impairment through activating the Nrf2 signal pathway and facilitating mitophagy. HT, a natural polyphenolic substance, mainly exists in olives and presents potent antioxidant capacity [91]. DQ induced OS and damaged intestinal barrier function, whereas HT mitigated these adverse phenomena. This research showed that targeting the Nrf2 signaling pathway and mitophagy may become a hopeful tactic for curing OS-related damage induced by DQ [92]. For instance, recent advances in selenium research revealed that dietary selenium nanoparticles (SeNPs) triggered the Nrf2 pathway to suppress the activation of inflammasome caused by OS in mice treated with DQ, consequently alleviating OS-related intestinal barrier dysfunction [81,93].
KEGG analysis of an original research showed that differentially expressed genes (DEGs) in mice after exposure to DQ were primarily enriched in the NF-κB signaling pathway [81]. Evidence supported that Nrf2 can inhibit the activation of NLRP3 inflammasomes, which are crucial for the processing and release of inflammatory cytokines, including IL-6, IL-18, IL-1β and TNF-α [94]. Qiao et al. (2022) demonstrated that DQ exposure increased the level of the NLRP3 inflammasome and subsequently caused an increase in IL-18 and IL-1β in mice [95]. Another study has shown that the concentration of IL-6 and TNF-α is significantly elevated in the piglet model of DQ-induced OS [96]. TNF-α, a proinflammatory cytokine, plays an important role in systemic inflammation. The collected data confirmed that TNF-α can result in an increase in intestinal epithelial TJ permeability [97,98].
As mentioned before, due to its characteristic biological structure and functional location, the gut is especially vulnerable to DQ, which causes severe OS. Thus, regulating OS has become a significant means to ameliorate the damage induced by DQ. Measures such as developing antioxidants and understanding their mechanisms and pathways can help alleviate OS induced by DQ and reduce the risk of intestinal damage.

2.3. Mitophagy

DQ is also identified as leading to mitochondrial damage. Studies have revealed that mitochondrial dysfunction is involved in DQ poisoning [12,82,99]. IECs contain abundant mitochondria, which are the main production place and the primary target of ROS [82,100,101,102]. Once exposed to external noxious stimuli, the impaired mitochondria produce excessive ROS, which further worsens oxidative damage [103,104,105]. Chen et al. (2021) [106] found that DQ exposure led to ROS accumulation, which destroyed mitochondrial DNA (mtDNA) and mitochondrial enzymes and caused mitochondrial dysfunction in the jejunum of weanling piglets. Such damaged mitochondrial function, in turn, exacerbated the production of mtROS, creating a vicious cycle. Moreover, research has shown that the accumulation of mtROS reduces the microbial diversity of the gut as well as antimicrobial defenses [107,108]. In this regard, alleviating mitochondrial disorders may be a promising strategy for the improvement of OS-related damage caused by DQ.
Previous investigations have demonstrated that OS can trigger mitophagy in DQ-induced intestinal damage [109]. It has been reported that DQ-induced OS could impair the function of the epithelium in piglets, with the mitochondrial disorder of the jejunal mucosa and mitophagy [83,110]. Mitophagy can eliminate excess ROS through phagocytosing impaired mitochondria and degrading them [111,112,113]. It was reported that OS can trigger mitophagy through the PINK1-Parkin signaling pathway [114,115]. In existing studies, researchers found that DQ exposure induces the production of autophagic vesicles and augments mitophagy-associated proteins, indicating that the mitophagy pathway has been initiated [92]. Recent studies have indicated that mitophagy plays a protective role in preventing cellular death triggered by OS in DQ-treated piglets. This process involves the selective engulfment of impaired mitochondria by autophagosomes, followed by their subsequent breakdown within lysosomes. Such cellular machinery not only maintains intestinal epithelial function but also decreases additional oxidative injury [82].
Recently, a growing body of studies have indicated that metabolites derived from gut microbiota play a significant role in modulating OS through microbial activity in the intestinal environment. SCFAs, especially acetate, propionate, and butyrate, are metabolic byproducts generated by beneficial gut microbiota during the breakdown and fermentation of dietary fiber [116,117,118]. These SCFAs serve as crucial energy substrates for IECs, facilitating their growth and differentiation. Additionally, they play a significant role in modulating gut functionality, immune responses, and preserving the homeostasis of microbial communities. In addition, SCFAs have been confirmed to possess antioxidant abilities, and among them, butyrate has received particular attention [119]. Extensive studies conducted both in animal models and cell cultures have verified that butyrate possesses the ability to effectively alleviate OS within the intestinal environment [120,121,122,123,124]. A study has shown that butyrate serves as a signal molecule safeguarding the liver from OS through stimulating the Nrf2 signal pathway in rats [124]. Furthermore, butyrate also possesses numerous functions, involving providing the major energy for intestinal cells, promoting TJ protein formation [119,125], suppressing the proliferation of pathogens, contributing to the growth of enterocytes, and restraining the intestinal inflammation [126,127,128]. In addition, some researchers indicated that butyrate has the capacity to restore mitochondrial energy deficiencies [129]. Data from Wang et al. (2019) [55] indicated that DQ injection caused serious intestinal OS in pigs, while butyrate relieved intestinal OS and inflammation and improved mitochondrial function through selectively inducing mitophagy. Wang’s research revealed that butyrate notably elevated protein levels of PINK1 and Parkin in mitochondria. Moreover, Lee et al. (2012) found that butyrate increased the survival of hamster ovary cells through recruiting Parkin and inducing mitophagy to eliminate damaged mitochondria [130]. Furthermore, previous studies have demonstrated that elevated butyrate concentrations upregulated mitophagy-related genes in cell lines from autistic boys under OS [131].
As previously mentioned, Nrf2 reduces OS through regulating the transcription of genes involved in antioxidant defense mechanisms, whereas mitophagy eliminates impaired mitochondria at the source. The synergistic effect of these two processes contributes to preserving cellular stability and improving the impairments induced by OS. Nevertheless, the precise interplay between Nrf2 and mitophagy is not yet entirely clear and needs further investigation.
The molecular mechanisms mentioned above are outlined in Figure 1.

2.4. Impact of DQ on Intestinal Barrier Integrity

It is generally acknowledged that the gut barrier, as the main shield against the harsh conditions within the lumen of the intestine, is critical for safeguarding the organism against toxins, antigens, and pathogens [132,133,134]. The intestinal epithelium comprises enterocytes, Paneth cells, and goblet cells, collectively constituting the principal cellular constituents of the intestinal barrier system that orchestrates mucosal defense mechanisms [135]. Goblet cells, a significant component of IECs, are the primary source of mucin secretion. In the gut, mucin 2 (MUC2) is the predominant mucin secreted by goblet cells [136] and serves as a key regulatory gene for the intestinal mucus layer, playing a significant role in maintaining intestinal barrier integrity [137]. Research has found that DQ exposure decreases the number of goblet cells, thereby diminishing MUC2 secretion. The deficiency of MUC2 compromises the integrity of the inner mucus layer, permitting pathogenic bacterial–epithelial interactions that trigger innate immune activation and chronic mucosal inflammation [138]. Animal studies have revealed that MUC2-deficient mice have chronic colonic inflammation [139].
Studies have demonstrated that DQ impaired the integrity of the gut barrier in mice, leading to fragmented brush borders and a disarrayed epithelium [83]. The excessive ROS in the intestinal epithelium can elevate intestinal permeability, enabling the translocation of detrimental luminal components into the bloodstream (as described in Figure 2) and elevating the likelihood of systemic inflammatory responses [140]. Gut integrity plays a significant role in evaluating gut health and could be quantified by various measurable parameters, such as diamine oxidase (DAO), D-lactate (DLA), VH, and CD [141]. DAO, an enzyme found within cells, is primarily produced and distributed in the intestinal epithelium [142]. DLA is a special byproduct generated by microbial metabolic processes. Increased serum concentrations of DAO and DLA commonly suggest gut barrier injury [143,144]. Chen et al. (2022) demonstrated that DQ-treated pigs showed elevated serum DAO and DLA levels, verifying the impairment of gut barrier integrity [145].
VH and CD serve as direct morphological parameters for assessing the structural integrity of the gut mucosa [146]. TJs form the fundamental building blocks of the gut barrier, with occludin being the earliest discovered TJ protein, while ZO-1 and claudin play critical roles in maintaining the physiological and structural integrity of the paracellular barrier [147]. It has been reported that claudins are crucial components of TJs and determine their barrier properties and paracellular permeability [148]. Both animal and cell culture studies showed that DQ-related OS compromises the integrity of IECs, evidenced by the disruption of TJs and the reduced viability of IECs, ultimately leading to the dysfunction of the nutrient metabolism [149,150,151]. Additionally, Wen et al. (2020) found that DQ negatively impacted the morphological development of the small intestine, decreasing VH and the V/C ratio, while also downregulating the expression of ZO-1 and occludin proteins in piglet intestinal tissue [152].
Furthermore, research has found that the atrophy of intestinal villi and barrier dysfunction caused by OS were partially related to the programmed cell death of IECs [153]. It was found that the impairment of the gut barrier is always accompanied by the apoptosis of IECs [154,155,156]. As everyone knows, apoptosis is modulated by various molecular mechanisms, with significant involvement from the Bcl-2 and caspase protein families [157]. Chen et al. (2022) revealed that DQ exposure obviously increased the expression levels of caspase-9, caspase-3, and Bax in the mucosal layers of the duodenum and jejunum, indicating that DQ induced apoptosis of IECs [145]. Furthermore, studies showed that DQ exposure led to atrophied intestinal villi, featuring fragmented intestinal brush borders and disordered epithelial layers, resulting from the apoptosis and shedding of IECs [21,158].

3. Effects of DQ on the Composition and Function of the Intestinal Microbiome

Increasing evidence has confirmed the crucial function of the intestinal microbiome in the physiological and pathological states of the body. Studies have revealed that the intestinal microbiome helps repair intestinal mucosal barrier injury [159] and also plays vital roles in modulating host OS and inflammation [160,161,162]. It has been reported that DQ reduced gut microbiota diversity, whereas HtSOD alleviated the decrease in microbial diversity caused by DQ and contributed to the growth of beneficial microbiota [42]. The distinct gut microbial profiles between DQ-exposed animals and the normal group have been exhibited in several studies.
Yuan et al. (2024) [81] found altered ileal mucosal microbiota composition in DQ-treated piglets. In comparison with normal piglets, DQ exposure reduced Acidobacteria populations while elevating the levels of Clostridium and Turicibacter. Clostridium genera are Gram-positive obligate anaerobic bacteria and are usually deemed as pathogenic microorganisms [163], while Turicibacter might influence gut health and contribute to the development of multiple disorders, such as diabetes and inflammation [164]. The elevated levels of Clostridium and Turicibacter showed exacerbated ileal inflammation resulting from DQ exposure. 16S rRNA analyses in the study by Wu et al. (2022) suggested that DQ administration disrupted the microbial balance in rats, markedly reducing Firmicutes and Lactobacillus populations while augmenting Proteobacteria abundance [165]. This is an inconsistent result to that of Yuan et al., which we hypothesize may be due to interspecies differences. Firmicutes, a Gram-positive bacterial phylum, constitutes the predominant microbial group in healthy human gut microbiota [17,166]. It encompasses various beneficial bacterial species capable of metabolizing SCFA salts, including acetate and lactate, thus influencing the balance of symbiotic microorganisms in the intestine, alleviating gut inflammation, and maintaining immune homeostasis [167,168,169]. Proteobacteria phylum contains the majority of conditional pathogens, such as Helicobacter, Vibrio, Salmonella and Escherichia [170]. There is a broad consensus that numerous bacterial species within the Proteobacteria phylum can induce persistent gut inflammation and tissue damage in piglets [171,172]. STAMP analyses in this study showed notably elevated levels of conditional pathogens Escherichia coli and Proteobacteria, alongside reduced populations of potentially beneficial microbes, including Firmicutes, Lactobacillus, and Akkermansia, in DQ-treated rats. Pearson’s correlation analysis revealed that beneficial microorganisms, including Butyricicoccus and Faecalibacterium, showed inverse relationships with MDA levels, further verifying the link between microbial community changes and DQ-induced OS [165]. Han et al. (2023) demonstrated that DQ exposure decreased Firmicutes populations, consistent with previous research findings, while simultaneously increasing Bacteroidetes levels in mice [173]. Recent research has indicated that the Firmicutes/Bacteroidetes (F/B) ratio is positively related to the antioxidant ability in piglets [174] and is recognized as a significant indicator of the health of the gut microbiota [175]. Furthermore, an article reported that DQ exposure increased Parabacteroides populations in mice, consistent with another study exhibiting a positive correlation between Parabacteroides abundance and OS markers in murine models [176]. The collected data confirmed that Parabacteroides is a core member of the intestinal microbiome and is related to the development of OS and inflammation [177]. Yang et al. (2024) found that DQ markedly decreased Lactobacillus and Alistipes populations in broilers [178]. Alistipes serves as a key contributor to SCFAs production, including acetate and propionate [179]. Research has indicated that Alistipes potentially exerts protective influences against certain pathological conditions, involving colitis, cardiovascular diseases, and tumor immunotherapy [180]. This further verifies that DQ can reduce the populations of advantageous microbial species, thus impairing gut homeostasis. The results of Fu et al. (2021) indicated that DQ exposure increased the populations of Ruminococcaceae UCG-005 and Eubacterium coprostanoligenes in weaned piglets [171]. A previous study demonstrated that Ruminococcaceae UCG-005 is a stable constituent of the intestinal microbiome in piglets [181]. Research has revealed that Ruminococcaceae UCG-005 is positively related to metabolic diseases and chronic inflammation in weaned piglets [182]. Furthermore, another study has indicated that the increase in Ruminococcaceae UCG-005 in piglets exposed to DQ implies the exacerbation of the gut milieu because the population levels of Ruminococcus within the gastrointestinal system significantly influence the occurrence of diarrhea [183,184]. Eubacterium coprostanoligenes is widely recognized for its capacity to transform cholesterol into coprostanol, thereby decreasing circulating cholesterol levels. Some findings have suggested that Eubacterium coprostanoligenes populations are elevated in correlation with rising serum cholesterol concentrations in piglets exposed to thermal stress [185]. Based on the data acquired, we hypothesize that OS induced an elevation in circulating cholesterol levels, subsequently elevating the abundance of Eubacterium coprostanoligenes.
Recent studies have indicated that there exists a connection between mitochondria and the intestinal microbiome. On the one hand, the intestinal microbiome has been found to modulate pivotal transcriptional factors that play crucial roles in the process of mitochondrial biogenesis. In addition, the intestinal microbiome and its byproducts, including SCFAs and secondary bile acids, also play a stimulative role in energy generation, OS, and inflammation reduction through diminishing TNF-α-driven responses and inflammasomes like NLRP3 [95]. Furthermore, mitochondrial events, especially mtROS generation, are crucial for modulating the intestinal microbiome through regulating the functionality of the gut barrier [186,187]. One study has shown that selenium deprivation disrupted redox homeostasis and altered the intestinal microbiome composition, which is more vulnerable to gut barrier impairment upon DQ exposure. This effect is mediated via the Nrf2-dependent regulation of NLRP3 inflammasome signaling in murine models subjected to DQ stimulation. Nutritional intervention using biologically synthesized SeNPs produced by Lactobacillus casei ATCC 393 significantly improved the impairment of the gut barrier through increasing the antioxidative ability, ameliorating mitochondrial integrity and functionality, and preserving intestinal ecological balance through the Nrf2-regulated NLRP3 signal cascade [95].
This field would be worth investigating further, as although there is information available sketching the effects of DQ on the composition and functionality of intestinal microbiota, the exact mechanisms remain challenging to completely characterize owing to interspecies and individual differences, necessitating further comprehensive research. Various lifestyle determinants contribute to the unique composition of individual intestinal microbiota, including dietary habits, probiotic consumption, age, environmental exposures, and exercise regimens [188,189].
In conclusion, DQ exerts toxic effects on intestinal microbial communities through modifying their compositional profile and functional characteristics. These microbial changes may provide valuable biomarker potential for evaluating DQ-induced toxicity.

4. Impact of DQ on Metabolites

Microbial-derived metabolites, including SCFAs, tryptophan metabolites, and bile acids (BAs), have been demonstrated to significantly influence gut homeostasis [190]. These metabolites from different microorganisms have been shown to regulate intestinal physiology and influence metabolic processes across multiple organ systems, including the liver, muscle, and brain [191,192,193]. They possess dual regulatory capabilities, facilitating the development and activity of immunoregulatory cells while simultaneously restraining pro-inflammatory pathways, thereby preserving both intestinal and systemic physiological balance in the host [194]. BAs are synthesized through cholesterol catabolism, undergoing hepatic conversion to primary forms (PBAs) before intestinal transit, where microbial biotransformation generates secondary metabolites (SBAs), preserving physiological balance via enterohepatic recirculation [195]. Studies in humans and mice have indicated that BAs are involved in modulating intestinal inflammation, tumorigenesis, and immune function [196,197]. SBAs undergo microbial-mediated conversion to generate TUDCA, a bioactive compound shown to stimulate Nrf2 signaling, increase antioxidative enzyme expression, and exert antioxidative effects [195,198]. Thereby, the regulation of intestinal microbial communities and their metabolic byproducts emerges as a promising therapeutic approach for ameliorating OS in the gastrointestinal tract. It was reported that polyphenolic substances exert beneficial effects on microbial ecosystem stability and normal metabolism [199,200]. As previously mentioned, HT is a natural polyphenolic compound and possesses potent antioxidant ability [201]. Wen et al. (2024) [137] reported that DQ caused disorders in BA metabolism, including lower levels of PBAs, hyocholic acid (HCA), hyodeoxycholic acid (HDCA), and TUDCA, while treatment with HT partially reversed these alterations. This study also revealed a positive association among the expression levels of HCA, Nrf2, and CAT, suggesting that HCA may possess antioxidant properties. Furthermore, HDCA was found to enhance the activity of antioxidative enzymes and then alleviate oxidative damage and inflammation. Moreover, some studies have revealed that HDCA has the potential to serve as an intrinsic regulator that inhibits inflammatory signaling mechanisms [202]. Accumulating evidence has demonstrated that TUDCA is capable of modulating the activity of antioxidative enzymes, which helps reduce OS [203,204,205]. This perspective was further supported by a positive association observed among the expression levels of TUDCA, Nrf2 and CAT. It is widely recognized that butyrate-producing bacteria are predominantly classified within the Firmicutes phylum, among which Faecalibacterium prausnitzii and Roseburia are acknowledged as the most potent butyrate producers. Notably, Faecalibacterium prausnitzii serves as a principal contributor to colonic butyrate levels due to its high relative abundance and efficient butyrogenic pathways [206]. In another study that also used HT as an intervention, researchers reported that HT administration counteracts the DQ-induced decrease in the relative abundance of Firmicutes and butyrate levels in mice, implying a potential connection between butyrate and the antioxidant ability enhanced by HT [173].
Studies have revealed that DQ decreases the production of microbial tryptophan, thereby decreasing its availability in the host [207]. Tryptophan and its metabolites are recognized for their function as signaling molecules that facilitate communication between the intestinal microbiome and host cells [208]. Research has found that OS triggered by DQ may increase tryptophan metabolism in pigs [84,207], and this metabolic shift drives indoxyl sulfate (IS) overproduction. IS, a uremic toxin, builds up in plasma as kidney function declines, accelerating the progression of chronic kidney disease (CKD) [209]. In some situations, IS can be beneficial to the host, most markedly by inhibiting inflammation [210] and enhancing gut barrier function [211]. Nevertheless, in patients with CKD, impaired kidney function results in a significant elevation of IS levels in the plasma, inducing detrimental effects [212]. Tryptophanases produced by gut bacteria transform tryptophan into indole, which is subsequently absorbed and transformed by the host into IS. TDO, the hepatic rate-limiting enzyme governing systemic tryptophan catabolism, mediates the oxidative cleavage of tryptophan into kynurenine pathway metabolites [213,214]. Previous studies have demonstrated that DQ exposure can elevate the TDO mRNA level in the liver of piglets [84]. Researchers speculated that the observed decrease in circulating tryptophan levels may be mechanistically linked to the DQ-induced upregulation of hepatic TDO activity [207]. The collected data confirmed that Bacteroides species predominantly harbor the most abundant tryptophanases in the intestines of most individuals. Research has found that DQ exposure increased the abundance of Bacteroides [63]. Researchers identified a widely distributed family of tryptophanases in gut-commensal Bacteroides and found that the targeted ablation of this gene eliminated the generation of indole in vitro [209]. This gives us some new insights: modulating the gut microbiota to reduce the indole concentration, thus decreasing IS in the circulation, may offer a novel strategy to lower the detrimental effects of kidney injury caused by DQ (the gut–kidney axis). Studies have revealed that most human-related bacterial species capable of generating indole are typically low-abundance colonizers of the intestine or pathogens [215,216]. This information may help us better comprehend, forecast, and reprogram IS levels in vivo. For instance, researchers demonstrated that through deleting or changing a single gene and its orthologs, they could clear or obviously decrease the content of urinary IS. In addition, it was also observed that rationally the changing diet could contribute to the proliferation of a tryptophanase-negative Bacteroides species into a model intestinal community, proving the feasibility of utilizing dietary interventions to reduce the microbiota’s ability to generate indole [209].
In the research by Fu et al. (2021) [171], the effects of DQ on the intestinal metabolic profiles was investigated. 3-methyldioxyindole is an oxidative metabolite of 3-methylindole, generated by colonic bacteria, and it plays a role in tryptophan metabolism. In Fu’s results, a notable reduction in 3-methyldioxyindole was found in DQ-treated weaned piglets. Correlation analysis helped identify several bacterial genera that may play a role in host metabolism. The levels of Acidaminobacter showed a negative relationship with two metabolites and a positive relationship with eight metabolites, while Terrisporobacter exhibited a positive relationship with one metabolite and a negative relationship with six metabolites. Integrating these results, researchers suggested that the destruction of intestinal microbial structure and metabolic balance is likely the primary factor contributing to the reduction in the antioxidative ability of DQ-exposed piglets.
Studies have found that the intestinal microbiome also affects host health by influencing the host metabolome [217,218]. Modifications in the structure of the intestinal microbiome can alleviate host OS through altering serum metabolomics. It was reported that HT administration increased the levels of multiple serum metabolites that help mitigate OS [173]. As an example, 20-carboxy arachidonic acid undergoes metabolism through cytochrome P450 omega-oxidase, generating 20-hydroxyeicosatetraenoic acid, a metabolite with demonstrated antioxidative and anti-inflammatory activities [219]. Likewise, 3-hydroxytetradecanoyl carnitine plays a role in mitochondrial activity and oxidation [220]. Malic acid serves as a crucial intermediate metabolite of the tricarboxylic acid cycle (TCA). Evidence has demonstrated that it increased the antioxidative ability in the liver of fish [221].

5. Several Therapies to Alleviate the Toxic Effects of DQ

Out of the current treatment strategies, probiotics have obtained the most attention [222]. Studies have revealed that probiotics can exhibit the antioxidative capacity via diverse approaches, including upregulating the antioxidases in the host and modulating intestinal microbiota [223]. As is well-known to all, lactic acid bacteria (LAB) possess antioxidative and immunoregulatory abilities and are regarded as ideal probiotics [224]. It was reported that LAB could be utilized as a supplement to regulate Nrf2 and NF-κB signal pathways, and ameliorate intestinal inflammation [225]. The dominant genus among LAB is Lactobacillus [189]. It was reported that the genus Lactobacillus is positively related to serum total antioxidant [63,226] and can suppress ROS generation through restraining the activities of critical enzymes like NADPH oxidase and increase the ROS elimination capacity of the antioxidative system through modulating Nrf2 and NF-κB signaling pathways [227]. Research has shown that Lactobacillus can suppress the expression of TNF-α by the NF-κB pathway to relieve intestinal inflammation [225]. Feng et al. (2023) found that Lactobacillus-derived extracellular vesicles (EVs) [228,229] obtained from piglets could elevate antioxidant activity through ameliorating the gut barrier and reorganizing gut microbiota in mice after exposure to DQ [230]. LAB-EVs reshaped the gut microbiota through reducing the growth of detrimental bacteria like Enterococcus while promoting the growth of beneficial bacteria, including Parasutterella and Erysipelatoclostridium. As a result, these alterations ameliorated the intestinal barrier function, as demonstrated by elongated intestinal villi; elevated the activities of the antioxidative enzymes; and alleviated the oxidative injury. Spearman’s correlation analysis exhibited consistent results: Erysipelotrichales and Parasutterella showed a positive relationship with the antioxidative enzyme activities and a negative relationship with oxidative injury. Another study has shown that the inclusion of Lactiplantibacillus plantarum P8 in the diet facilitated an amelioration of the antioxidative ability, microstructure, and barrier integrity in the jejunal mucosa and a reduction in apoptosis in DQ-treated piglets. Transcriptome analysis of the jejunum further revealed that the positive effects of P8 might be correlated with the modulation of the NF-κB signal pathway [231].
The gut plays a significant role in preserving an organism’s arginine balance and arginine absorption, endogenous synthesis, and metabolism [232]. It was reported that OS reduced the content of serum arginine in piglets [85], and supplementation with arginine can diminish OS through elevating the total antioxidative ability in piglets [233]. Furthermore, another study has shown that arginine could maintain barrier function and decrease bacterial translocation in the gut of mice [234]. Research has demonstrated that supplementation with arginine notably decreased intestinal CD and TNF-α mRNA levels in jejunum exposed to DQ. This study supports that arginine supplementation in the diet promotes the gut health via ameliorating the intestinal morphology, modulating arginine availability and lowering inflammatory cytokine levels [149].
Resveratrol, a polyphenolic phytoalexin derived from plants like peanuts, has acquired significant attention for its antioxidative and anti-apoptotic properties. Recent studies have indicated that the administration of resveratrol as a dietary supplement elevated the antioxidative capacity in the host [235,236]. Additionally, resveratrol possesses a potent inhibitory effect on ROS generation and exhibits a wide range of biological activities, including anti-inflammatory, anti-obesity, and anti-aging properties [237,238]. Apigenin, a naturally occurring phytochemical flavonoid, is found in various plant-based foods, such as fruits and vegetables [239]. Recent studies have indicated that apigenin exhibits potential antioxidative, anti-apoptotic, and anti-inflammatory properties [239,240,241,242]. Zhou et al. (2022) [243] found that resveratrol and apigenin could reduce the MDA level and increase SOD and GSH-PX levels in DQ-treated pullets. Furthermore, resveratrol upregulated the levels of HO-1 mRNA in ileac and jejunal tissue, while apigenin increased NRF2 and HO-1. Both resveratrol and apigenin upregulated the mRNA levels of claudin-1 and ZO-1 in the ileum after exposure to DQ. These results demonstrated that resveratrol and apigenin supplementation alleviated OS via the Nrf2 signal pathway and improved intestinal barrier function in pullets exposed to DQ to a certain extent. Curcumin (CUR), a diketone substance derived from the rhizomes of Curcuma longa, is known for its various biological properties, including antioxidative, anti-inflammatory, and antiviral effects [244,245]. Research by Wu et al. (2024) revealed that the concentrations of acetate and total SCFAs in the cecum of broilers treated with CUR were notably elevated compared to those in the DQ group, suggesting that CUR facilitated acetate generation [246]. This might be because Ruminococcaceae_Clostridium populations in the CUR group obviously increased. Recent studies have indicated that Ruminococcaceae shows a crucial effect on degrading starch and fiber, thereby facilitating the generation of SCFAs [247]. As the primary component of SCFAs, acetate is directly related to the content of total SCFAs. Furthermore, the beneficial effects of CUR on preserving cecal physiological equilibrium facilitated acetate generation. Additionally, the LEfSe analysis in Wu’s study indicated that in the pairwise comparison group, Lactobacillaceae and Lactobacillus exhibited the highest LDA values. Based on this, researchers hypothesized that these two taxa might play an important role in the mitigatory effects of CUR on OS and inflammatory responses [246].
Selenium, a critical micronutrient, exhibits important effects on antioxidant, anticancer, antiviral, and immune-modulatory activities [248]. The low poisonousness and highly bioavailable properties of SeNPs make them promising candidates for therapeutic applications and selenium supplements. REG3G is closely linked to the integrity and functionality of the gut barrier. A deficiency in REG3G can promote the proliferation of mucosa-adherent bacteria, ultimately contributing to gut barrier impairment [249]. A study by Qiao et al. (2022) [95] indicated that the jejunal villi were arranged disorderly, the VH was shortened, and the number of goblet cells was reduced in mice under DQ stimulation. However, the supplementation of biogenic SeNPs in the diet increased jejunal MUC2 and REG3G expression, demonstrating their efficacy in mitigating DQ-caused gut barrier damage. Additionally, a decrease in the F/B ratio and the levels of Desulfovibrio and an obvious increase in the levels of Bacteroides and Clostridium_XlVa were observed in mice fed with SeNPs. Bacteroides, an essential component of the gut microbiota, plays a vital role in carbohydrate metabolism and propionate production [250,251]. Recent advances in microbiome research revealed that Bacteroides thetaiotaomicron is crucial for absorbing nutrition and contributing to barrier integrity via promoting goblet cell maturation and regulating mucus secretion [252]. Clostridium_XlVa is found to possess the ability to break down carbohydrates and generate SCFAs, including acetate and butyrate [253,254]. Desulfovibrio, an opportunistic pathogen, generates the LPS endotoxin and exhibits higher levels in the gut of obese individuals and patients with ulcerative colitis compared to healthy subjects. Furthermore, elevated levels of Desulfovibrio can impair IECs and the intestinal barrier integrity [255,256]. As key metabolites of the intestinal microbiota, changes in SCFA levels were consistently found in the cecum of mice following SeNP supplements. In Qiao’s other article, metabolic pathway analysis using the KEGG database suggested that the predominant metabolic pathways affected by SeNP intervention were primarily associated with aldosterone synthesis and secretion, as well as glutathione metabolism [257]. Aldosterone plays a crucial regulatory role in the sodium balance and has been demonstrated to directly contribute to end-organ injury through various pathophysiological mechanisms [258]. Glutathione, a potent tripeptide antioxidant, plays a crucial role in xenobiotic metabolism and biotransformation processes. Its reactive sulfhydryl group, primarily located on the cysteine residue, enables the effective binding and neutralization of various toxic compounds, thereby conferring significant detoxification capacity [259]. The information above offered substantial evidence supporting the critical interplay between selenium supplementation, gut microbiome, and systemic metabolic regulation.
Periplaneta americana L., commonly known as the American cockroach, is a medically significant insect species belonging to the genus Periplaneta within the family Blattidae. This arthropod has been traditionally utilized in Chinese medicine and is recognized as a distinctive therapeutic agent. Recent pharmacological studies have demonstrated that Periplaneta americana extract (PAE) exhibits significant therapeutic potential in the management of various inflammatory disorders [260,261]. Lu et al. (2022) [262] elaborated that DQ damaged gut microbiota homeostasis, characterized by elevated levels of Bacteroidetes and Proteobacteria phyla, coupled with a marked reduction in Firmicutes populations. This microbial dysbiosis was associated with the development of intestinal barrier dysfunction (leaky gut), as evidenced by elevated levels of gut-derived bacterial metabolite DLA in systemic circulation. Notably, PAE intervention demonstrated protective effects by ameliorating DQ-induced reduction in α-diversity indices of gut microbiota. Furthermore, PAE treatment significantly modulated microbial composition, particularly through enhancing the proliferation of the beneficial bacterium Akkermansia muciniphila while suppressing the growth of Bacteroidetes in DQ-treated mice. Akkermansia muciniphila, an anaerobic bacterium, was first isolated two decades ago and has emerged as a significant member of the complex microbial community in the gut [263]. Moreover, A. muciniphila is identified as a promising next-generation probiotic [264,265]. A. muciniphila has been found to upregulate the levels of intestinal TJs including occludin and ZO-1 [266]. In addition, it was also reported that A. muciniphila can potentially offer numerous health benefits by elevating SCFAs generation [267].
Changes in gut microbiota and metabolites observed in experimental animal models after treatment with DQ or intervention are shown in Table 1.

6. Conclusions and Perspective

As previously mentioned, DQ is prevalent in the environment because of extensive contamination. This results in DQ being imperceptibly ingested by humans via contaminated crops and drinking water. Therefore, it is necessary to attenuate DQ-caused OS and elevate antioxidant ability. The intervention of antioxidative medicines can be effective in eliminating or inhibiting the production of ROS and RNS, which are known to play prominent roles in DQ poisoning. As the gut serves as the primary interface between the body and its luminal environment, it is particularly susceptible to oxidative injury compared to other organs. This heightened susceptibility is due to the substantial accumulation of diet-derived oxidants, carcinogens, and mutagens within the intestinal lumen [268,269]. Based on the above information, researchers have increasingly recognized that targeting the gut microbiota and its metabolites will pave the way for innovative strategies in the prevention and intervention of OS. This article highlighted the impact of DQ toxicity on the intestinal microbiota and may act as a valuable resource for both clinical practice and fundamental investigation. By exploring the effects of DQ exposure on gut health, it aims to deepen our comprehension of the intricate relationship between environmental toxins and intestinal well-being. Nevertheless, the results of the animal experiments mentioned above are not entirely consistent. We hypothesize that these differences may arise from interspecies discrepancies. For instance, some studies were conducted in mouse models, some were performed in piglets. This hypothesis is reasonable given the well-documented physiological, metabolic, and intestinal microbial discrepancies between mice and piglets. Furthermore, host genetics, individual variations, dietary habits, age, sex, comorbidities, and environmental factors may also affect the results [270,271]. It is worth mentioning that these results acquired from animal models may not directly translate to humans. Therefore, future studies conducted in humans or in preclinical animal models are needed to reveal complex and multifactorial mechanisms responsible for gut dysbiosis establishment in order to meet the demand for personalized therapies. There is still a wide space to explore the mechanism behind the toxicity caused by DQ exposure to gut microbiota and related metabolites. Multiple strategies have been investigated in clinical trials, ranging from therapies aimed at depleting the pathogens with antimicrobials to therapies aimed at remodeling “normal” commensals with probiotics or fecal transplantation.
Notably, beyond elucidating the mechanisms of intestinal injury induced by DQ, our research team has extended the investigative scope to explore the multi-organ crosstalk mediated by the gut–brain axis, gut–kidney axis, and gut–liver axis. By establishing a cross-organ toxicity assessment model, we revealed that DQ-induced gut barrier disruption not only triggers localized inflammatory responses but also exacerbates systemic pathophysiology through distinct yet interconnected pathways such as (1) neuroinflammatory cascades in the central nervous system via circulating microparticles (gut–brain axis), (2) aggravated renal OS due to dysregulated gut microbiota-derived metabolites (gut–kidney axis), and (3) impaired hepatic detoxification capacity mediated through the portal venous system (gut–liver axis). Looking ahead, integrating organ-on-a-chip platforms with multi-omics technologies will further unravel the centrality of the “gut-X axis” in environmental-exposure-related pathologies, thereby advancing toxicology into a systems biology era focused on holistic mechanistic decoding.

Author Contributions

J.H., Q.T., Y.-C.L., L.-J.W. and Y.-F.C. equally contributed to conceptualization, validation, writing—original draft preparation, writing—review and editing, visualization, and supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Tianjin Municipal Medical Key Construction Project, grant number TJYXZDXK-007A; and Beijing Union Medical Foundation—Rui E Emergency Medicine Research, grant number PUMF01010010-2024-18.

Data Availability Statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

Acknowledgments

Figures are created with Adobe Illustrator 2024.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

The following abbreviations are used in this manuscript:
BABile acid
CATCatalase
CURCurcumin
CDCrypt depth
CKDChronic kidney disease
DAODiamine oxidase
DQDiquat
DLAD-lactate
DEGsDifferentially expressed genes
EVsExtracellular vesicles
EUPSEucommia ulmoides polysaccharide
F/BFirmicutes/Bacteroidetes
GSH-PXGlutathione peroxidase
HCAHyocholic acid
HDCAHyodeoxycholic acid
HTHydroxytyrosol
HO-1Heme oxygenase-1
HtHydrogenobacter thermophilus strain
IECsIntestinal epithelial cells
ISIndoxyl sulfate
LABLactic acid bacteria
LAB-EVsLactic acid bacteria derived extracellular vesicles
LPSLipopolysaccharides
MDAMalondialdehyde
MUC2Mucin 2
Nrf2Nuclear factor erythroid 2-related factor 2
NLRP3NLR family pyrin domain containing 3
NF-κBNuclear factor kappa-B
NADPHNicotinamide adenine dinucleotide phosphate
OSOxidative stress
PAEPeriplaneta americana extract
PBAPrimary bile acid
PQParaquat
QGQuercetagetin
RNSReactive nitrogen species
RESResveratrol
ROSReactive oxygen species
SCFAsShort-chain fatty acids
SBASecondary bile acid
SODSuperoxide dismutase
SeNPsSelenium nanoparticles
TUDCATauroursodeoxycholic acid
TMAOTrimethylamine-N-oxide
TJsTight junctions
TDOTryptophan 2,3-dioxygenase
VHVillus height
ZO-1Zonula occludens-1

References

  1. Zhou, J.-N.; Lu, Y.-Q. Lethal Diquat Poisoning Manifests as Acute Central Nervous System Injury and Circulatory Failure: A Retrospective Cohort Study of 50 Cases. eClinicalMedicine 2022, 52, 101609. [Google Scholar] [CrossRef]
  2. Ren, Y.; Guo, F.; Wang, L. Imaging Findings and Toxicological Mechanisms of Nervous System Injury Caused by Diquat. Mol. Neurobiol. 2024, 61, 9272–9283. [Google Scholar] [CrossRef] [PubMed]
  3. Magalhães, N.; Carvalho, F.; Dinis-Oliveira, R. Human and Experimental Toxicology of Diquat Poisoning: Toxicokinetics, Mechanisms of Toxicity, Clinical Features, and Treatment. Hum. Exp. Toxicol. 2018, 37, 1131–1160. [Google Scholar] [CrossRef]
  4. Pateiro-Moure, M.; Martínez-Carballo, E.; Arias-Estévez, M.; Simal-Gándara, J. Determination of Quaternary Ammonium Herbicides in Soils. Comparison of Digestion, Shaking and Microwave-Assisted Extractions. J. Chromatogr. A 2008, 1196–1197, 110–116. [Google Scholar] [CrossRef]
  5. Pizzutti, I.R.; Vela, G.M.E.; de Kok, A.; Scholten, J.M.; Dias, J.V.; Cardoso, C.D.; Concenço, G.; Vivian, R. Determination of Paraquat and Diquat: LC-MS Method Optimization and Validation. Food Chem. 2016, 209, 248–255. [Google Scholar] [CrossRef] [PubMed]
  6. Chen, J.; Su, Y.; Lin, R.; Lin, F.; Shang, P.; Hussain, R.; Shi, D. Effects of Acute Diquat Poisoning on Liver Mitochondrial Apoptosis and Autophagy in Ducks. Front. Vet. Sci. 2021, 8, 727766. [Google Scholar] [CrossRef]
  7. Rose, M.S.; Crabtree, H.C.; Fletcher, K.; Wyatt, I. Biochemical Effects of Diquat and Paraquat. Disturbance of the Control of Corticosteroid Synthesis in Rat Adrenal and Subsequent Effects on the Control of Liver Glycogen Utilization. Biochem. J. 1974, 138, 437–443. [Google Scholar] [CrossRef]
  8. Li, W.; Wei, H.; Li, N.; Li, S.; Liu, Y.; Liu, R.; Zou, W.; Hu, P.; Zhang, Z.; Wang, C. Rapid Identification and Quantification of Diquat in Biological Fluids within 30 s Using a Portable Raman Spectrometer. Biosens. Bioelectron. 2023, 225, 115083. [Google Scholar] [CrossRef]
  9. Wageed, M.; Mahdy, H.M.; Kalaba, M.H.; Kelany, M.A.; Soliman, M. Development of LC-MS/MS Analytical Method for the Rapid Determination of Diquat in Water and Beverages. Food Chem. 2024, 438, 137869. [Google Scholar] [CrossRef] [PubMed]
  10. Zhang, H.; Sun, H.; Qian, J.; Sun, L.; Zong, C.; Zhang, J.; Yuan, B. High Expression of 3-Hydroxy-3-Methylglutaryl-CoA Synthase 2 (HMGCS2) Associated with Diquat-Induced Damage. Ecotoxicol. Environ. Saf. 2024, 281, 116623. [Google Scholar] [CrossRef]
  11. Koch, R.E.; Hill, G.E. An Assessment of Techniques to Manipulate Oxidative Stress in Animals. Funct. Ecol. 2017, 31, 9–21. [Google Scholar] [CrossRef]
  12. Li, X.; Zhu, J.; Lin, Q.; Yu, M.; Lu, J.; Feng, J.; Hu, C. Effects of Curcumin on Mitochondrial Function, Endoplasmic Reticulum Stress, and Mitochondria-Associated Endoplasmic Reticulum Membranes in the Jejunum of Oxidative Stress Piglets. J. Agric. Food Chem. 2022, 70, 8974–8985. [Google Scholar] [CrossRef]
  13. Yu, G.; Jian, T.; Cui, S.; Shi, L.; Kan, B.; Jian, X. Acute Diquat Poisoning Resulting in Toxic Encephalopathy: A Report of Three Cases. Clin. Toxicol. 2022, 60, 647–650. [Google Scholar] [CrossRef]
  14. Zhang, H.; Zhang, J.; Li, J.; Mao, Z.; Qian, J.; Zong, C.; Sun, H.; Yuan, B. Multi-Omics Analyses Reveal the Mechanisms of Early Stage Kidney Toxicity by Diquat. Toxics 2023, 11, 184. [Google Scholar] [CrossRef]
  15. Liu, G.; Xiao, L.; Cao, W.; Fang, T.; Jia, G.; Chen, X.; Zhao, H.; Wu, C.; Wang, J. Changes in the Metabolome of Rats after Exposure to Arginine and N-Carbamylglutamate in Combination with Diquat, a Compound That Causes Oxidative Stress, Assessed by 1H NMR Spectroscopy. Food Funct. 2016, 7, 964–974. [Google Scholar] [CrossRef]
  16. Sharma, T.; Natesh, N.S.; Pothuraju, R.; Batra, S.K.; Rachagani, S. Gut Microbiota: A Non-Target Victim of Pesticide-Induced Toxicity. Gut Microbes 2023, 15, 2187578. [Google Scholar] [CrossRef]
  17. Sosnowski, K.; Przybyłkowski, A. Ethanol-Induced Changes to the Gut Microbiome Compromise the Intestinal Homeostasis: A Review. Gut Microbes 2024, 16, 2393272. [Google Scholar] [CrossRef]
  18. Ma, L.; Ni, Y.; Wang, Z.; Tu, W.; Ni, L.; Zhuge, F.; Zheng, A.; Hu, L.; Zhao, Y.; Zheng, L.; et al. Spermidine Improves Gut Barrier Integrity and Gut Microbiota Function in Diet-Induced Obese Mice. Gut Microbes 2020, 12, 1832857. [Google Scholar] [CrossRef]
  19. Assimakopoulos, S.F.; Triantos, C.; Maroulis, I.; Gogos, C. The Role of the Gut Barrier Function in Health and Disease. Gastroenterol. Res. 2018, 11, 261–263. [Google Scholar] [CrossRef]
  20. Huo, J.; Wu, Z.; Sun, W.; Wang, Z.; Wu, J.; Huang, M.; Wang, B.; Sun, B. Protective Effects of Natural Polysaccharides on Intestinal Barrier Injury: A Review. J. Agric. Food Chem. 2022, 70, 711–735. [Google Scholar] [CrossRef]
  21. Song, D.; Cheng, Y.; Li, X.; Wang, F.; Lu, Z.; Xiao, X.; Wang, Y. Biogenic Nanoselenium Particles Effectively Attenuate Oxidative Stress-Induced Intestinal Epithelial Barrier Injury by Activating the Nrf2 Antioxidant Pathway. ACS Appl. Mater. Interfaces 2017, 9, 14724–14740. [Google Scholar] [CrossRef]
  22. Willott, E.; Balda, M.S.; Fanning, A.S.; Jameson, B.; Van Itallie, C.; Anderson, J.M. The Tight Junction Protein ZO-1 Is Homologous to the Drosophila Discs-Large Tumor Suppressor Protein of Septate Junctions. Proc. Natl. Acad. Sci. USA 1993, 90, 7834–7838. [Google Scholar] [CrossRef]
  23. Chassaing, B.; Etienne-Mesmin, L.; Gewirtz, A.T. Microbiota-Liver Axis in Hepatic Disease. Hepatology 2014, 59, 328–339. [Google Scholar] [CrossRef]
  24. Coutry, N.; Gasmi, I.; Herbert, F.; Jay, P. Mechanisms of Intestinal Dysbiosis: New Insights into Tuft Cell Functions. Gut Microbes 2024, 16, 2379624. [Google Scholar] [CrossRef]
  25. Bäckhed, F.; Ley, R.E.; Sonnenburg, J.L.; Peterson, D.A.; Gordon, J.I. Host-Bacterial Mutualism in the Human Intestine. Science 2005, 307, 1915–1920. [Google Scholar] [CrossRef]
  26. Marchesi, J.R.; Adams, D.H.; Fava, F.; Hermes, G.D.A.; Hirschfield, G.M.; Hold, G.; Quraishi, M.N.; Kinross, J.; Smidt, H.; Tuohy, K.M.; et al. The Gut Microbiota and Host Health: A New Clinical Frontier. Gut 2016, 65, 330–339. [Google Scholar] [CrossRef]
  27. Wong, C.C.; Yu, J. Gut Microbiota in Colorectal Cancer Development and Therapy. Nat. Rev. Clin. Oncol. 2023, 20, 429–452. [Google Scholar] [CrossRef]
  28. Mancin, L.; Wu, G.D.; Paoli, A. Gut Microbiota–Bile Acid–Skeletal Muscle Axis. Trends Microbiol. 2023, 31, 254–269. [Google Scholar] [CrossRef]
  29. Suzuki, T. Regulation of the Intestinal Barrier by Nutrients: The Role of Tight Junctions. Anim. Sci. J. 2020, 91, e13357. [Google Scholar] [CrossRef]
  30. Chen, L.; Zhang, S.; Wu, S.; Ren, Z.; Liu, G.; Wu, J. Synergistic Protective Effect of Konjac Mannan Oligosaccharides and Bacillus Subtilis on Intestinal Epithelial Barrier Dysfunction in Caco-2 Cell Model and Mice Model of Lipopolysaccharide Stimulation. Front. Immunol. 2021, 12, 696148. [Google Scholar] [CrossRef]
  31. Vergauwen, H.; Prims, S.; Degroote, J.; Wang, W.; Casteleyn, C.; van Cruchten, S.; de Smet, S.; Michiels, J.; van Ginneken, C. In Vitro Investigation of Six Antioxidants for Pig Diets. Antioxidants 2016, 5, 41. [Google Scholar] [CrossRef]
  32. Tian, J.; Jiang, Q.; Bao, X.; Yang, F.; Li, Y.; Sun, H.; Yao, K.; Yin, Y. Plant-Derived Squalene Supplementation Improves Growth Performance and Alleviates Acute Oxidative Stress-Induced Growth Retardation and Intestinal Damage in Piglets. Anim. Nutr. 2023, 15, 386–398. [Google Scholar] [CrossRef]
  33. Sule, R.O.; Condon, L.; Gomes, A.V. A Common Feature of Pesticides: Oxidative Stress—The Role of Oxidative Stress in Pesticide-Induced Toxicity. Oxidative Med. Cell. Longev. 2022, 2022, 5563759. [Google Scholar] [CrossRef]
  34. Hajhashemi, V.; Vaseghi, G.; Pourfarzam, M.; Abdollahi, A. Are Antioxidants Helpful for Disease Prevention? Res. Pharm. Sci. 2010, 5, 1–8. [Google Scholar]
  35. Li, W.; Liu, C.; Yang, Q.; Zhou, Y.; Liu, M.; Shan, H. Oxidative Stress and Antioxidant Imbalance in Ovulation Disorder in Patients with Polycystic Ovary Syndrome. Front. Nutr. 2022, 9, 1018674. [Google Scholar] [CrossRef]
  36. Bhattacharyya, A.; Chattopadhyay, R.; Mitra, S.; Crowe, S.E. Oxidative Stress: An Essential Factor in the Pathogenesis of Gastrointestinal Mucosal Diseases. Physiol. Rev. 2014, 94, 329–354. [Google Scholar] [CrossRef]
  37. Lightfoot, T.J.; Skibola, C.F.; Smith, A.G.; Forrest, M.S.; Adamson, P.J.; Morgan, G.J.; Bracci, P.M.; Roman, E.; Smith, M.T.; Holly, E.A. Polymorphisms in the Oxidative Stress Genes, Superoxide Dismutase, Glutathione Peroxidase and Catalase and Risk of Non-Hodgkin’s Lymphoma. Haematologica 2006, 91, 1222–1227. [Google Scholar]
  38. Halliwell, B. Biochemistry of Oxidative Stress. Biochem. Soc. Trans. 2007, 35 Pt 5, 1147–1150. [Google Scholar] [CrossRef]
  39. Wang, M.; Huang, H.; Liu, S.; Zhuang, Y.; Yang, H.; Li, Y.; Chen, S.; Wang, L.; Yin, L.; Yao, Y.; et al. Tannic Acid Modulates Intestinal Barrier Functions Associated with Intestinal Morphology, Antioxidative Activity, and Intestinal Tight Junction in a Diquat-Induced Mouse Model. RSC Adv. 2019, 9, 31988–31998. [Google Scholar] [CrossRef]
  40. Akbari, P.; Braber, S.; Varasteh, S.; Alizadeh, A.; Garssen, J.; Fink-Gremmels, J. The Intestinal Barrier as an Emerging Target in the Toxicological Assessment of Mycotoxins. Arch. Toxicol. 2017, 91, 1007–1029. [Google Scholar] [CrossRef]
  41. Zhuang, Y.; Wu, H.; Wang, X.; He, J.; He, S.; Yin, Y. Resveratrol Attenuates Oxidative Stress-Induced Intestinal Barrier Injury through PI3K/Akt-Mediated Nrf2 Signaling Pathway. Oxidative Med. Cell Longev. 2019, 2019, 7591840. [Google Scholar] [CrossRef] [PubMed]
  42. Li, Y.; Chen, W.; Yin, J.; Xia, S.; Jiang, Y.; Ge, Q.; Liu, J.; Wang, M.; Hou, Z.; Bai, Y.; et al. Biomineralized ZIF-8 Encapsulating SOD from Hydrogenobacter Thermophilus: Maintaining Activity in the Intestine and Alleviating Intestinal Oxidative Stress. Small 2024, 20, 2402812. [Google Scholar] [CrossRef] [PubMed]
  43. Ames, B.N. Dietary Carcinogens and Anticarcinogens. Oxygen Radicals and Degenerative Diseases. Science 1983, 221, 1256–1264. [Google Scholar] [CrossRef] [PubMed]
  44. Qiao, L.; Dou, X.; Yan, S.; Zhang, B.; Xu, C. Biogenic Selenium Nanoparticles Synthesized by Lactobacillus Casei ATCC 393 Alleviate Diquat-Induced Intestinal Barrier Dysfunction in C57BL/6 Mice through Their Antioxidant Activity. Food Funct. 2020, 11, 3020–3031. [Google Scholar] [CrossRef]
  45. Cao, S.; Wang, C.; Yan, J.; Li, X.; Wen, J.; Hu, C. Curcumin Ameliorates Oxidative Stress-Induced Intestinal Barrier Injury and Mitochondrial Damage by Promoting Parkin Dependent Mitophagy through AMPK-TFEB Signal Pathway. Free Radic. Biol. Med. 2020, 147, 8–22. [Google Scholar] [CrossRef]
  46. Xu, Q.; Liu, M.; Chao, X.; Zhang, C.; Yang, H.; Chen, J.; Zhao, C.; Zhou, B. Acidifiers Attenuate Diquat-Induced Oxidative Stress and Inflammatory Responses by Regulating NF-κB/MAPK/COX-2 Pathways in IPEC-J2 Cells. Antioxidants 2022, 11, 2002. [Google Scholar] [CrossRef]
  47. Guo, Y.; Liu, Y.; Zhao, S.; Xu, W.; Li, Y.; Zhao, P.; Wang, D.; Cheng, H.; Ke, Y.; Zhang, X. Oxidative Stress-Induced FABP5 S-Glutathionylation Protects against Acute Lung Injury by Suppressing Inflammation in Macrophages. Nat. Commun. 2021, 12, 7094. [Google Scholar] [CrossRef]
  48. Ghelli Luserna di Rora’, A.; Iacobucci, I.; Martinelli, G. The Cell Cycle Checkpoint Inhibitors in the Treatment of Leukemias. J. Hematol. Oncol. 2017, 10, 77. [Google Scholar] [CrossRef]
  49. Sies, H.; Stahl, W.; Sevanian, A. Nutritional, Dietary and Postprandial Oxidative Stress. J. Nutr. 2005, 135, 969–972. [Google Scholar] [CrossRef]
  50. Awad, W.A.; Aschenbach, J.R.; Setyabudi, F.M.C.S.; Razzazi-Fazeli, E.; Böhm, J.; Zentek, J. In Vitro Effects of Deoxynivalenol on Small Intestinal D-Glucose Uptake and Absorption of Deoxynivalenol across the Isolated Jejunal Epithelium of Laying Hens. Poult. Sci. 2007, 86, 15–20. [Google Scholar] [CrossRef]
  51. Zhao, G.-P.; Wang, X.-Y.; Li, J.-W.; Wang, R.; Ren, F.-Z.; Pang, G.-F.; Li, Y.-X. Imidacloprid Increases Intestinal Permeability by Disrupting Tight Junctions. Ecotoxicol. Env. Saf. 2021, 222, 112476. [Google Scholar] [CrossRef] [PubMed]
  52. Mittal, M.; Siddiqui, M.R.; Tran, K.; Reddy, S.P.; Malik, A.B. Reactive Oxygen Species in Inflammation and Tissue Injury. Antioxid. Redox Signal. 2014, 20, 1126–1167. [Google Scholar] [CrossRef] [PubMed]
  53. Guzik, T.J.; Korbut, R.; Adamek-Guzik, T. Nitric Oxide and Superoxide in Inflammation and Immune Regulation. J. Physiol. Pharmacol. 2003, 54, 469–487. [Google Scholar]
  54. Lorenzen, I.; Mullen, L.; Bekeschus, S.; Hanschmann, E.-M. Redox Regulation of Inflammatory Processes Is Enzymatically Controlled. Oxidative Med. Cell Longev. 2017, 2017, 8459402. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, C.; Cao, S.; Zhang, Q.; Shen, Z.; Feng, J.; Hong, Q.; Lu, J.; Xie, F.; Peng, Y.; Hu, C. Dietary Tributyrin Attenuates Intestinal Inflammation, Enhances Mitochondrial Function, and Induces Mitophagy in Piglets Challenged with Diquat. J. Agric. Food Chem. 2019, 67, 1409–1417. [Google Scholar] [CrossRef]
  56. Wang, P.; Song, C.-Y.; Lu, X.; Zhou, J.-N.; Lin, L.-Y.; Li, T.; Zhang, Q.; Lu, Y.-Q. Diquat Exacerbates Oxidative Stress and Neuroinflammation by Blocking the Autophagic Flux of Microglia in the Hippocampus. Ecotoxicol. Environ. Saf. 2024, 286, 117188. [Google Scholar] [CrossRef]
  57. Srivastava, S.K.; Yadav, U.C.S.; Reddy, A.B.M.; Saxena, A.; Tammali, R.; Shoeb, M.; Ansari, N.H.; Bhatnagar, A.; Petrash, M.J.; Srivastava, S.; et al. Aldose Reductase Inhibition Suppresses Oxidative Stress-Induced Inflammatory Disorders. Chem. Biol. Interact. 2011, 191, 330–338. [Google Scholar] [CrossRef]
  58. Wei, H.; Chen, G.; Wang, R.-J.; Peng, J. Oregano Essential Oil Decreased Susceptibility to Oxidative Stress-Induced Dysfunction of Intestinal Epithelial Barrier in Rats. J. Funct. Food. 2015, 18, 1191–1199. [Google Scholar] [CrossRef]
  59. Hou, Y.; Wang, L.; Ding, B.; Liu, Y.; Zhu, H.; Liu, J.; Li, Y.; Wu, X.; Yin, Y.; Wu, G. Dietary Alpha-Ketoglutarate Supplementation Ameliorates Intestinal Injury in Lipopolysaccharide-Challenged Piglets. Amino Acids 2010, 39, 555–564. [Google Scholar] [CrossRef]
  60. Xiao, L.; Cao, W.; Liu, G.; Fang, T.; Wu, X.; Jia, G.; Chen, X.; Zhao, H.; Wang, J.; Wu, C.; et al. Arginine, N-Carbamylglutamate, and Glutamine Exert Protective Effects against Oxidative Stress in Rat Intestine. Anim. Nutr. 2016, 2, 242–248. [Google Scholar] [CrossRef]
  61. Wang, Z.; Jiang, Q.; Li, P.; Shi, P.; Liu, C.; Wang, W.; Huang, K.; Yin, Y.; Huang, P. The Water Extract of Ampelopsis Grossedentata Alleviates Oxidative Stress and Intestinal Inflammation. Antioxidants 2023, 12, 547. [Google Scholar] [CrossRef] [PubMed]
  62. Han, H.; Liu, Z.; Yin, J.; Gao, J.; He, L.; Wang, C.; Hou, R.; He, X.; Wang, G.; Li, T.; et al. D-Galactose Induces Chronic Oxidative Stress and Alters Gut Microbiota in Weaned Piglets. Front. Physiol. 2021, 12, 634283. [Google Scholar] [CrossRef] [PubMed]
  63. Hao, L.; Cheng, Y.; Su, W.; Wang, C.; Lu, Z.; Jin, M.; Wang, F.; Wang, Y. Pediococcus Pentosaceus ZJUAF-4 Relieves Oxidative Stress and Restores the Gut Microbiota in Diquat-Induced Intestinal Injury. Appl. Microbiol. Biotechnol. 2021, 105, 1657–1668. [Google Scholar] [CrossRef] [PubMed]
  64. Sun, L.; Jia, H.; Li, J.; Yu, M.; Yang, Y.; Tian, D.; Zhang, H.; Zou, Z. Cecal Gut Microbiota and Metabolites Might Contribute to the Severity of Acute Myocardial Ischemia by Impacting the Intestinal Permeability, Oxidative Stress, and Energy Metabolism. Front. Microbiol. 2019, 10, 1745. [Google Scholar] [CrossRef]
  65. Houde, V.; Grenier, D.; Chandad, F. Protective Effects of Grape Seed Proanthocyanidins against Oxidative Stress Induced by Lipopolysaccharides of Periodontopathogens. J. Periodontol. 2006, 77, 1371–1379. [Google Scholar] [CrossRef]
  66. Spiljar, M.; Merkler, D.; Trajkovski, M. The Immune System Bridges the Gut Microbiota with Systemic Energy Homeostasis: Focus on TLRs, Mucosal Barrier, and SCFAs. Front. Immunol. 2017, 8, 1353. [Google Scholar] [CrossRef]
  67. Lee, H.H.; Molla, M.N.; Cantor, C.R.; Collins, J.J. Bacterial Charity Work Leads to Population-Wide Resistance. Nature 2010, 467, 82–85. [Google Scholar] [CrossRef]
  68. Han, M.; Song, P.; Huang, C.; Rezaei, A.; Farrar, S.; Brown, M.A.; Ma, X. Dietary Grape Seed Proanthocyanidins (GSPs) Improve Weaned Intestinal Microbiota and Mucosal Barrier Using a Piglet Model. Oncotarget 2016, 7, 80313–80326. [Google Scholar] [CrossRef]
  69. Sun, X.; Jiao, X.; Ma, Y.; Liu, Y.; Zhang, L.; He, Y.; Chen, Y. Trimethylamine N-Oxide Induces Inflammation and Endothelial Dysfunction in Human Umbilical Vein Endothelial Cells via Activating ROS-TXNIP-NLRP3 Inflammasome. Biochem. Biophys. Res. Commun. 2016, 481, 63–70. [Google Scholar] [CrossRef]
  70. Vašíček, O.; Lojek, A.; Číž, M. Serotonin and Its Metabolites Reduce Oxidative Stress in Murine RAW264.7 Macrophages and Prevent Inflammation. J. Physiol. Biochem. 2020, 76, 49–60. [Google Scholar] [CrossRef]
  71. Mishra, V.; Shah, C.; Mokashe, N.; Chavan, R.; Yadav, H.; Prajapati, J. Probiotics as Potential Antioxidants: A Systematic Review. J. Agric. Food Chem. 2015, 63, 3615–3626. [Google Scholar] [CrossRef]
  72. Park, A.; Koh, H.C. NF-κB/mTOR-Mediated Autophagy Can Regulate Diquat-Induced Apoptosis. Arch. Toxicol. 2019, 93, 1239–1253. [Google Scholar] [CrossRef]
  73. Wen, C.; Li, F.; Guo, Q.; Zhang, L.; Duan, Y.; Wang, W.; Li, J.; He, S.; Chen, W.; Yin, Y. Protective Effects of Taurine against Muscle Damage Induced by Diquat in 35 Days Weaned Piglets. J. Anim. Sci. Biotechnol. 2020, 11, 56. [Google Scholar] [CrossRef]
  74. Osburn, W.O.; Wakabayashi, N.; Misra, V.; Nilles, T.; Biswal, S.; Trush, M.A.; Kensler, T.W. Nrf2 Regulates an Adaptive Response Protecting against Oxidative Damage Following Diquat-Mediated Formation of Superoxide Anion. Arch. Biochem. Biophys. 2006, 454, 7–15. [Google Scholar] [CrossRef]
  75. Zha, A.; Yan, J.; Li, J.; Wang, J.; Qi, M.; Liao, P.; Chun, G.; Yin, Y. Melatonin Increased Antioxidant Capacity to Ameliorate Growth Retardation and Intestinal Epithelial Barrier Dysfunction in Diquat-Challenged Piglets. J. Sci. Food Agric. 2024, 104, 2262–2271. [Google Scholar] [CrossRef]
  76. Abdollahi, M.; Ranjbar, A.; Shadnia, S.; Nikfar, S.; Rezaie, A. Pesticides and Oxidative Stress: A Review. Med. Sci. Monit. 2004, 10, RA141–RA147. [Google Scholar]
  77. Xie, L.-W.; Cai, S.; Zhao, T.-S.; Li, M.; Tian, Y. Green Tea Derivative (-)-Epigallocatechin-3-Gallate (EGCG) Confers Protection against Ionizing Radiation-Induced Intestinal Epithelial Cell Death Both in Vitro and in Vivo. Free Radic. Biol. Med. 2020, 161, 175–186. [Google Scholar] [CrossRef]
  78. Hu, Q.; Ren, J.; Li, G.; Wu, J.; Wu, X.; Wang, G.; Gu, G.; Ren, H.; Hong, Z.; Li, J. The Mitochondrially Targeted Antioxidant MitoQ Protects the Intestinal Barrier by Ameliorating Mitochondrial DNA Damage via the Nrf2/ARE Signaling Pathway. Cell Death Dis. 2018, 9, 403. [Google Scholar] [CrossRef]
  79. Dong, Y.; Zhao, J.; Zhu, Q.; Liu, H.; Wang, J.; Lu, W. Melatonin Inhibits the Apoptosis of Rooster Leydig Cells by Suppressing Oxidative Stress via AKT-Nrf2 Pathway Activation. Free Radic. Biol. Med. 2020, 160, 1–12. [Google Scholar] [CrossRef]
  80. Zorov, D.B.; Juhaszova, M.; Sollott, S.J. Mitochondrial Reactive Oxygen Species (ROS) and ROS-Induced ROS Release. Physiol. Rev. 2014, 94, 909–950. [Google Scholar] [CrossRef]
  81. Yuan, J.; Meng, H.; Liu, Y.; Wang, L.; Zhu, Q.; Wang, Z.; Liu, H.; Zhang, K.; Zhao, J.; Li, W.; et al. Bacillus Amyloliquefaciens Attenuates the Intestinal Permeability, Oxidative Stress and Endoplasmic Reticulum Stress: Transcriptome and Microbiome Analyses in Weaned Piglets. Front. Microbiol. 2024, 15, 1362487. [Google Scholar] [CrossRef]
  82. Cao, S.; Wu, H.; Wang, C.; Zhang, Q.; Jiao, L.; Lin, F.; Hu, C.H. Diquat-Induced Oxidative Stress Increases Intestinal Permeability, Impairs Mitochondrial Function, and Triggers Mitophagy in Piglets. J. Anim. Sci. 2018, 96, 1795–1805. [Google Scholar] [CrossRef]
  83. Yin, J.; Liu, M.; Ren, W.; Duan, J.; Yang, G.; Zhao, Y.; Fang, R.; Chen, L.; Li, T.; Yin, Y. Effects of Dietary Supplementation with Glutamate and Aspartate on Diquat-Induced Oxidative Stress in Piglets. PLoS ONE 2015, 10, e0122893. [Google Scholar] [CrossRef]
  84. Lv, M.; Yu, B.; Mao, X.B.; Zheng, P.; He, J.; Chen, D.W. Responses of Growth Performance and Tryptophan Metabolism to Oxidative Stress Induced by Diquat in Weaned Pigs. Animal 2012, 6, 928–934. [Google Scholar] [CrossRef]
  85. Zheng, P.; Yu, B.; Lv, M.; Chen, D. Effects of Oxidative Stress Induced by Diquat on Arginine Metabolism of Postweaning Pigs. Asian-Australasions J. Anim. Sci. 2010, 23, 98–105. [Google Scholar] [CrossRef]
  86. Bafana, A.; Dutt, S.; Kumar, S.; Ahuja, P.S. Superoxide Dismutase: An Industrial Perspective. Crit. Rev. Biotechnol. 2011, 31, 65–76. [Google Scholar] [CrossRef]
  87. Hwang, J.; Jin, J.; Jeon, S.; Moon, S.H.; Park, M.Y.; Yum, D.-Y.; Kim, J.H.; Kang, J.-E.; Park, M.H.; Kim, E.-J.; et al. SOD1 Suppresses Pro-Inflammatory Immune Responses by Protecting against Oxidative Stress in Colitis. Redox Biol. 2020, 37, 101760. [Google Scholar] [CrossRef]
  88. Liu, P.; Luo, G.; Dodson, M.; Schmidlin, C.J.; Wei, Y.; Kerimoglu, B.; Ooi, A.; Chapman, E.; Garcia, J.G.; Zhang, D.D. The NRF2-LOC344887 Signaling Axis Suppresses Pulmonary Fibrosis. Redox Biol. 2021, 38, 101766. [Google Scholar] [CrossRef]
  89. Baird, L.; Yamamoto, M. The Molecular Mechanisms Regulating the KEAP1-NRF2 Pathway. Mol. Cell Biol. 2020, 40, e00099-20. [Google Scholar] [CrossRef]
  90. Jia, H.; Zhang, Y.; Si, X.; Jin, Y.; Jiang, D.; Dai, Z.; Wu, Z. Quercetin Alleviates Oxidative Damage by Activating Nuclear Factor Erythroid 2-Related Factor 2 Signaling in Porcine Enterocytes. Nutrients 2021, 13, 375. [Google Scholar] [CrossRef]
  91. Robles-Almazan, M.; Pulido-Moran, M.; Moreno-Fernandez, J.; Ramirez-Tortosa, C.; Rodriguez-Garcia, C.; Quiles, J.L.; Ramirez-Tortosa, M. Hydroxytyrosol: Bioavailability, Toxicity, and Clinical Applications. Food Res. Int. 2018, 105, 654–667. [Google Scholar] [CrossRef]
  92. Wen, X.; Tang, S.; Wan, F.; Zhong, R.; Chen, L.; Zhang, H. The PI3K/Akt-Nrf2 Signaling Pathway and Mitophagy Synergistically Mediate Hydroxytyrosol to Alleviate Intestinal Oxidative Damage. Int. J. Biol. Sci. 2024, 20, 4258–4276. [Google Scholar] [CrossRef]
  93. Xu, C.; Qiao, L.; Guo, Y.; Ma, L.; Cheng, Y. Preparation, Characteristics and Antioxidant Activity of Polysaccharides and Proteins-Capped Selenium Nanoparticles Synthesized by Lactobacillus Casei ATCC 393. Carbohydr. Polym. 2018, 195, 576–585. [Google Scholar] [CrossRef]
  94. Liu, Q.; Zhang, D.; Hu, D.; Zhou, X.; Zhou, Y. The Role of Mitochondria in NLRP3 Inflammasome Activation. Mol. Immunol. 2018, 103, 115–124. [Google Scholar] [CrossRef]
  95. Qiao, L.; Zhang, X.; Pi, S.; Chang, J.; Dou, X.; Yan, S.; Song, X.; Chen, Y.; Zeng, X.; Zhu, L.; et al. Dietary Supplementation with Biogenic Selenium Nanoparticles Alleviate Oxidative Stress-Induced Intestinal Barrier Dysfunction. npj Sci. Food 2022, 6, 30. [Google Scholar] [CrossRef] [PubMed]
  96. Yuan, D.; Hussain, T.; Tan, B.; Liu, Y.; Ji, P.; Yin, Y. The Evaluation of Antioxidant and Anti-Inflammatory Effects of Eucommia Ulmoides Flavones Using Diquat-Challenged Piglet Models. Oxidative Med. Cell Longev. 2017, 2017, 8140962. [Google Scholar] [CrossRef]
  97. Ma, T.Y.; Boivin, M.A.; Ye, D.; Pedram, A.; Said, H.M. Mechanism of TNF-α Modulation of Caco-2 Intestinal Epithelial Tight Junction Barrier: Role of Myosin Light-Chain Kinase Protein Expression. Am. J. Physiol. Gastrointest. Liver Physiol. 2005, 288, G422–G430. [Google Scholar] [CrossRef]
  98. Al-Sadi, R.M.; Ma, T.Y. IL-1beta Causes an Increase in Intestinal Epithelial Tight Junction Permeability. J. Immunol. 2007, 178, 4641–4649. [Google Scholar] [CrossRef]
  99. Chen, K.; Tang, Y.; Lan, L.; Li, M.; Lu, Z. Autophagy Mediated FTH1 Degradation Activates Gasdermin E Dependent Pyroptosis Contributing to Diquat Induced Kidney Injury. Food Chem. Toxicol. 2024, 184, 114411. [Google Scholar] [CrossRef]
  100. Wen, X.; Tang, L.; Zhong, R.; Liu, L.; Chen, L.; Zhang, H. Role of Mitophagy in Regulating Intestinal Oxidative Damage. Antioxidants 2023, 12, 480. [Google Scholar] [CrossRef]
  101. Figueira, T.R.; Barros, M.H.; Camargo, A.A.; Castilho, R.F.; Ferreira, J.C.B.; Kowaltowski, A.J.; Sluse, F.E.; Souza-Pinto, N.C.; Vercesi, A.E. Mitochondria as a Source of Reactive Oxygen and Nitrogen Species: From Molecular Mechanisms to Human Health. Antioxid. Redox Signal. 2013, 18, 2029–2074. [Google Scholar] [CrossRef]
  102. Zorov, D.B.; Juhaszova, M.; Sollott, S.J. Mitochondrial ROS-Induced ROS Release: An Update and Review. Biochim. Biophys. Acta 2006, 1757, 509–517. [Google Scholar] [CrossRef]
  103. Sweetman, E.; Kleffmann, T.; Edgar, C.; de Lange, M.; Vallings, R.; Tate, W. A SWATH-MS Analysis of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome Peripheral Blood Mononuclear Cell Proteomes Reveals Mitochondrial Dysfunction. J. Transl. Med. 2020, 18, 365. [Google Scholar] [CrossRef]
  104. De Gaetano, A.; Gibellini, L.; Zanini, G.; Nasi, M.; Cossarizza, A.; Pinti, M. Mitophagy and Oxidative Stress: The Role of Aging. Antioxidants 2021, 10, 794. [Google Scholar] [CrossRef]
  105. Balaban, R.S.; Nemoto, S.; Finkel, T. Mitochondria, Oxidants, and Aging. Cell 2005, 120, 483–495. [Google Scholar] [CrossRef]
  106. Chen, Y.; Zhang, H.; Ji, S.; Jia, P.; Chen, Y.; Li, Y.; Wang, T. Resveratrol and Its Derivative Pterostilbene Attenuate Oxidative Stress-Induced Intestinal Injury by Improving Mitochondrial Redox Homeostasis and Function via SIRT1 Signaling. Free Radic. Biol. Med. 2021, 177, 1–14. [Google Scholar] [CrossRef]
  107. Yardeni, T.; Tanes, C.E.; Bittinger, K.; Mattei, L.M.; Schaefer, P.M.; Singh, L.N.; Wu, G.D.; Murdock, D.G.; Wallace, D.C. Host Mitochondria Influence Gut Microbiome Diversity: A Role for ROS. Sci. Signal. 2019, 12, eaaw3159. [Google Scholar] [CrossRef]
  108. Ballard, J.W.O.; Towarnicki, S.G. Mitochondria, the Gut Microbiome and ROS. Cell. Signal. 2020, 75, 109737. [Google Scholar] [CrossRef]
  109. Zhou, Y.; Wang, Q.; Mark Evers, B.; Chung, D.H. Oxidative Stress-Induced Intestinal Epithelial Cell Apoptosis Is Mediated by P38 MAPK. Biochem. Biophys. Res. Commun. 2006, 350, 860–865. [Google Scholar] [CrossRef] [PubMed]
  110. Xu, X.; Wei, Y.; Hua, H.; Jing, X.; Zhu, H.; Xiao, K.; Zhao, J.; Liu, Y. Polyphenols Sourced from Ilex Latifolia Thunb. Relieve Intestinal Injury via Modulating Ferroptosis in Weanling Piglets under Oxidative Stress. Antioxidants 2022, 11, 966. [Google Scholar] [CrossRef] [PubMed]
  111. Alan, P.; Vandevoorde, K.R.; Joshi, B.; Cardoen, B.; Gao, G.; Mohammadzadeh, Y.; Hamarneh, G.; Nabi, I.R. Basal Gp78-Dependent Mitophagy Promotes Mitochondrial Health and Limits Mitochondrial ROS. Cell. Mol. Life Sci. 2022, 79, 565. [Google Scholar] [CrossRef]
  112. Saita, S.; Shirane, M.; Nakayama, K.I. Selective Escape of Proteins from the Mitochondria during Mitophagy. Nat. Commun. 2013, 4, 1410. [Google Scholar] [CrossRef]
  113. Kim, I.; Lemasters, J.J. Mitophagy Selectively Degrades Individual Damaged Mitochondria after Photoirradiation. Antioxid. Redox Signal. 2011, 14, 1919–1928. [Google Scholar] [CrossRef]
  114. Zhang, C.; Nie, P.; Zhou, C.; Hu, Y.; Duan, S.; Gu, M.; Jiang, D.; Wang, Y.; Deng, Z.; Chen, J.; et al. Oxidative Stress-Induced Mitophagy Is Suppressed by the miR-106b-93-25 Cluster in a Protective Manner. Cell Death Dis. 2021, 12, 209. [Google Scholar] [CrossRef]
  115. Wang, Y.; Nartiss, Y.; Steipe, B.; McQuibban, G.A.; Kim, P.K. ROS-Induced Mitochondrial Depolarization Initiates PARK2/PARKIN-Dependent Mitochondrial Degradation by Autophagy. Autophagy 2012, 8, 1462–1476. [Google Scholar] [CrossRef]
  116. Chen, T.; Kim, C.Y.; Kaur, A.; Lamothe, L.; Shaikh, M.; Keshavarzian, A.; Hamaker, B.R. Dietary Fibre-Based SCFA Mixtures Promote Both Protection and Repair of Intestinal Epithelial Barrier Function in a Caco-2 Cell Model. Food Funct. 2017, 8, 1166–1173. [Google Scholar] [CrossRef]
  117. Martin-Gallausiaux, C.; Marinelli, L.; Blottière, H.M.; Larraufie, P.; Lapaque, N. SCFA: Mechanisms and Functional Importance in the Gut. Proc. Nutr. Soc. 2021, 80, 37–49. [Google Scholar] [CrossRef]
  118. Yang, J.; Pei, G.; Sun, X.; Xiao, Y.; Miao, C.; Zhou, L.; Wang, B.; Yang, L.; Yu, M.; Zhang, Z.-S.; et al. RhoB Affects Colitis through Modulating Cell Signaling and Intestinal Microbiome. Microbiome 2022, 10, 149. [Google Scholar] [CrossRef]
  119. Hays, K.E.; Pfaffinger, J.M.; Ryznar, R. The Interplay between Gut Microbiota, Short-Chain Fatty Acids, and Implications for Host Health and Disease. Gut Microbes 2024, 16, 2393270. [Google Scholar] [CrossRef]
  120. Gonçalves, P.; Gregório, I.; Catarino, T.A.; Martel, F. The Effect of Oxidative Stress upon the Intestinal Epithelial Uptake of Butyrate. Eur. J. Pharmacol. 2013, 699, 88–100. [Google Scholar] [CrossRef] [PubMed]
  121. Lin, Y.; Fang, Z.; Che, L.; Xu, S.; Wu, D.; Wu, C.; Wu, X. Use of Sodium Butyrate as an Alternative to Dietary Fiber: Effects on the Embryonic Development and Anti-Oxidative Capacity of Rats. PLoS ONE 2014, 9, e97838. [Google Scholar] [CrossRef]
  122. Bai, Y.; Mansell, T.J. Production and Sensing of Butyrate in a Probiotic Escherichia coli Strain. Int. J. Mol. Sci. 2020, 21, 3615. [Google Scholar] [CrossRef]
  123. Guo, W.; Liu, J.; Sun, J.; Gong, Q.; Ma, H.; Kan, X.; Cao, Y.; Wang, J.; Fu, S. Butyrate Alleviates Oxidative Stress by Regulating NRF2 Nuclear Accumulation and H3K9/14 Acetylation via GPR109A in Bovine Mammary Epithelial Cells and Mammary Glands. Free Radic. Biol. Med. 2020, 152, 728–742. [Google Scholar] [CrossRef] [PubMed]
  124. Sun, B.; Jia, Y.; Yang, S.; Zhao, N.; Hu, Y.; Hong, J.; Gao, S.; Zhao, R. Sodium Butyrate Protects against High-Fat Diet-Induced Oxidative Stress in Rat Liver by Promoting Expression of Nuclear Factor E2-Related Factor 2. Br. J. Nutr. 2019, 122, 400–410. [Google Scholar] [CrossRef]
  125. Dicks, L.M.T. How Important Are Fatty Acids in Human Health and Can They Be Used in Treating Diseases? Gut Microbes 2024, 16, 2420765. [Google Scholar] [CrossRef]
  126. Nielsen, D.S.G.; Jensen, B.B.; Theil, P.K.; Nielsen, T.S.; Knudsen, K.E.B.; Purup, S. Effect of Butyrate and Fermentation Products on Epithelial Integrity in a Mucus-Secreting Human Colon Cell Line. J. Funct. Food. 2018, 40, 9–17. [Google Scholar] [CrossRef]
  127. Hamer, H.M.; Jonkers, D.; Venema, K.; Vanhoutvin, S.; Troost, F.J.; Brummer, R.-J. Review Article: The Role of Butyrate on Colonic Function. Aliment. Pharmacol. Ther. 2008, 27, 104–119. [Google Scholar] [CrossRef]
  128. Galvez, J.; Rodríguez-Cabezas, M.E.; Zarzuelo, A. Effects of Dietary Fiber on Inflammatory Bowel Disease. Mol. Nutr. Food Res. 2005, 49, 601–608. [Google Scholar] [CrossRef]
  129. Donohoe, D.R.; Garge, N.; Zhang, X.; Sun, W.; O’Connell, T.M.; Bunger, M.K.; Bultman, S.J. The Microbiome and Butyrate Regulate Energy Metabolism and Autophagy in the Mammalian Colon. Cell Metab. 2011, 13, 517–526. [Google Scholar] [CrossRef] [PubMed]
  130. Lee, J.S.; Lee, G.M. Effect of Sodium Butyrate on Autophagy and Apoptosis in Chinese Hamster Ovary Cells. Biotechnol. Prog. 2012, 28, 349–357. [Google Scholar] [CrossRef] [PubMed]
  131. Rose, S.; Bennuri, S.C.; Davis, J.E.; Wynne, R.; Slattery, J.C.; Tippett, M.; Delhey, L.; Melnyk, S.; Kahler, S.G.; MacFabe, D.F.; et al. Butyrate Enhances Mitochondrial Function during Oxidative Stress in Cell Lines from Boys with Autism. Transl. Psychiatry 2018, 8, 42. [Google Scholar] [CrossRef] [PubMed]
  132. Groschwitz, K.R.; Hogan, S.P. Intestinal Barrier Function: Molecular Regulation and Disease Pathogenesis. J. Allergy Clin. Immunol. 2009, 124, 3–20. [Google Scholar] [CrossRef]
  133. Gou, F.; Lin, Q.; Tu, X.; Zhu, J.; Li, X.; Chen, S.; Hu, C. Hesperidin Alleviated Intestinal Barrier Injury, Mitochondrial Dysfunction, and Disorder of Endoplasmic Reticulum Mitochondria Contact Sites under Oxidative Stress. J. Agric. Food Chem. 2024, 72, 16276–16286. [Google Scholar] [CrossRef]
  134. Wijtten, P.J.A.; van der Meulen, J.; Verstegen, M.W.A. Intestinal Barrier Function and Absorption in Pigs after Weaning: A Review. Br. J. Nutr. 2011, 105, 967–981. [Google Scholar] [CrossRef]
  135. Tang, X.; Ma, W.; Zhan, W.; Wang, X.; Dong, H.; Zhao, H.; Yang, L.; Ji, C.; Han, Q.; Ji, C.; et al. Internal Biliary Drainage Superior to External Biliary Drainage in Improving Gut Mucosa Barrier Because of Goblet Cells and Mucin-2 up-Regulation. Biosci. Rep. 2018, 38, BSR20171241. [Google Scholar] [CrossRef]
  136. Vancamelbeke, M.; Vermeire, S. The Intestinal Barrier: A Fundamental Role in Health and Disease. Expert. Rev. Gastroenterol. Hepatol. 2017, 11, 821–834. [Google Scholar] [CrossRef]
  137. Wen, X.; Wan, F.; Zhong, R.; Chen, L.; Zhang, H. Hydroxytyrosol Alleviates Intestinal Oxidative Stress by Regulating Bile Acid Metabolism in a Piglet Model. Int. J. Mol. Sci. 2024, 25, 5590. [Google Scholar] [CrossRef]
  138. Johansson, M.E.V.; Phillipson, M.; Petersson, J.; Velcich, A.; Holm, L.; Hansson, G.C. The Inner of the Two Muc2 Mucin-Dependent Mucus Layers in Colon Is Devoid of Bacteria. Proc. Natl. Acad. Sci. USA 2008, 105, 15064–15069. [Google Scholar] [CrossRef]
  139. Van Der Sluis, M.; De Koning, B.A.E.; De Bruijn, A.C.J.M.; Velcich, A.; Meijerink, J.P.P.; Van Goudoever, J.B.; Büller, H.A.; Dekker, J.; Van Seuningen, I.; Renes, I.B.; et al. Muc2-Deficient Mice Spontaneously Develop Colitis, Indicating That MUC2 Is Critical for Colonic Protection. Gastroenterology 2006, 131, 117–129. [Google Scholar] [CrossRef]
  140. Rao, R. Oxidative Stress-Induced Disruption of Epithelial and Endothelial Tight Junctions. Front. Biosci. 2008, 13, 7210–7226. [Google Scholar] [CrossRef]
  141. Qin, Q.; Xu, X.; Wang, X.; Wu, H.; Zhu, H.; Hou, Y.; Dai, B.; Liu, X.; Liu, Y. Glutamate Alleviates Intestinal Injury, Maintains mTOR and Suppresses TLR4 and NOD Signaling Pathways in Weanling Pigs Challenged with Lipopolysaccharide. Sci. Rep. 2018, 8, 15124. [Google Scholar] [CrossRef]
  142. Chen, J.; Yu, B.; Chen, D.; Huang, Z.; Mao, X.; Zheng, P.; Yu, J.; Luo, J.; He, J. Chlorogenic Acid Improves Intestinal Barrier Functions by Suppressing Mucosa Inflammation and Improving Antioxidant Capacity in Weaned Pigs. J. Nutr. Biochem. 2018, 59, 84–92. [Google Scholar] [CrossRef] [PubMed]
  143. Chen, Y.; Miao, L.; Yao, Y.; Wu, W.; Wu, X.; Gong, C.; Qiu, L.; Chen, J. Dexmedetomidine Ameliorate CLP-Induced Rat Intestinal Injury via Inhibition of Inflammation. Mediat. Inflamm. 2015, 2015, 918361. [Google Scholar] [CrossRef] [PubMed]
  144. Yi, H.; Zhang, L.; Gan, Z.; Xiong, H.; Yu, C.; Du, H.; Wang, Y. High Therapeutic Efficacy of Cathelicidin-WA against Postweaning Diarrhea via Inhibiting Inflammation and Enhancing Epithelial Barrier in the Intestine. Sci. Rep. 2016, 6, 25679. [Google Scholar] [CrossRef]
  145. Chen, J.; Chen, D.; Yu, B.; Luo, Y.; Zheng, P.; Mao, X.; Yu, J.; Luo, J.; Huang, Z.; Yan, H.; et al. Chlorogenic Acid Attenuates Oxidative Stress-Induced Intestinal Mucosa Disruption in Weaned Pigs. Front. Vet. Sci. 2022, 9, 806253. [Google Scholar] [CrossRef]
  146. Giannenas, I.; Bonos, E.; Anestis, V.; Filioussis, G.; Papanastasiou, D.K.; Bartzanas, T.; Papaioannou, N.; Tzora, A.; Skoufos, I. Effects of Protease Addition and Replacement of Soybean Meal by Corn Gluten Meal on the Growth of Broilers and on the Environmental Performances of a Broiler Production System in Greece. PLoS ONE 2017, 12, e0169511. [Google Scholar] [CrossRef]
  147. Elkouby-Naor, L.; Ben-Yosef, T. Functions of Claudin Tight Junction Proteins and Their Complex Interactions in Various Physiological Systems. Int. Rev. Cell Mol. Biol. 2010, 279, 1–32. [Google Scholar] [CrossRef]
  148. Apostolova, D.; Apostolov, G.; Moten, D.; Batsalova, T.; Dzhambazov, B. Claudin-12: Guardian of the Tissue Barrier or Friend of Tumor Cells. Tissue Barriers 2024, 12, 2387408. [Google Scholar] [CrossRef]
  149. Zheng, P.; Yu, B.; He, J.; Yu, J.; Mao, X.; Luo, Y.; Luo, J.; Huang, Z.; Tian, G.; Zeng, Q.; et al. Arginine Metabolism and Its Protective Effects on Intestinal Health and Functions in Weaned Piglets under Oxidative Stress Induced by Diquat. Br. J. Nutr. 2017, 117, 1495–1502. [Google Scholar] [CrossRef] [PubMed]
  150. Jiao, N.; Wu, Z.; Ji, Y.; Wang, B.; Dai, Z.; Wu, G. L-Glutamate Enhances Barrier and Antioxidative Functions in Intestinal Porcine Epithelial Cells. J. Nutr. 2015, 145, 2258–2264. [Google Scholar] [CrossRef]
  151. Liu, F.; Cottrell, J.J.; Furness, J.B.; Rivera, L.R.; Kelly, F.W.; Wijesiriwardana, U.; Pustovit, R.V.; Fothergill, L.J.; Bravo, D.M.; Celi, P.; et al. Selenium and Vitamin E Together Improve Intestinal Epithelial Barrier Function and Alleviate Oxidative Stress in Heat-Stressed Pigs. Exp. Physiol. 2016, 101, 801–810. [Google Scholar] [CrossRef] [PubMed]
  152. Wen, C.; Guo, Q.; Wang, W.; Duan, Y.; Zhang, L.; Li, J.; He, S.; Chen, W.; Li, F. Taurine Alleviates Intestinal Injury by Mediating Tight Junction Barriers in Diquat-Challenged Piglet Models. Front. Physiol. 2020, 11, 449. [Google Scholar] [CrossRef] [PubMed]
  153. Williams, J.M.; Duckworth, C.A.; Watson, A.J.M.; Frey, M.R.; Miguel, J.C.; Burkitt, M.D.; Sutton, R.; Hughes, K.R.; Hall, L.J.; Caamaño, J.H.; et al. A Mouse Model of Pathological Small Intestinal Epithelial Cell Apoptosis and Shedding Induced by Systemic Administration of Lipopolysaccharide. Dis. Model. Mech. 2013, 6, 1388–1399. [Google Scholar] [CrossRef] [PubMed]
  154. Turner, J.R. Intestinal Mucosal Barrier Function in Health and Disease. Nat. Rev. Immunol. 2009, 9, 799–809. [Google Scholar] [CrossRef]
  155. Song, D.; Zong, X.; Zhang, H.; Wang, T.; Yi, H.; Luan, C.; Wang, Y. Antimicrobial Peptide Cathelicidin-BF Prevents Intestinal Barrier Dysfunction in a Mouse Model of Endotoxemia. Int. Immunopharmacol. 2015, 25, 141–147. [Google Scholar] [CrossRef]
  156. Zhang, H.; Xia, X.; Han, F.; Jiang, Q.; Rong, Y.; Song, D.; Wang, Y. Cathelicidin-BF, a Novel Antimicrobial Peptide from Bungarus Fasciatus, Attenuates Disease in a Dextran Sulfate Sodium Model of Colitis. Mol. Pharm. 2015, 12, 1648–1661. [Google Scholar] [CrossRef]
  157. Luo, X.; Budihardjo, I.; Zou, H.; Slaughter, C.; Wang, X. Bid, a Bcl2 Interacting Protein, Mediates Cytochrome c Release from Mitochondria in Response to Activation of Cell Surface Death Receptors. Cell 1998, 94, 481–490. [Google Scholar] [CrossRef]
  158. Zhou, N.; Tian, Y.; Liu, W.; Tu, B.; Gu, T.; Xu, W.; Zou, K.; Lu, L. Effects of Quercetin and Coated Sodium Butyrate Dietary Supplementation in Diquat-Challenged Pullets. Anim. Biosci. 2022, 35, 1434–1443. [Google Scholar] [CrossRef]
  159. Cheng, H.-Y.; Ning, M.-X.; Chen, D.-K.; Ma, W.-T. Interactions Between the Gut Microbiota and the Host Innate Immune Response Against Pathogens. Front. Immunol. 2019, 10, 607. [Google Scholar] [CrossRef]
  160. Kulkarni, D.H.; Rusconi, B.; Floyd, A.N.; Joyce, E.L.; Talati, K.B.; Kousik, H.; Alleyne, D.; Harris, D.L.; Garnica, L.; McDonough, R.; et al. Gut Microbiota Induces Weight Gain and Inflammation in the Gut and Adipose Tissue Independent of Manipulations in Diet, Genetics, and Immune Development. Gut Microbes 2023, 15, 2284240. [Google Scholar] [CrossRef]
  161. Strati, F.; Pujolassos, M.; Burrello, C.; Giuffrè, M.R.; Lattanzi, G.; Caprioli, F.; Troisi, J.; Facciotti, F. Antibiotic-Associated Dysbiosis Affects the Ability of the Gut Microbiota to Control Intestinal Inflammation upon Fecal Microbiota Transplantation in Experimental Colitis Models. Microbiome 2021, 9, 39. [Google Scholar] [CrossRef] [PubMed]
  162. Clemente, J.C.; Manasson, J.; Scher, J.U. The Role of the Gut Microbiome in Systemic Inflammatory Disease. BMJ 2018, 360, j5145. [Google Scholar] [CrossRef]
  163. Samul, D.; Worsztynowicz, P.; Leja, K.; Grajek, W. Beneficial and Harmful Roles of Bacteria from the Clostridium Genus. Acta Biochim. Pol. 2013, 60, 515–521. [Google Scholar] [CrossRef] [PubMed]
  164. Jones-Hall, Y.L.; Kozik, A.; Nakatsu, C. Ablation of Tumor Necrosis Factor Is Associated with Decreased Inflammation and Alterations of the Microbiota in a Mouse Model of Inflammatory Bowel Disease. PLoS ONE 2015, 10, e0119441. [Google Scholar] [CrossRef]
  165. Wu, Y.; Wang, B.; Tang, L.; Zhou, Y.; Wang, Q.; Gong, L.; Ni, J.; Li, W. Probiotic Bacillus Alleviates Oxidative Stress-Induced Liver Injury by Modulating Gut-Liver Axis in a Rat Model. Antioxidants 2022, 11, 291. [Google Scholar] [CrossRef] [PubMed]
  166. Pandit, R.J.; Hinsu, A.T.; Patel, N.V.; Koringa, P.G.; Jakhesara, S.J.; Thakkar, J.R.; Shah, T.M.; Limon, G.; Psifidi, A.; Guitian, J.; et al. Microbial Diversity and Community Composition of Caecal Microbiota in Commercial and Indigenous Indian Chickens Determined Using 16s rDNA Amplicon Sequencing. Microbiome 2018, 6, 115. [Google Scholar] [CrossRef]
  167. Tremaroli, V.; Bäckhed, F. Functional Interactions between the Gut Microbiota and Host Metabolism. Nature 2012, 489, 242–249. [Google Scholar] [CrossRef]
  168. Peng, Y.; Yang, Y.; Tian, Y.; Zhang, M.; Cheng, K.; Zhang, X.; Zhou, M.; Hui, M.; Zhang, Y. Extraction, Characterization, and Antioxidant Activity of Eucommia Ulmoides Polysaccharides. Molecules 2024, 29, 4793. [Google Scholar] [CrossRef]
  169. Wang, H.; Ren, P.; Mang, L.; Shen, N.; Chen, J.; Zhang, Y. In Vitro Fermentation of Novel Microwave-Synthesized Non-Digestible Oligosaccharides and Their Impact on the Composition and Metabolites of Human Gut Microbiota. J. Funct. Food. 2019, 55, 156–166. [Google Scholar] [CrossRef]
  170. Shin, N.-R.; Whon, T.W.; Bae, J.-W. Proteobacteria: Microbial Signature of Dysbiosis in Gut Microbiota. Trends Biotechnol. 2015, 33, 496–503. [Google Scholar] [CrossRef]
  171. Fu, Q.; Tan, Z.; Shi, L.; Xun, W. Resveratrol Attenuates Diquat-Induced Oxidative Stress by Regulating Gut Microbiota and Metabolome Characteristics in Piglets. Front. Microbiol. 2021, 12, 695155. [Google Scholar] [CrossRef] [PubMed]
  172. Chen, J.; Yu, B.; Chen, D.; Zheng, P.; Luo, Y.; Huang, Z.; Luo, J.; Mao, X.; Yu, J.; He, J. Changes of Porcine Gut Microbiota in Response to Dietary Chlorogenic Acid Supplementation. Appl. Microbiol. Biotechnol. 2019, 103, 8157–8168. [Google Scholar] [CrossRef] [PubMed]
  173. Han, H.; Zhong, R.; Zhang, S.; Wang, M.; Wen, X.; Yi, B.; Zhao, Y.; Chen, L.; Zhang, H. Hydroxytyrosol Attenuates Diquat-Induced Oxidative Stress by Activating Nrf2 Pathway and Modulating Colonic Microbiota in Mice. J. Nutr. Biochem. 2023, 113, 109256. [Google Scholar] [CrossRef]
  174. Hu, R.; He, Z.; Liu, M.; Tan, J.; Zhang, H.; Hou, D.-X.; He, J.; Wu, S. Dietary Protocatechuic Acid Ameliorates Inflammation and Up-Regulates Intestinal Tight Junction Proteins by Modulating Gut Microbiota in LPS-Challenged Piglets. J. Anim. Sci. Biotechnol. 2020, 11, 92. [Google Scholar] [CrossRef]
  175. Cockburn, D.W.; Koropatkin, N.M. Polysaccharide Degradation by the Intestinal Microbiota and Its Influence on Human Health and Disease. J. Mol. Biol. 2016, 428, 3230–3252. [Google Scholar] [CrossRef]
  176. Xia, T.; Zhang, B.; Li, S.; Fang, B.; Duan, W.; Zhang, J.; Song, J.; Wang, M. Vinegar Extract Ameliorates Alcohol-Induced Liver Damage Associated with the Modulation of Gut Microbiota in Mice. Food Funct. 2020, 11, 2898–2909. [Google Scholar] [CrossRef] [PubMed]
  177. Wang, Y.-J.; Li, Q.-M.; Zha, X.-Q.; Luo, J.-P. Dendrobium Fimbriatum Hook Polysaccharide Ameliorates Dextran-Sodium-Sulfate-Induced Colitis in Mice via Improving Intestinal Barrier Function, Modulating Intestinal Microbiota, and Reducing Oxidative Stress and Inflammatory Responses. Food Funct. 2022, 13, 143–160. [Google Scholar] [CrossRef]
  178. Yang, S.; Huo, M.; Su, Z.; Wang, F.; Zhang, Y.; Zhong, C.; Shi, Y. The Impact of Dietary Supplementation of Quercetagetin on Growth, Antioxidant Capacity, and Gut Microbiota of Diquat-Challenged Broilers. Front. Microbiol. 2024, 15, 1453145. [Google Scholar] [CrossRef]
  179. Li, L.; Ma, L.; Fu, P. Gut Microbiota-Derived Short-Chain Fatty Acids and Kidney Diseases. Drug Des. Dev. Ther. 2017, 11, 3531–3542. [Google Scholar] [CrossRef]
  180. Parker, B.J.; Wearsch, P.A.; Veloo, A.C.M.; Rodriguez-Palacios, A. The Genus Alistipes: Gut Bacteria with Emerging Implications to Inflammation, Cancer, and Mental Health. Front. Immunol. 2020, 11, 906. [Google Scholar] [CrossRef]
  181. Liu, B.; Wang, W.; Zhu, X.; Sun, X.; Xiao, J.; Li, D.; Cui, Y.; Wang, C.; Shi, Y. Response of Gut Microbiota to Dietary Fiber and Metabolic Interaction with SCFAs in Piglets. Front. Microbiol. 2018, 9, 2344. [Google Scholar] [CrossRef]
  182. Mateos, I.; Combes, S.; Pascal, G.; Cauquil, L.; Barilly, C.; Cossalter, A.-M.; Laffitte, J.; Botti, S.; Pinton, P.; Oswald, I.P. Fumonisin-Exposure Impairs Age-Related Ecological Succession of Bacterial Species in Weaned Pig Gut Microbiota. Toxins 2018, 10, 230. [Google Scholar] [CrossRef]
  183. Hung, D.-Y.; Cheng, Y.-H.; Chen, W.-J.; Hua, K.-F.; Pietruszka, A.; Dybus, A.; Lin, C.-S.; Yu, Y.-H. Bacillus Licheniformis-Fermented Products Reduce Diarrhea Incidence and Alter the Fecal Microbiota Community in Weaning Piglets. Animals 2019, 9, 1145. [Google Scholar] [CrossRef] [PubMed]
  184. Gaukroger, C.H.; Stewart, C.J.; Edwards, S.A.; Walshaw, J.; Adams, I.P.; Kyriazakis, I. Changes in Faecal Microbiota Profiles Associated with Performance and Birthweight of Piglets. Front. Microbiol. 2020, 11, 917. [Google Scholar] [CrossRef] [PubMed]
  185. He, J.; Zheng, W.; Lu, M.; Yang, X.; Xue, Y.; Yao, W. A Controlled Heat Stress during Late Gestation Affects Thermoregulation, Productive Performance, and Metabolite Profiles of Primiparous Sow. J. Therm. Biol. 2019, 81, 33–40. [Google Scholar] [CrossRef]
  186. Vezza, T.; Abad-Jiménez, Z.; Marti-Cabrera, M.; Rocha, M.; Víctor, V.M. Microbiota-Mitochondria Inter-Talk: A Potential Therapeutic Strategy in Obesity and Type 2 Diabetes. Antioxidants 2020, 9, 848. [Google Scholar] [CrossRef] [PubMed]
  187. Clark, A.; Mach, N. The Crosstalk between the Gut Microbiota and Mitochondria during Exercise. Front. Physiol. 2017, 8, 319. [Google Scholar] [CrossRef]
  188. Walsh, L.; Hill, C.; Ross, R.P. Impact of Glyphosate (RoundupTM) on the Composition and Functionality of the Gut Microbiome. Gut Microbes 2023, 15, 2263935. [Google Scholar] [CrossRef]
  189. Shah, A.B.; Baiseitova, A.; Zahoor, M.; Ahmad, I.; Ikram, M.; Bakhsh, A.; Shah, M.A.; Ali, I.; Idress, M.; Ullah, R.; et al. Probiotic Significance of Lactobacillus Strains: A Comprehensive Review on Health Impacts, Research Gaps, and Future Prospects. Gut Microbes 2024, 16, 2431643. [Google Scholar] [CrossRef]
  190. Chu, H.; Duan, Y.; Yang, L.; Schnabl, B. Small Metabolites, Possible Big Changes: A Microbiota-Centered View of Non-Alcoholic Fatty Liver Disease. Gut 2019, 68, 359–370. [Google Scholar] [CrossRef]
  191. Frost, G.; Sleeth, M.L.; Sahuri-Arisoylu, M.; Lizarbe, B.; Cerdan, S.; Brody, L.; Anastasovska, J.; Ghourab, S.; Hankir, M.; Zhang, S.; et al. The Short-Chain Fatty Acid Acetate Reduces Appetite via a Central Homeostatic Mechanism. Nat. Commun. 2014, 5, 3611. [Google Scholar] [CrossRef] [PubMed]
  192. Canfora, E.E.; Jocken, J.W.; Blaak, E.E. Short-Chain Fatty Acids in Control of Body Weight and Insulin Sensitivity. Nat. Rev. Endocrinol. 2015, 11, 577–591. [Google Scholar] [CrossRef] [PubMed]
  193. Rastelli, M.; Cani, P.D.; Knauf, C. The Gut Microbiome Influences Host Endocrine Functions. Endocr. Rev. 2019, 40, 1271–1284. [Google Scholar] [CrossRef]
  194. Wang, J.; Zhu, N.; Su, X.; Gao, Y.; Yang, R. Gut-Microbiota-Derived Metabolites Maintain Gut and Systemic Immune Homeostasis. Cells 2023, 12, 793. [Google Scholar] [CrossRef]
  195. Feng, Z.; Jia, C.; Lin, X.; Hao, H.; Li, S.; Li, F.; Cui, Q.; Chen, Y.; Wu, F.; Xiao, X. The Inhibition of Enterocyte Proliferation by Lithocholic Acid Exacerbates Necrotizing Enterocolitis through Downregulating the Wnt/β-Catenin Signalling Pathway. Cell Prolif. 2022, 55, e13228. [Google Scholar] [CrossRef] [PubMed]
  196. Pan, Y.; Zhang, H.; Li, M.; He, T.; Guo, S.; Zhu, L.; Tan, J.; Wang, B. Novel Approaches in IBD Therapy: Targeting the Gut Microbiota-Bile Acid Axis. Gut Microbes 2024, 16, 2356284. [Google Scholar] [CrossRef]
  197. Kriaa, A.; Mariaule, V.; Jablaoui, A.; Rhimi, S.; Mkaouar, H.; Hernandez, J.; Korkmaz, B.; Lesner, A.; Maguin, E.; Aghdassi, A.; et al. Bile Acids: Key Players in Inflammatory Bowel Diseases? Cells 2022, 11, 901. [Google Scholar] [CrossRef]
  198. Lin, M.; Li, L.; Zhang, Y.; Zheng, L.; Xu, M.; Rong, R.; Zhu, T. Baicalin Ameliorates H2O2 Induced Cytotoxicity in HK-2 Cells through the Inhibition of ER Stress and the Activation of Nrf2 Signaling. Int. J. Mol. Sci. 2014, 15, 12507–12522. [Google Scholar] [CrossRef]
  199. Cheng, H.; Zhang, D.; Wu, J.; Liu, J.; Zhou, Y.; Tan, Y.; Feng, W.; Peng, C. Interactions between Gut Microbiota and Polyphenols: A Mechanistic and Metabolomic Review. Phytomedicine 2023, 119, 154979. [Google Scholar] [CrossRef]
  200. Xie, F.; Yang, W.; Xing, M.; Zhang, H.; Ai, L. Natural Polyphenols-Gut Microbiota Interactions and Effects on Glycolipid Metabolism via Polyphenols-Gut-Brain Axis: A State-of-the-Art Review. Trends Food Sci. Technol. 2023, 140, 104171. [Google Scholar] [CrossRef]
  201. Martínez, L.; Ros, G.; Nieto, G. Hydroxytyrosol: Health Benefits and Use as Functional Ingredient in Meat. Medicines 2018, 5, 13. [Google Scholar] [CrossRef]
  202. Li, J.; Chen, Y.; Li, R.; Zhang, X.; Chen, T.; Mei, F.; Liu, R.; Chen, M.; Ge, Y.; Hu, H.; et al. Gut Microbial Metabolite Hyodeoxycholic Acid Targets the TLR4/MD2 Complex to Attenuate Inflammation and Protect against Sepsis. Mol. Ther. 2023, 31, 1017–1032. [Google Scholar] [CrossRef] [PubMed]
  203. Paumgartner, G.; Beuers, U. Ursodeoxycholic Acid in Cholestatic Liver Disease: Mechanisms of Action and Therapeutic Use Revisited. Hepatology 2002, 36, 525–531. [Google Scholar] [CrossRef] [PubMed]
  204. Kusaczuk, M. Tauroursodeoxycholate-Bile Acid with Chaperoning Activity: Molecular and Cellular Effects and Therapeutic Perspectives. Cells 2019, 8, 1471. [Google Scholar] [CrossRef]
  205. Zangerolamo, L.; Vettorazzi, J.F.; Rosa, L.R.O.; Carneiro, E.M.; Barbosa, H.C.L. The Bile Acid TUDCA and Neurodegenerative Disorders: An Overview. Life Sci. 2021, 272, 119252. [Google Scholar] [CrossRef] [PubMed]
  206. Fu, X.; Liu, Z.; Zhu, C.; Mou, H.; Kong, Q. Nondigestible Carbohydrates, Butyrate, and Butyrate-Producing Bacteria. Crit. Rev. Food Sci. Nutr. 2019, 59 (Suppl. S1), S130–S152. [Google Scholar] [CrossRef]
  207. Mao, X.; Lv, M.; Yu, B.; He, J.; Zheng, P.; Yu, J.; Wang, Q.; Chen, D. The Effect of Dietary Tryptophan Levels on Oxidative Stress of Liver Induced by Diquat in Weaned Piglets. J. Anim. Sci. Biotechnol. 2014, 5, 49. [Google Scholar] [CrossRef]
  208. Roager, H.M.; Licht, T.R. Microbial Tryptophan Catabolites in Health and Disease. Nat. Commun. 2018, 9, 3294. [Google Scholar] [CrossRef]
  209. Devlin, A.S.; Marcobal, A.; Dodd, D.; Nayfach, S.; Plummer, N.; Meyer, T.; Pollard, K.S.; Sonnenburg, J.L.; Fischbach, M.A. Modulation of a Circulating Uremic Solute via Rational Genetic Manipulation of the Gut Microbiota. Cell Host Microbe 2016, 20, 709–715. [Google Scholar] [CrossRef]
  210. Rothhammer, V.; Mascanfroni, I.D.; Bunse, L.; Takenaka, M.C.; Kenison, J.E.; Mayo, L.; Chao, C.-C.; Patel, B.; Yan, R.; Blain, M.; et al. Type I Interferons and Microbial Metabolites of Tryptophan Modulate Astrocyte Activity and Central Nervous System Inflammation via the Aryl Hydrocarbon Receptor. Nat. Med. 2016, 22, 586–597. [Google Scholar] [CrossRef]
  211. Shimada, Y.; Kinoshita, M.; Harada, K.; Mizutani, M.; Masahata, K.; Kayama, H.; Takeda, K. Commensal Bacteria-Dependent Indole Production Enhances Epithelial Barrier Function in the Colon. PLoS ONE 2013, 8, e80604. [Google Scholar] [CrossRef] [PubMed]
  212. Niwa, T.; Shimizu, H. Indoxyl Sulfate Induces Nephrovascular Senescence. J. Ren. Nutr. 2012, 22, 102–106. [Google Scholar] [CrossRef] [PubMed]
  213. Britan, A.; Maffre, V.; Tone, S.; Drevet, J.R. Quantitative and Spatial Differences in the Expression of Tryptophan-Metabolizing Enzymes in Mouse Epididymis. Cell Tissue Res. 2006, 324, 301–310. [Google Scholar] [CrossRef]
  214. Dairam, A.; Antunes, E.M.; Saravanan, K.S.; Daya, S. Non-Steroidal Anti-Inflammatory Agents, Tolmetin and Sulindac, Inhibit Liver Tryptophan 2,3-Dioxygenase Activity and Alter Brain Neurotransmitter Levels. Life Sci. 2006, 79, 2269–2274. [Google Scholar] [CrossRef]
  215. Kraal, L.; Abubucker, S.; Kota, K.; Fischbach, M.A.; Mitreva, M. The Prevalence of Species and Strains in the Human Microbiome: A Resource for Experimental Efforts. PLoS ONE 2014, 9, e97279. [Google Scholar] [CrossRef]
  216. Lee, J.-H.; Lee, J. Indole as an Intercellular Signal in Microbial Communities. FEMS Microbiol. Rev. 2010, 34, 426–444. [Google Scholar] [CrossRef] [PubMed]
  217. Han, H.; Yi, B.; Zhong, R.; Wang, M.; Zhang, S.; Ma, J.; Yin, Y.; Yin, J.; Chen, L.; Zhang, H. From Gut Microbiota to Host Appetite: Gut Microbiota-Derived Metabolites as Key Regulators. Microbiome 2021, 9, 162. [Google Scholar] [CrossRef]
  218. Han, H.; Li, Y.; Fang, J.; Liu, G.; Yin, J.; Li, T.; Yin, Y. Gut Microbiota and Type 1 Diabetes. Int. J. Mol. Sci. 2018, 19, 995. [Google Scholar] [CrossRef]
  219. Waldman, M.; Peterson, S.J.; Arad, M.; Hochhauser, E. The Role of 20-HETE in Cardiovascular Diseases and Its Risk Factors. Prostaglandins Other Lipid Mediat. 2016, 125, 108–117. [Google Scholar] [CrossRef]
  220. Nambo-Venegas, R.; Valdez-Vargas, C.; Cisneros, B.; Palacios-González, B.; Vela-Amieva, M.; Ibarra-González, I.; Cerecedo-Zapata, C.M.; Martínez-Cruz, E.; Cortés, H.; Reyes-Grajeda, J.P.; et al. Altered Plasma Acylcarnitines and Amino Acids Profile in Spinocerebellar Ataxia Type 7. Biomolecules 2020, 10, 390. [Google Scholar] [CrossRef]
  221. Chen, Y.-J.; Luo, L.; Zhang, G.-Z.; Li, Z.; Bai, F.-J.; Shi, Y.-Q.; Yang, H.-S. Effect of Dietary L-Malic Acid Supplementation on Growth, Feed Utilization and Digestive Function of Juvenile GIFT Tilapia Oreochromis Niloticus (Linnaeus, 1758). J. Appl. Ichthyol. 2016, 32, 1118–1123. [Google Scholar] [CrossRef]
  222. Oniszczuk, A.; Oniszczuk, T.; Gancarz, M.; Szymańska, J. Role of Gut Microbiota, Probiotics and Prebiotics in the Cardiovascular Diseases. Molecules 2021, 26, 1172. [Google Scholar] [CrossRef]
  223. Wang, Y.; Wu, Y.; Wang, Y.; Xu, H.; Mei, X.; Yu, D.; Wang, Y.; Li, W. Antioxidant Properties of Probiotic Bacteria. Nutrients 2017, 9, 521. [Google Scholar] [CrossRef]
  224. Reuben, R.C.; Roy, P.C.; Sarkar, S.L.; Alam, R.-U.; Jahid, I.K. Isolation, Characterization, and Assessment of Lactic Acid Bacteria toward Their Selection as Poultry Probiotics. BMC Microbiol. 2019, 19, 253. [Google Scholar] [CrossRef]
  225. Kong, Y.; Olejar, K.J.; On, S.L.W.; Chelikani, V. The Potential of Lactobacillus Spp. for Modulating Oxidative Stress in the Gastrointestinal Tract. Antioxidants 2020, 9, 610. [Google Scholar] [CrossRef] [PubMed]
  226. He, J.; He, Y.; Pan, D.; Cao, J.; Sun, Y.; Zeng, X. Associations of Gut Microbiota with Heat Stress-Induced Changes of Growth, Fat Deposition, Intestinal Morphology, and Antioxidant Capacity in Ducks. Front. Microbiol. 2019, 10, 903. [Google Scholar] [CrossRef] [PubMed]
  227. Clavijo, V.; Flórez, M.J.V. The Gastrointestinal Microbiome and Its Association with the Control of Pathogens in Broiler Chicken Production: A Review. Poult. Sci. 2018, 97, 1006–1021. [Google Scholar] [CrossRef]
  228. Li, M.; Mao, B.; Tang, X.; Zhang, Q.; Zhao, J.; Chen, W.; Cui, S. Lactic Acid Bacteria Derived Extracellular Vesicles: Emerging Bioactive Nanoparticles in Modulating Host Health. Gut Microbes 2024, 16, 2427311. [Google Scholar] [CrossRef]
  229. Savcı, Y.; Kırbaş, O.K.; Bozkurt, B.T.; Abdik, E.A.; Taşlı, P.N.; Şahin, F.; Abdik, H. Grapefruit-Derived Extracellular Vesicles as a Promising Cell-Free Therapeutic Tool for Wound Healing. Food Funct. 2021, 12, 5144–5156. [Google Scholar] [CrossRef]
  230. Feng, S.; Liu, Y.; Xu, J.; Fan, J.; Li, J.; Wu, Z.; Sun, Y.; Xiong, W. Three Strains of Lactobacillus Derived from Piglets Alleviated Intestinal Oxidative Stress Induced by Diquat through Extracellular Vesicles. Nutrients 2023, 15, 4198. [Google Scholar] [CrossRef]
  231. Liu, Y.; Yuan, J.; Xi, W.; Wang, Z.; Liu, H.; Zhang, K.; Zhao, J.; Wang, Y. Lactiplantibacillus Plantarum Ameliorated Morphological Damage and Barrier Dysfunction and Reduced Apoptosis and Ferroptosis in the Jejunum of Oxidatively Stressed Piglets. Animals 2024, 14, 3335. [Google Scholar] [CrossRef]
  232. Wu, G.; Bazer, F.W.; Davis, T.A.; Jaeger, L.A.; Johnson, G.A.; Kim, S.W.; Knabe, D.A.; Meininger, C.J.; Spencer, T.E.; Yin, Y.-L. Important Roles for the Arginine Family of Amino Acids in Swine Nutrition and Production. Livest. Sci. 2007, 112, 8–22. [Google Scholar] [CrossRef]
  233. Zheng, P.; Yu, B.; He, J.; Tian, G.; Luo, Y.; Mao, X.; Zhang, K.; Che, L.; Chen, D. Protective Effects of Dietary Arginine Supplementation against Oxidative Stress in Weaned Piglets. Br. J. Nutr. 2013, 109, 2253–2260. [Google Scholar] [CrossRef] [PubMed]
  234. Viana, M.L.; Santos, R.G.C.; Generoso, S.V.; Arantes, R.M.E.; Correia, M.I.T.D.; Cardoso, V.N. Pretreatment with Arginine Preserves Intestinal Barrier Integrity and Reduces Bacterial Translocation in Mice. Nutrition 2010, 26, 218–223. [Google Scholar] [CrossRef] [PubMed]
  235. Bitterman, J.L.; Chung, J.H. Metabolic Effects of Resveratrol: Addressing the Controversies. Cell Mol. Life Sci. 2015, 72, 1473–1488. [Google Scholar] [CrossRef]
  236. Zhang, C.; Luo, J.; Yu, B.; Zheng, P.; Huang, Z.; Mao, X.; He, J.; Yu, J.; Chen, J.; Chen, D. Dietary Resveratrol Supplementation Improves Meat Quality of Finishing Pigs through Changing Muscle Fiber Characteristics and Antioxidative Status. Meat Sci. 2015, 102, 15–21. [Google Scholar] [CrossRef]
  237. Xia, N.; Daiber, A.; Förstermann, U.; Li, H. Antioxidant Effects of Resveratrol in the Cardiovascular System. Br. J. Pharmacol. 2017, 174, 1633–1646. [Google Scholar] [CrossRef]
  238. Zhang, H.Z.; Chen, D.W.; He, J.; Zheng, P.; Yu, J.; Mao, X.B.; Huang, Z.Q.; Luo, Y.H.; Luo, J.Q.; Yu, B. Long-Term Dietary Resveratrol Supplementation Decreased Serum Lipids Levels, Improved Intramuscular Fat Content, and Changed the Expression of Several Lipid Metabolism-Related miRNAs and Genes in Growing-Finishing Pigs1. J. Anim. Sci. 2019, 97, 1745–1756. [Google Scholar] [CrossRef]
  239. Madunić, J.; Madunić, I.V.; Gajski, G.; Popić, J.; Garaj-Vrhovac, V. Apigenin: A Dietary Flavonoid with Diverse Anticancer Properties. Cancer Lett. 2018, 413, 11–22. [Google Scholar] [CrossRef]
  240. Li, F.; Lang, F.; Zhang, H.; Xu, L.; Wang, Y.; Zhai, C.; Hao, E. Apigenin Alleviates Endotoxin-Induced Myocardial Toxicity by Modulating Inflammation, Oxidative Stress, and Autophagy. Oxidative Med. Cell Longev. 2017, 2017, 2302896. [Google Scholar] [CrossRef]
  241. Sánchez-Marzo, N.; Pérez-Sánchez, A.; Ruiz-Torres, V.; Martínez-Tébar, A.; Castillo, J.; Herranz-López, M.; Barrajón-Catalán, E. Antioxidant and Photoprotective Activity of Apigenin and Its Potassium Salt Derivative in Human Keratinocytes and Absorption in Caco-2 Cell Monolayers. Int. J. Mol. Sci. 2019, 20, 2148. [Google Scholar] [CrossRef] [PubMed]
  242. Wang, D.; Yang, Y.; Zou, X.; Zhang, J.; Zheng, Z.; Wang, Z. Antioxidant Apigenin Relieves Age-Related Muscle Atrophy by Inhibiting Oxidative Stress and Hyperactive Mitophagy and Apoptosis in Skeletal Muscle of Mice. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, 2081–2088. [Google Scholar] [CrossRef]
  243. Zhou, N.; Tian, Y.; Liu, W.; Tu, B.; Xu, W.; Gu, T.; Zou, K.; Lu, L. Protective Effects of Resveratrol and Apigenin Dietary Supplementation on Serum Antioxidative Parameters and mRNAs Expression in the Small Intestines of Diquat-Challenged Pullets. Front. Vet. Sci. 2022, 9, 850769. [Google Scholar] [CrossRef]
  244. Ak, T.; Gülçin, I. Antioxidant and Radical Scavenging Properties of Curcumin. Chem. Biol. Interact. 2008, 174, 27–37. [Google Scholar] [CrossRef] [PubMed]
  245. Kotha, R.R.; Luthria, D.L. Curcumin: Biological, Pharmaceutical, Nutraceutical, and Analytical Aspects. Molecules 2019, 24, 2930. [Google Scholar] [CrossRef]
  246. Wu, F.; Zhao, M.; Tang, Z.; Wang, F.; Han, S.; Liu, S.; Chen, B. Curcumin Alleviates Cecal Oxidative Injury in Diquat-Induced Broilers by Regulating the Nrf2/ARE Pathway and Microflora. Poult. Sci. 2024, 103, 103651. [Google Scholar] [CrossRef]
  247. Shi, H.; Li, X.; Hou, C.; Chen, L.; Zhang, Y.; Li, J. Effects of Pomegranate Peel Polyphenols Combined with Inulin on Gut Microbiota and Serum Metabolites of High-Fat-Induced Obesity Rats. J. Agric. Food Chem. 2023, 71, 5733–5744. [Google Scholar] [CrossRef]
  248. Avery, J.C.; Hoffmann, P.R. Selenium, Selenoproteins, and Immunity. Nutrients 2018, 10, 1203. [Google Scholar] [CrossRef] [PubMed]
  249. Chopyk, D.M.; Grakoui, A. Contribution of the Intestinal Microbiome and Gut Barrier to Hepatic Disorders. Gastroenterology 2020, 159, 849–863. [Google Scholar] [CrossRef]
  250. Wexler, H.M. Bacteroides: The Good, the Bad, and the Nitty-Gritty. Clin. Microbiol. Rev. 2007, 20, 593–621. [Google Scholar] [CrossRef]
  251. Zafar, H.; Saier, M.H. Gut Bacteroides Species in Health and Disease. Gut Microbes 2021, 13, 1848158. [Google Scholar] [CrossRef] [PubMed]
  252. Durant, L.; Stentz, R.; Noble, A.; Brooks, J.; Gicheva, N.; Reddi, D.; O’Connor, M.J.; Hoyles, L.; McCartney, A.L.; Man, R.; et al. Bacteroides Thetaiotaomicron-Derived Outer Membrane Vesicles Promote Regulatory Dendritic Cell Responses in Health but Not in Inflammatory Bowel Disease. Microbiome 2020, 8, 88. [Google Scholar] [CrossRef] [PubMed]
  253. Van den Abbeele, P.; Belzer, C.; Goossens, M.; Kleerebezem, M.; De Vos, W.M.; Thas, O.; De Weirdt, R.; Kerckhof, F.-M.; Van de Wiele, T. Butyrate-Producing Clostridium Cluster XIVa Species Specifically Colonize Mucins in an in Vitro Gut Model. ISME J. 2013, 7, 949–961. [Google Scholar] [CrossRef]
  254. Gong, X.; Wu, M.; Jiang, Y.; Wang, H. Effects of Different Temperatures and pH Values on Volatile Fatty Acids Production during Codigestion of Food Waste and Thermal-Hydrolysed Sewage Sludge and Subsequent Volatile Fatty Acids for Polyhydroxyalkanoates Production. Bioresour. Technol. 2021, 333, 125149. [Google Scholar] [CrossRef]
  255. Chen, Y.; Jin, L.; Li, Y.; Xia, G.; Chen, C.; Zhang, Y. Bamboo-Shaving Polysaccharide Protects against High-Diet Induced Obesity and Modulates the Gut Microbiota of Mice. J. Funct. Food. 2018, 49, 20–31. [Google Scholar] [CrossRef]
  256. Verstreken, I.; Laleman, W.; Wauters, G.; Verhaegen, J. Desulfovibrio Desulfuricans Bacteremia in an Immunocompromised Host with a Liver Graft and Ulcerative Colitis. J. Clin. Microbiol. 2012, 50, 199–201. [Google Scholar] [CrossRef]
  257. Qiao, L.; Dou, X.; Song, X.; Chang, J.; Pi, S.; Zhang, X.; Zhu, L.; Zeng, X.; Xu, C. Protective Effect of Biogenic Selenium Nanoparticles against Diquat-Induced Acute Toxicity via Regulation of Gut Microbiota and Its Metabolites. Food Chem. Toxicol. 2022, 170, 113480. [Google Scholar] [CrossRef]
  258. Bollag, W.B. Regulation of Aldosterone Synthesis and Secretion. Compr. Physiol. 2014, 4, 1017–1055. [Google Scholar] [CrossRef]
  259. Dominko, K.; Đikić, D. Glutathionylation: A Regulatory Role of Glutathione in Physiological Processes. Arch. Ind. Hyg. Toxicol. 2018, 69, 1–24. [Google Scholar] [CrossRef]
  260. Li, Q.J.; Wang, Z.G.; Liu, Q.; Xie, Y.; Hu, H.L. [Research Status of Periplaneta Americana with Analyses and Prospects of Key Issues]. China J. Chin. Mater. Med. 2018, 43, 1507–1516. [Google Scholar] [CrossRef]
  261. Nguyen, T.; Chen, X.; Chai, J.; Li, R.; Han, X.; Chen, X.; Liu, S.; Chen, M.; Xu, X. Antipyretic, Anti-Inflammatory and Analgesic Activities of Periplaneta Americana Extract and Underlying Mechanisms. Biomed. Pharmacother. 2020, 123, 109753. [Google Scholar] [CrossRef] [PubMed]
  262. Lu, S.; Xu, S.; Chen, L.; Deng, Y.; Feng, J. Periplaneta Americana Extract Pretreatment Alleviates Oxidative Stress and Inflammation and Increases the Abundance of Gut Akkermansia Muciniphila in Diquat-Induced Mice. Antioxidants 2022, 11, 1806. [Google Scholar] [CrossRef] [PubMed]
  263. Panzetta, M.E.; Valdivia, R.H. Akkermansia in the Gastrointestinal Tract as a Modifier of Human Health. Gut Microbes 2024, 16, 2406379. [Google Scholar] [CrossRef]
  264. Hagi, T.; Belzer, C. The Interaction of Akkermansia Muciniphila with Host-Derived Substances, Bacteria and Diets. Appl. Microbiol. Biotechnol. 2021, 105, 4833–4841. [Google Scholar] [CrossRef]
  265. Zhang, T.; Li, Q.; Cheng, L.; Buch, H.; Zhang, F. Akkermansia Muciniphila Is a Promising Probiotic. Microb. Biotechnol. 2019, 12, 1109–1125. [Google Scholar] [CrossRef]
  266. Chelakkot, C.; Choi, Y.; Kim, D.-K.; Park, H.T.; Ghim, J.; Kwon, Y.; Jeon, J.; Kim, M.-S.; Jee, Y.-K.; Gho, Y.S.; et al. Akkermansia Muciniphila-Derived Extracellular Vesicles Influence Gut Permeability through the Regulation of Tight Junctions. Exp. Mol. Med. 2018, 50, e450. [Google Scholar] [CrossRef] [PubMed]
  267. LeBlanc, J.G.; Chain, F.; Martín, R.; Bermúdez-Humarán, L.G.; Courau, S.; Langella, P. Beneficial Effects on Host Energy Metabolism of Short-Chain Fatty Acids and Vitamins Produced by Commensal and Probiotic Bacteria. Microb. Cell Fact. 2017, 16, 79. [Google Scholar] [CrossRef]
  268. Xiao, H.; Wu, M.; Shao, F.; Guan, G.; Huang, B.; Tan, B.; Yin, Y. N-Acetyl-L-Cysteine Protects the Enterocyte against Oxidative Damage by Modulation of Mitochondrial Function. Mediat. Inflamm. 2016, 2016, 8364279. [Google Scholar] [CrossRef]
  269. Biasi, F.; Deiana, M.; Guina, T.; Gamba, P.; Leonarduzzi, G.; Poli, G. Wine Consumption and Intestinal Redox Homeostasis. Redox Biol. 2014, 2, 795–802. [Google Scholar] [CrossRef]
  270. Wang, S.; Mu, L.; Yu, C.; He, Y.; Hu, X.; Jiao, Y.; Xu, Z.; You, S.; Liu, S.-L.; Bao, H. Microbial Collaborations and Conflicts: Unraveling Interactions in the Gut Ecosystem. Gut Microbes 2024, 16, 2296603. [Google Scholar] [CrossRef]
  271. Cho, N.A.; Strayer, K.; Dobson, B.; McDonald, B. Pathogenesis and Therapeutic Opportunities of Gut Microbiome Dysbiosis in Critical Illness. Gut Microbes 2024, 16, 2351478. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic overview illustrating the multiple pathways through which diquat triggers ROS production, inflammatory factor release, and the activation of mitophagy and apoptosis in IECs. (1) SCFAs (e.g., butyrate) and TUDCA derived from intestinal homeostasis activate Nrf2 signaling to upregulate antioxidant enzymes (HO-1/CAT/SOD/GSH-PX), constituting a critical defense against oxidative injury. (2) DQ elicits OS through dual mechanisms: the direct induction of ROS generation and the indirect disruption of redox homeostasis via dysbiosis. (3) Pro-inflammatory signaling: NF-κB/NLRP3 activation upregulates cytokines (IL-6, IL-1β, IL-18, TNF-α). (4) Mitochondrial–lysosomal axis dysfunction: ROS accumulation induces PINK1-Parkin-dependent mitophagy, and lysosomal activity mediates the clearance of damaged mitochondria. (5) OS triggers mitochondrial-mediated apoptosis (caspase-9/caspase-3 cascade activation). SCFA: Short-chain fatty acid; TUDCA: Tauroursodeoxycholic acid; Nrf2: Nuclear factor erythroid 2-related factor 2; HO-1: Heme oxygenase-1; CAT: Catalase; SOD: Superoxide dismutase; GSH-PX: Glutathione peroxidase; ROS: Reactive oxygen species; NF-kB: Nuclear factor kappa-B; NLRP3: NLR family pyrin domain containing 3.
Figure 1. Schematic overview illustrating the multiple pathways through which diquat triggers ROS production, inflammatory factor release, and the activation of mitophagy and apoptosis in IECs. (1) SCFAs (e.g., butyrate) and TUDCA derived from intestinal homeostasis activate Nrf2 signaling to upregulate antioxidant enzymes (HO-1/CAT/SOD/GSH-PX), constituting a critical defense against oxidative injury. (2) DQ elicits OS through dual mechanisms: the direct induction of ROS generation and the indirect disruption of redox homeostasis via dysbiosis. (3) Pro-inflammatory signaling: NF-κB/NLRP3 activation upregulates cytokines (IL-6, IL-1β, IL-18, TNF-α). (4) Mitochondrial–lysosomal axis dysfunction: ROS accumulation induces PINK1-Parkin-dependent mitophagy, and lysosomal activity mediates the clearance of damaged mitochondria. (5) OS triggers mitochondrial-mediated apoptosis (caspase-9/caspase-3 cascade activation). SCFA: Short-chain fatty acid; TUDCA: Tauroursodeoxycholic acid; Nrf2: Nuclear factor erythroid 2-related factor 2; HO-1: Heme oxygenase-1; CAT: Catalase; SOD: Superoxide dismutase; GSH-PX: Glutathione peroxidase; ROS: Reactive oxygen species; NF-kB: Nuclear factor kappa-B; NLRP3: NLR family pyrin domain containing 3.
Antioxidants 14 00721 g001
Figure 2. DQ compromises the intestinal barrier and increases intestinal permeability. (1) Tight junction (TJ) disruption: the downregulation of claudin, ZO-1, and occludin compromises epithelial barrier integrity. (2) Reduced MUC2 secretion from goblet cells exacerbates mucosal vulnerability. (3) DQ elevates serum DAO and DLA levels, verifying the impairment of gut barrier integrity. (4) DQ enhances tryptophan metabolism, thereby augmenting the production of IS, a uremic toxin associated with renal pathophysiology. ↑: upregulation; ↓: downregulation; TJ: Tight junction; ZO-1: Zonula occludens-1; MUC2: Mucin 2; DLA: D-lactate; DAO: Diamine oxidase; IS: Indoxyl sulfate.
Figure 2. DQ compromises the intestinal barrier and increases intestinal permeability. (1) Tight junction (TJ) disruption: the downregulation of claudin, ZO-1, and occludin compromises epithelial barrier integrity. (2) Reduced MUC2 secretion from goblet cells exacerbates mucosal vulnerability. (3) DQ elevates serum DAO and DLA levels, verifying the impairment of gut barrier integrity. (4) DQ enhances tryptophan metabolism, thereby augmenting the production of IS, a uremic toxin associated with renal pathophysiology. ↑: upregulation; ↓: downregulation; TJ: Tight junction; ZO-1: Zonula occludens-1; MUC2: Mucin 2; DLA: D-lactate; DAO: Diamine oxidase; IS: Indoxyl sulfate.
Antioxidants 14 00721 g002
Table 1. Changes in gut microbiota and metabolites of experimental animal models after treatment with DQ or intervention.
Table 1. Changes in gut microbiota and metabolites of experimental animal models after treatment with DQ or intervention.
SubjectsChanges (DQ)InterventionChanges (Intervention)Ref.
C57BL/6 miceAllobaculum, Providencia, Escherichia-Shigella, Bacteroidetes, proteobacteria↑; FirmicutesPeriplaneta americana extract (PAE)Akkermansia muciniphila↑; Bacteroidetes[262]
Weaned pigletsHCA, HDCA, TUDCA↓HydroxytyrosolHCA, HDCA, TUDCA↑[137]
ICR miceFirmicutes↓; BacteroidetesHydroxytyrosolFirmicutes, Lactobacillus↑; Bacteroidetes↓; butyrate↑; glycerophospholipid metabolism, pentose, glucuronate interconversions↓[173]
C57BL/6J miceBacteroidota, Coriobacteriia, Enterorhabdus↓; Escherichia–ShigellaL. delbrueckii, L. amylovorus, and L. salivarius EVsEnterococcus↓; Parasutterella, Bifidobacterium, Erysipelatoclostridium[230]
MiceFirmicutesEucommia ulmoides polysaccharide (EUPS)Firmicutes, Ligilactobacillus↑; Helicobacter[168]
WOD168 broilersLactobacillus, AlistipesQuercetagetin (QG)Lactobacillus, Alistipes[178]
Weaned pigletsFirmicutes, Actinobacteria, Ruminococcaceae UCG-005, Eubacterium coprostanoligenesResveratrol (RES)Clostridium sensu stricto 1, Lachnospiraceae[171]
C57BL/6 mice Selenium nanoparticles (SeNPs)Akkermansia, Muribaculaceae, Bacteroides, Parabacteroides[257]
C57BL/6 miceBacteroides, Helicobacter↑; Firmicutes, Pediococcus, Enterococcus, DubosiellaPediococcus pentosaceus ZJUAF-4; VCZJUAF-4 reversed these changes induced by DQ and the reversed abilities of ZJUAF-4 seemed to be higher than those of VC[63]
Cobb broilersLactobacillaceae, Victivallis, Bacillus↓; Saccharopolyspora_hir-suta, Staphylococcus_succinusCurcumin (CUR)Lactobacillaceae, Ruminococcaceae_Clostridium↑; Saccharopolyspora_hirsuta, Staphylococcus_succinus↓;
acetate, total SCFAs↑
[246]
C57BL/6J mice Hydrogenobacter thermophilus
strain (HtSOD)
Dubosiella, Alistipes[42]
Weaned pigletsAcidobacteria↓; Turicibacteraceae, Clostridium, TuricibacterBacillus amyloliquefaciens SC06Ruminococcaceae, Clostridium↓; Pasteurellaceae, Lactobacillus, Actinobacillus[81]
SD ratsg_Lactobacillus, p_Firmicutes, g_Akkermansia, p_Verrucomicrobia↓; p_Proteobacteria, g_Escherichia, s_EscherichiacoliBacillus SC06g_Anaerofilum, s_Bacteroides uniformis↑; s_Oscillospira guilliermondil[165]
C57BL/6 mice SeNPsBacteroidetes, Clostridium_XlVa↑; Verrucomicrobia, Desulfovibrio↓;
total SCFAs, butyrate, isobutyrate, valerate, isovalerate↑
[95]
↑: upregulation; ↓: downregulation; DQ: Diquat; Ref.: References; HCA: Hyocholic acid; HDCA: Hyodeoxycholic acid; TUDCA: Tauroursodeoxycholic acid; EVs: Extracellular vesicles; SCFAs: Short-chain fatty acids.
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

He, J.; Tang, Q.; Liu, Y.-C.; Wang, L.-J.; Chai, Y.-F. Impact of Diquat on the Intestinal Health and the Composition and Function of the Gut Microbiome. Antioxidants 2025, 14, 721. https://doi.org/10.3390/antiox14060721

AMA Style

He J, Tang Q, Liu Y-C, Wang L-J, Chai Y-F. Impact of Diquat on the Intestinal Health and the Composition and Function of the Gut Microbiome. Antioxidants. 2025; 14(6):721. https://doi.org/10.3390/antiox14060721

Chicago/Turabian Style

He, Jiao, Qing Tang, Yan-Cun Liu, Li-Jun Wang, and Yan-Fen Chai. 2025. "Impact of Diquat on the Intestinal Health and the Composition and Function of the Gut Microbiome" Antioxidants 14, no. 6: 721. https://doi.org/10.3390/antiox14060721

APA Style

He, J., Tang, Q., Liu, Y.-C., Wang, L.-J., & Chai, Y.-F. (2025). Impact of Diquat on the Intestinal Health and the Composition and Function of the Gut Microbiome. Antioxidants, 14(6), 721. https://doi.org/10.3390/antiox14060721

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