Next Article in Journal
Pneumocystis jirovecii Pneumonia Diagnosis with Oropharyngeal Wash PCR in Immunocompromised Patients—A Systematic Review
Previous Article in Journal
Serum Granulin Concentrations Are Elevated in Prediabetes and Newly Diagnosed Diabetes: A Cross-Sectional Study
Previous Article in Special Issue
Tranilast Reduces Intestinal Ischemia Reperfusion Injury in Rats Through the Upregulation of Heme-Oxygenase (HO)-1
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Relevance of Experimental Models in Assessing the Impact of Oxidative Stress on Intestinal Pathology

by
Cristian Dan Pavel
1,
Cristina Gales
1,
Irina Andreea Pavel
2,* and
Carmen Lăcrămioara Zamfir
1
1
Department of Morpho-Functional Sciences I, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, University Street, No.16, 700115 Iasi, Romania
2
Department of Surgery II, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, University Street, No.16, 700115 Iasi, Romania
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2025, 14(18), 6569; https://doi.org/10.3390/jcm14186569
Submission received: 5 August 2025 / Revised: 4 September 2025 / Accepted: 15 September 2025 / Published: 18 September 2025

Abstract

Oxidative stress is a state of imbalance between the process of producing and removing reactive oxygen species (ROS). With advancing age or in certain situations where oxidative stress cannot be combated, various pathologies such as inflammatory bowel diseases or neoplasia may occur. Over the past decade, a surge of intriguing discoveries has linked subtoxic levels of oxidative stress to key processes, including the maintenance of mucosal homeostasis, regulation of protective inflammation, and even the control of tissue wound healing. Given the complexity and limited understanding of oxidative mechanisms involved in human intestinal pathology, the relevance of experimental models becomes a critical consideration in efforts to elucidate these processes. Although diverse, none of these models fully replicate human digestive pathology; however, they remain valuable for developing new therapeutic strategies. This paper examines the main markers of oxidative stress and its mechanism and their impact on the intestinal tract, as well as the most widely used animal models that have contributed valuable insights into the pathogenesis of inflammatory bowel diseases (IBD).

1. Introduction

Oxidative stress occurs when antioxidant mechanisms cannot remove excess of reactive oxygen species (ROS) from the body and cause damage to proteins, lipids, and DNA. Over the past decade, a surge of intriguing discoveries has linked subtoxic levels of oxidative stress to key processes, including the maintenance of mucosal homeostasis, regulation of protective inflammation, and even the control of tissue wound healing. The production of ROS is an important factor in the pathogenesis of many inflammatory disorders, such as those in the gastrointestinal system, activating specific inflammatory signaling pathways and affecting immune reactivity. If the contribution of oxidative stress to the onset and progression of intestinal diseases is now broadly accepted, many of its underlying mechanisms and their relationships remain poorly understood [1,2].
Heightened oxidative stress directly affects intestinal morphology and permeability, together with gut microbiota, digestion, absorption, metabolic processes, and the immune response. In the gut, disruption of the mucosal barrier produces an activation of the innate immune system and an acute inflammatory reaction, initially located in lamina propria [1,2]. Elucidating the role of oxidative stress in digestive disorders not only facilitates the understanding of particular mechanisms involved but also is important in advancing new therapeutic strategies [3].
Given the current gaps in understanding these mechanisms, experimental models are essential tools for assessing oxidative stress-induced alterations in the intestine [3]. There are a lot of factors which should be considered when determining the relevance of experimental models. On this basis it can be concluded that no experimental model can fully replicate the diversity and complexity of the intestinal pathology, but they remain essential tools to investigate oxidative stress-induced alterations in intestinal structure and function [4].
Therefore, the aim of this narrative review is to synthesize the available evidence on experimental models of intestinal oxidative stress, highlighting their advantages, limitations, and the reasons why such models remain indispensable, despite the fact that no single experimental model can fully replicate the diversity and complexity of intestinal pathology.

2. Materials and Methods

The search was conducted in PubMed, Scopus, and ScienceDirect using the following keywords, experimental models, intestine, oxidative stress, glutathione, combined with Boolean operators (example: oxidative stress AND intestine AND experimental models). The last search was performed in March 2025.
We limited our search primarily to the last 10 years in order to capture the most recent developments in experimental models. However, we also included older, seminal studies that remain relevant for understanding fundamental mechanisms. Inclusion criteria included the following: prospective and retrospective studies, meta-analysis, and reviews. Exclusion criteria included the following: abstracts, conference proceedings, book chapters, editorials, and articles not addressing the scope of the review. Duplicates were removed manually. Our search resulted in 833 total references, and 45 papers were within our scope of interest. A schematic representation of the current study design is shown in Figure 1.

3. Intestinal Morphology and Redox Biology

As a prerequisite for understanding the role of experimental models, it is important to briefly review the main oxidative stress markers and antioxidant defense mechanisms.

3.1. Intestinal Morphology

The intestinal mucosa comprises a superficial layer of self-renewing epithelial cells and lamina propria with vascular, immune components. In the small intestine there are invaginations called Lieberkuhn’s crypts and protrusions into the lumen called differentiated cell villi. Crypts contain proliferative stem cells and Paneth cells responsible for innate immunity and antibacterial defense and for providing essential signals to intestinal stem cells [5].
The intestinal epithelium is made up of cells with an absorptive function (enterocytes), secretory cells (mucus-secreting goblet cells, Paneth cells-secreting antimicrobial peptides like lysozyme and α defensins, hormone-secreting enteroendocrine cells), and M-microfold cells that facilitate antigen presentation to lymphocytes from lamina propria. Enterocytes represent 80% of all cells, and enteroendocrine cells represent 1% of all epithelial cells. They originate in crypts, migrate to villi during differentiation, and then undergo spontaneous apoptosis and detach when they reach the tip of the villi after terminal differentiation [5].
Immune and inflammatory responses in the intestinal mucosa are marked by significant metabolic shifts in the tissue. These shifts involve high energy consumption and reduced oxygen availability (hypoxia) [6]. Such metabolic changes are driven by the recruitment of inflammatory cells, particularly neutrophils, other polymorphonuclear leukocytes (PMNs), and monocytes. A key feature of acute gut inflammation is the localized buildup of PMNs, known as crypt abscesses. Due to the substantial amounts of ROS generated by activated PMNs, crypt abscesses serve as critical hubs for reduction–oxidation (redox) signaling (a signaling response triggered by specific ROS) [7]. Resident immune cells in the gut, including intraepithelial lymphocytes and antigen-presenting cells (dendritic cells and macrophages), are primed to react to threats like bacterial and viral infections but also play a role in maintaining homeostasis through immune surveillance and promoting regulatory immune responses. Many of these cell types can either activate or bypass redox signaling, which has significant implications for mucosal homeostasis [8].

3.2. Intestinal Antioxidant Defense System

To combat oxidative stress, the body has an antioxidant defense system composed of endogenous enzymatic antioxidants and endogenous non-enzymatic antioxidants. Endogenous enzymatic antioxidants include superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px). Endogenous non-enzymatic antioxidants include glutathione, thioredoxin (Trx), and irisin. Exogenous antioxidants are represented by essential nutrients and nutritional supplements [9]. Table 1 summarizes the mechanisms of action of endo- and exogenous antioxidants.

3.3. Glutathione Synthesis and Its Particular Role in the Gut

The intestinal epithelium, a component of mucosa, has a particular luminal distribution, which makes it a significant protective structure against extraluminal diverse factors. Glutathione, a potent antioxidant abundantly present in intestinal epithelial cells, plays a central role in maintaining mucosal integrity. Glutathione is a tripeptide (g-Glu-Cys-Gly) present in high concentrations in tissues, essential for the proper functioning and maintenance of the structural integrity of the gastrointestinal tract. Epithelial cells in the jejunum and colon are dependent on the presence of GSH, and a deficiency could affect the gastrointestinal tract at this level [3]. The GSH/GSSG (glutathione disulfide) redox couple provides a redox environment that allows maintenance of gut microbiota, adequate nutrient absorption, reversal of oxidant-induced epithelial damage, modulation of intestinal cell transformation, and apoptosis [8].
GSH is essential for the proper functioning and maintenance of the structural integrity of the intestinal tract. Epithelial cells in the jejunum and colon are dependent on the presence of GSH, and a deficiency of GSH could affect the intestinal tract at this level [3].
GSH concentrations are much higher in the gastric glands, conferring protection against the effects of stomach acid. Inflammation induced by Helicobacter pylori infection results in damage due to the production of ROS, exceeding the protective capacity of gastric mucosal cells and glutathione. Regulation of glutathione concentration prevents injury caused by Helicobacter pylori infection, showing the importance of maintaining the relative balance between pro- and antioxidants [25]. Intestinal GSH concentrations are in the millimolar range (2–10 mmol/L). At the intracellular level it is found in the reduced thiol form, and the oxidized form is glutathione disulfide (GSSG) and is associated with oxidative stress [26].
GSH in the intestinal epithelium is synthesized in the cytosol via two ATP-dependent steps: glutamate-cysteine ligase forms γ-glutamyl-cysteine, and glutathione synthetase adds glycine to complete the tripeptide, while glutathione reductase reduces oxidized glutathione back to GSH using NADPH. At the brush border, γ-glutamyl transpeptidase and a dipeptidase degrade luminal GSH into amino acids that enterocytes re-use for GSH synthesis; together, these processes constitute the γ-glutamyl cycle [4].
Diet and bile provide luminal GSH; bile secretion into the duodenum supplies roughly half of hepatic GSH output. Enterocytes absorb intact GSH across the apical membrane through transporters that operate independently of GSH synthesis and are stimulated by monovalent cations. Any luminal GSH not taken up is hydrolyzed by apical γ-glutamyl transpeptidase, even at low concentrations [26].
Within the epithelium, the GSH/GSSG redox couple sustains a reducing environment that supports nutrient absorption, preserves the mucus layer, and modulates proliferation, differentiation, and apoptosis of enterocytes [4]. Luminal GSH reduces dietary disulfides, conjugates xenobiotics, chelates metals, and contributes to mucus fluidity, thereby protecting the mucosal surface [26]. Adequate GSH is particularly important for active calcium transport: depletion of mucosal GSH leads to epithelial degeneration and impaired Ca2+ absorption, whereas oral GSH or its monoester restores both epithelial integrity and transport capacity. GSH depletion or oxidant-rich diets trigger oxidative and nitrosative stress and apoptosis in enterocytes, diminishing nutrient absorption, while antioxidants such as quercetin, naringin, ursodeoxycholic acid, melatonin, and glutamine replenish GSH and normalize these functions [4].
At the brush border of the intestinal epithelium, a cysteine/cystine shuttle works alongside the γ-glutamyl cycle to control luminal thiol/disulfide balance, and within enterocytes, reduction in cystine and continuous GSH synthesis keep both the cysteine/cystine and GSH/GSSG redox couples in the proper range. This finely tuned system enables intestinal epithelial cells to cope with constant exposure to luminal oxidants, maintain barrier integrity, and support efficient nutrient uptake and detoxification [26].

3.4. Redox Signaling in the Gut

The redox state of the intestinal tract is determined by the balance between antioxidants and oxidants. When this balance is disrupted—either by an excess of oxidants or a deficiency of neutralizing antioxidants—the tissue experiences oxidative or nitrosative stress. In the intestinal tract, various ROS, such as superoxide (O2) and hydroxyl radicals (•OH), along with non-radicals like hypochlorous acid (HOCl) and hydrogen peroxide (H2O2), are produced by epithelial cells, endothelial cells, and immune cells to support mucosal defense [27].
The enzymatic use of molecular oxygen (O2) within the intestinal mucosa facilitates redox signaling, creating spatial and dynamic patterns of oxygen availability. In healthy intestinal mucosa, there is a steep oxygen gradient between the well-vascularized mucosa and the anoxic lumen. As a result, cells near crypt stem cells typically experience higher partial pressures of oxygen (pO2; ~100 mmHg) compared to lumen-facing epithelial cells (<10 mmHg) [28].

4. Oxidative Stress Involved in Inflammation-Based Intestinal Diseases

4.1. Collateral Damage to Digestive Tissues Induced by Reactive Oxygen Species

When antioxidant defenses fail, excess ROS damage proteins, lipids, and DNA, impairing intestinal morphology, barrier integrity, metabolism, and immune regulation [2]. Major ROS sources in the intestinal tract include xanthine oxidase, diamine oxidase, aldehyde oxidase, and NADPH oxidase [29]. Excessive ROS from pro-inflammatory cells drive chronic intestinal inflammation, linked to reduced GSH and increased GSSG. Patients with inflammatory bowel disease have lower mucosal GSH and decreased T lymphocyte proliferation in the lamina propria. Bacterial ROS can alter Trx status and inhibit NF-κB activity, while microbiota composition affects pathogen resistance. Chronic oxidative stress and inflammation raise cancer risk, with elevated Trx1 and Grx3 promoting colorectal cancer growth, whereas reduced Grx3 inhibits tumor progression [26].
Tight junction proteins are crucial for maintaining the intestinal barrier and regulating permeability; their disruption leads to gut barrier damage and increased permeability, contributing to diseases like inflammatory bowel disease [30]. Redox balance is vital for intestinal flora, with GSH in the lumen supporting mucus fluidity, detoxifying xenobiotics, and metabolizing peroxidized lipids [31]. Impaired redox balance weakens antimicrobial defenses, prolongs immune activation, and alters immune responses, leading to incomplete pathogen removal and chronic inflammation that can cause complications like fibrosis and neoplasia [1]. Both Crohn’s disease and ulcerative hemorrhagic colitis involve chronic inflammation due to inadequate inflammatory responses in genetically predisposed individuals, marked by elevated ROS and reduced antioxidants. ROS further enhances pro-inflammatory factors, worsening tissue damage and disease progression [25].
Both excess and deficiency of ROS contribute to inflammatory bowel disease (IBD). Genome-wide association studies reveal that risk factors vary among individuals, not solely linked to ulcerative colitis or Crohn’s disease. For instance, altered oxygen levels in chronic granulomatous disease significantly increase IBD risk [32]. Elevated oxidative stress from intestinal inflammation leads to tissue damage, impaired absorption, gut barrier dysfunction, and altered motility [28]. Nutrient malabsorption occurs after ischemia–reperfusion injury and in IBD [33]. Impaired absorption in the colon’s epithelial tissue results in diarrhea [34]. Extensive ROS-induced damage and increased immune mediators like TNF-α and IFN-γ enhance mucosal permeability by affecting tight junctions [35].
Increases in vascular permeability can precede epithelial permeability during active mucosal inflammation. Tolstanova et al. demonstrated that early endothelial injury causes perivascular edema and epithelial hypoxia, stabilizing hypoxia-inducible factors in the mucosa [36]. Additionally, intestinal motility is influenced by redox-sensitive mechanisms; Brown et al. found that glial cell-derived nitric oxide (NO) mediates enteric neuron death during active colitis, impacting connexin activity. Furthermore, exposure to microbial lipopolysaccharides (LPS) impairs motility by generating excess ROS and reactive nitrogen species (RNS) in submucosal smooth muscle cells [37]. These findings highlight the tissue damage linked to oxidative stress during gut inflammation.

4.2. Toxin/Drug Exposure

Oxidative stress plays a role in inflammation-driven intestinal diseases. The involvement of ROS in various GI dysfunctions, as well as their dual roles in cancer promotion and suppression, will be explored to deepen our understanding of inflammation-based GI disorders [38].
Chemotherapeutic agents produce high levels of ROS, which play a significant role in the pathology of drugs like anthracyclines and platinum-based compounds. While these agents use ROS to kill cancer cells, they also cause common side effects, including intestinal toxicity and mutagenesis. Excess ROS can lead to severe issues such as mucosal damage, loss of epithelial cells and tight junction proteins, microbiota imbalance, and enteric neuropathy. A major side effect of chemotherapy is mucositis, which results in significant gastrointestinal inflammation, nausea, diarrhea, bleeding, and abdominal pain [39].
Radiotherapy for abdominal and pelvic cancers often leads to gastrointestinal complications, with oxidative stress being a key mechanism for cancer cell death [40]. Prolonged NSAID use carries a high risk of gastrointestinal issues, including peptic ulcers, as NSAID metabolites may induce ROS generation [41]. Excessive alcohol consumption also contributes to oxidative stress through byproducts of ethanol metabolism and NAD depletion [39].

4.3. Ischemia–Reperfusion and Postoperative Injury

Ischemia–reperfusion injury is a major issue in ischemic syndromes and solid organ transplantation, occurring in various tissues upon reoxygenation. This injury is primarily driven by oxidative stress, which activates an immune response in the affected tissue [42]. The gut is particularly susceptible due to its ability to generate numerous free radicals [39].
Postoperative ileus, or paralytic ileus, is characterized by reduced intestinal activity following bowel manipulation during surgery. Research indicates that it is a complex condition involving initial activation of cells in the muscularis propria [43].

4.4. Congenital Pathologies

Genetic intestinal disorders may be linked to oxidative stress, though the complexity of mutations requires further investigation. For instance, Triple-A syndrome has been noted, and recent reports suggest oxidative stress may play a role in Hirschsprung’s disease [44,45].

4.5. Inflammation and Infection

Oxidative stress is crucial for normal immune functions, as free radicals help eliminate bacteria and diseased cells and activate inflammatory pathways. However, non-specific free radical signaling can cause tissue damage and lead to inflammation, a phenomenon well-studied in IBD [46,47].
Necrotizing enterocolitis (NEC) is a life-threatening intestinal condition that primarily affects premature, low-birth-weight infants. The pathogenesis of NEC is closely linked to tissue oxygenation levels. This multifactorial disorder involves immune stimulation in the preterm gut during colonization by intestinal flora [48].
Oxidative stress also seems to be involved in parasitic infections of the gut, such as Chagas disease which is caused by infection with the parasite Trypanosoma cruzi. In this case, megacolon or megaesophagus only occurs in a limited number of patients [49].

4.6. Cancer

Intestinal inflammation promotes oxidative stress and free radical accumulation, which accelerates carcinogenesis. Consequently, patients with IBD have a markedly increased risk of colorectal cancer [50,51,52]. Similarly, Barrett’s esophagus is linked to oxidative stress, which can lead to esophageal adenocarcinoma [53].
Table 2 summarizes information on oxidative stress in intestinal pathology.

5. Experimental Models of Inflammatory Bowel Diseases

Because oxidative mechanisms in human intestinal pathology remain insufficiently understood, experimental models are essential for clarifying their role.
A key feature is the remarkable diversity of experimental models and species used, reflecting the ongoing search for reliable systems to assess the impact of oxidative stress in the digestive tract.

5.1. Mice

The most used species in experimental models for studying oxidative stress in intestinal system is the mouse.

5.1.1. DSS- and TNBS-Induced Colitis Models

Most patients with chronic granulomatous disease exhibit symptoms characteristic of inflammatory bowel disease. A complicating factor in research in this area is the use of mouse models to differentiate the roles of phagocyte-derived ROS versus those originating from mucosal sources. In studies using 2,4,6-trinitrobenzenesulfonic acid (TNBS) to model colitis, researchers found that Nox2−/− mice developed significantly more severe colitis, as evidenced by weight loss, increased intestinal permeability, and an inability to resolve inflammation, compared to wild-type controls [6]. Conversely, Bao et al. used the same Nox2−/− mice to study dextran sulfate sodium (DSS)-induced colitis and found no difference in weight loss or disease severity compared to wild-type controls, concluding that reduced tissue damage was linked to a diminished oxidative response [65].
One potential explanation for the discrepancies between these studies is the nature of the models used to assess the relative importance of NOX phagocytosis. DSS-induced colitis models involve the denudation of epithelial cells, beginning with apical mucus erosion and epithelial apoptosis, leading to an immune infiltrate. In this context, it could be argued that phagocyte-derived ROS may not significantly contribute to the denudation of colonic epithelia; therefore, only interventions affecting epithelial viability or turnover would have a notable impact. In contrast, TNBS-induced colitis models involve pre-sensitization of the host immune system. DSS results in progressive tissue damage that extends from the rectum and is associated with gradual weight loss. By contrast, TNBS-treated animals exhibit rapid weight fluctuations and discontinuous lesions, often with relatively preserved epithelia. Furthermore, the immune infiltrates and inflammatory mediators differ significantly between the models. Thus, DSS serves as a model of tissue deterioration, whereas TNBS represents a model of acute to chronic inflammation and resolution. Consequently, it is plausible that the Nox2−/− mutation does not lead to increased mucosal damage [66].

5.1.2. Oxazolone-Induced Colitis Model

Oxazolone is a haptenizing agent commonly used to induce colitis in mice for studying the pathological processes involved in ulcerative colitis. This colitis model triggers a Th2 cell-mediated immune response and has been shown to closely resemble human ulcerative colitis, exhibiting mucosal membrane inflammation, epithelial microulcerations, and histopathological changes in the distal colon. The cellular and immune responses, as well as the cytokine secretion profiles in oxazolone-induced colitis, differ from those observed in TNBS colitis. Specifically, oxazolone-induced colitis is marked by significant IL-13 production from natural killer T (NKT) cells in the lamina propria CD4, rather than the typical IFN-γ production from conventional CD4+ T cells [67].

5.1.3. Adoptive T-Cell Transfer Experimental Model

In 1990, an adoptive T-cell transfer system was established to induce colitis in immunodeficient mice, significantly advancing the concept of “regulatory T cells” [68]. A major breakthrough in IBD research occurred in 1993 with the discovery of spontaneous colitis in three types of knockout (KO) mice: interleukin (IL)-2 KO, IL-10 KO, and T-cell receptor (TCR) KO [69]. Since then, over 40 different genetically engineered KO mouse strains and strains with congenital mutations that spontaneously develop colitis and/or ileitis have been identified [67].

5.1.4. IL-10 Knockout Model

The development of murine models to study the pathophysiology of IBD continued with the creation of a genetically engineered model using IL-10-deficient mice [70]. This model underscored the significant anti-inflammatory role of IL-10. Additionally, genetic polymorphisms at the IL-10 locus have been linked to the development of Crohn’s disease (CD) and ulcerative colitis (UC). IL-10 is a well-known regulatory cytokine and a key susceptibility gene for IBD (both UC and CD). IL-10 KO mice, which lack the IL-10 gene, spontaneously develop colitis after three months of age [71].

5.1.5. SAMP1/YitFc Colitis

The SAMP1/YitFc mouse strain serves as a model for CD-like ileitis, making it ideal for investigating the pathogenesis of chronic intestinal inflammation. Unlike most animal models of colitis, the ileal phenotype specific to SAMP1/YitFc mice occurs spontaneously, without any genetic, chemical, or immunological manipulation. Furthermore, SAMP1/YitFc mice exhibit remarkable similarities to human disease in terms of disease localization, histological features, incidence of extraintestinal manifestations, and response to conventional therapies [72].

5.1.6. Microbiome-Induced Mouse Models

Using culture-independent molecular methods, researchers have identified over 1000 species of bacteria residing in the gastrointestinal tract, with the collective genome of gut microbes containing approximately 100 times more genes than the human genome. In patients with IBD, there is a notable anti-inflammatory effect associated with decreased bacterial diversity and an increase in pro-inflammatory bacteria compared to healthy individuals. The most consistent changes include reduced gut microbiota diversity and lower levels of Firmicutes [67]. Table 3 summarizes various experimental models of inflammatory bowel disease in mice.

5.2. Guinea Pigs

Guinea pigs are commonly used to study the enteric nervous system due to their neurochemical similarities to humans with IBD. Their sensitivity to sensitizing substances makes them ideal for immunological research, particularly in DSS-induced colitis. However, their high care costs limit their utility [93].

5.3. The New Zealand White Rabbit

New Zealand white rabbits facilitate endoscopic biopsies due to their larger size. Inducing colitis with DSS allows observation of disease progression from acute to chronic stages. They are recommended for studies requiring endoscopic procedures [93].

5.4. Dogs

Dogs are frequently used in drug discovery and safety evaluation. Canine idiopathic IBD shares similarities with human IBD, involving immune, genetic, and environmental factors. Despite their usefulness as models, the high costs associated with using dogs can be a drawback [94].

5.5. Pigs

Pigs share comparable nutritional and immunological features with humans, making them a promising model for IBD. DSS-induced colitis has been successfully reproduced in young piglets, aiding research on mucosal immunity and dietary effects. Pigs also exhibit chronic inflammation patterns similar to human UC, despite the high costs involved [95,96,97].

5.6. Monkeys

Primates, such as rhesus macaques and cotton-top tamarins, are crucial for biomedical research due to their biological similarity to humans. They often show spontaneous enteritis, resembling human IBD. Studies reveal that many macaques share differentially expressed genes with humans and mice, making them valuable for IBD research, although their limited availability poses challenges [98].

5.7. Zebrafish

Zebrafish, with 87% genetic similarity to humans, are low-cost models for studying IBD. Their transparency allows dynamic observation of disease development. TNBS- and DSS-induced intestinal inflammation in zebrafish reflects changes in gut microbiota and immune responses similar to mammalian IBD. While their differences from humans limit direct applicability, they provide valuable insights into pathogenesis and drug testing [99,100,101,102].
A study conducted by Rastogi et al., showed that the ability of the GSH system to respond and regenerate after oxidation changes with developmental stage. Zebrafish embryos are resistant to oxidative stress 18 h after fertilization. After hatching, embryos become more sensitive to the action of pro-oxidizing factors. This is due to changes in GSH concentration during embryo development [103].
Glutathione is the most abundant and most commonly studied antioxidant in zebrafish when assessing oxidative stress. Oxidative stress results in decreased glutathione concentration. Zebrafish glutathione concentration decreases in response to multiple substances such as arsenic, atrazine, cadmium, chromium, cadmium sulfate, silver nanoparticles, titanium dioxide nanoparticles, bisphenol A, nonylphenol, and ionizing radiation. Glutathione is used in order to neutralize ROS causing an increase in GSSG concentration. Assessment of both GSH and GSSG allows calculation of redox status (ratio of GSSG to GSH) being a biomarker of oxidative stress. There are different concentrations of GSH and GSSG depending on the stage of embryonic development. GSH dynamics are important for maintaining redox homeostasis and suggest that there are stages at which embryos are susceptible to oxidative stress [104].
Many xenobiotic agents can induce oxidative stress, with the risk of overwhelming the body’s antioxidant defense system and damaging proteins, lipids, and genetic material. Zebrafish is a useful experimental model to study the toxicity of different xenobiotic agents and oxidative stress by having an extensive antioxidant system [104].
Zebrafish are extensively utilized as an animal model to investigate human development, genetics, and disease. Recently, they have gained prominence as a model for exploring the regulatory mechanisms of ion homeostasis in body fluids. Hormonal control plays a crucial role in regulating ions within these fluids. Several hormones, such as parathyroid hormone (PTH) and vitamin D, are well known for maintaining Ca2+ homeostasis in mammals. Recent studies have examined the roles of these calciotropic hormones in zebrafish. The expression and secretion of calciotropic hormones in zebrafish are influenced by extracellular Ca2+ levels. The calcium-sensing receptor (CaSR) located in specific endocrine organs detects these extracellular Ca2+ levels and modulates the expression and secretion of mammalian calciotropic hormones. Recent research has also defined the effects of CaSR on hormone regulation in zebrafish [105].

5.8. Drosophila melanogaster

Drosophila melanogaster serves as a valuable model for studying intestinal diseases due to its human-like anatomical features. It helps investigate the regulatory pathways of innate immunity related to IBD, such as the balance between microbe-induced epithelial cell damage and stem cell repair via Toll signaling. This model has also been used to explore the effects of Epstein–Barr virus DNA on IBD. Future research could leverage Drosophila for large-scale genetic screening to uncover processes related to intestinal cell proliferation, differentiation, and function, potentially offering new insights into the mechanisms of intestinal diseases [106,107,108].

5.9. Worms

Caenorhabditis elegans shares significant genomic homology with mammals and is transparent during growth, allowing for easy cell visualization. While it enables the study of bacterial interactions with intestinal cells, its applicability to IBD research is limited due to the absence of immune and circulatory systems. Nonetheless, worms are valuable for understanding inflammation mechanisms in the human gut and screening new drugs for IBD treatment [109,110,111]. Table 4 summarizes different experimental models of inflammatory bowel disease.

6. Conclusions

Oxidative stress is central to the onset and progression of intestinal disease. Experimental models remain indispensable for identifying biomarkers, elucidating mechanisms, and testing potential therapies. Across models, oxidative stress is commonly assessed by the GSH/GSSG redox couple and antioxidant enzymes (SOD, CAT), together with global ROS-related readouts where applicable. Although no single model fully replicates human intestinal diseases, chemically induced colitis models are useful for acute inflammation, genetically modified animals provide insights into chronic mechanisms, and cell culture systems allow mechanistic exploration but lack systemic relevance. The choice of model should depend on the research question. Future studies should standardize redox panels and reporting, validate findings across models/species, and expand human-relevant platforms that better reproduce the complexity of human intestinal pathology.

Author Contributions

Conceptualization, C.D.P. and C.L.Z.; methodology, C.L.Z.; software, I.A.P. and C.G.; validation, C.G. and I.A.P.; formal analysis, C.G.; investigation, C.D.P. and C.G.; resources, C.D.P.; data curation, I.A.P.; writing—original draft preparation, C.D.P.; writing—review and editing, I.A.P.; visualization, C.L.Z.; supervision, C.L.Z.; project administration, I.A.P.; funding acquisition, C.D.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Aviello, G.; Knaus, U.G. ROS in gastrointestinal inflammation: Rescue or Sabotage? Br. J. Pharmacol. 2017, 174, 1704–1718. [Google Scholar] [CrossRef]
  2. Zhang, S. From Challenge to Opportunity: Addressing Oxidative Stress in Animal Husbandry. Antioxidants 2023, 12, 1543. [Google Scholar] [CrossRef] [PubMed]
  3. Balmus, M.I.; Ciobica, A.; Trifan, A.; Stanciu, C. The implications of oxidative stress and antioxidant therapies in Inflammatory Bowel Disease: Clinical aspects and experimental models. Saudi J. Gastroenterol. 2016, 22, 3–17. [Google Scholar]
  4. Razvani, M. Oxidative Stress induced Gastrointestinal Disease: Biology and Nanomedicine—A Review. BioChem 2024, 4, 189–216. [Google Scholar] [CrossRef]
  5. Moine, L.; Rivoira, M.; Díaz de Barboza, G.; Pérez, A.; Tolosa de Talamoni, N. Glutathione depleting drugs, antioxidants and intestinal calcium absorption. World J. Gastroenterol. 2018, 24, 4979–4988. [Google Scholar] [CrossRef] [PubMed]
  6. Taylor, C.T.; Colgan, S.P. Regulation of immunity and inflammation by hypoxia in immunological niches. Nat. Rev. Immunol. 2017, 17, 774–785. [Google Scholar] [CrossRef] [PubMed]
  7. Campbell, E.L.; Bruyninckx, W.J.; Kelly, C.J.; Glover, L.E.; McNamee, E.N.; Bowers, B.E.; Bayless, A.J.; Scully, M.; Saeedi, B.J.; Golden-Mason, L.; et al. Transmigrating neutrophils shape the mucosal microenvironment through localized oxygen depletion to influence resolution of inflammation. Immunity 2014, 40, 66–77. [Google Scholar] [CrossRef]
  8. Chang, S.Y.; Ko, H.J.; Kweon, M.N. Mucosal dendritic cells shape mucosal immunity. Exp. Mol. Med. 2014, 46, e84. [Google Scholar] [CrossRef]
  9. Yajie, W.; Yue, C.; Xiaoyu, Z. New insights in intestinal oxidative stress damage and the health intervention effects of nutrients: A review. J. Funct. Foods 2020, 75, 104248. [Google Scholar] [CrossRef]
  10. Rampon, C.; Volovitch, M.; Joliot, A.; Vriz, S. Hydrogen Peroxide and Redox Regulation of Developments. Antioxidants 2018, 7, 159. [Google Scholar] [CrossRef]
  11. Liang, X.; Wen, K.; Chen, Y.; Fang, G.; Yang, S.; Li, Q. Oral Administration of Therapeutic Enzyme Capsule for the Management of Inflammatory Bowel Disease. Int. J. Nanomed. 2022, 17, 4843–4860. [Google Scholar] [CrossRef]
  12. Vašková, J.; Kočan, L.; Vaško, L.; Perjési, P. Glutathione-Related Enzymes and Proteins: A Review. Molecules 2023, 28, 1447. [Google Scholar] [CrossRef] [PubMed]
  13. Morris, G.; Gevezova, M.; Sarafian, V.; Maes, M. Redox regulation of the immune response. Cell. Mol. Immunol. 2022, 19, 1079–1101. [Google Scholar] [CrossRef]
  14. Iborra, M.; Moret, I.; Busó, E.; García-Giménez, J.L.; Ricart, E.; Gisbert, J.P.; Cabré, E.; Esteve, M.; Márquez-Mosquera, L.; García-Planella, E. The Genetic Diversity and Dysfunctionality of Catalase Associated with a Worse Outcome in Crohn’s Disease. Int. J. Mol. Sci. 2022, 23, 15881. [Google Scholar] [CrossRef] [PubMed]
  15. Elste, V.; Troesch, B.; Eggersdorfer, M.; Weber, P. Emerging Evidence on Neutrophil Motility Supporting Its Usefulness to Define Vitamin C Intake Requirements. Nutrients 2017, 9, 503. [Google Scholar] [CrossRef]
  16. Rizvi, S.; Raza, S.T.; Ahmed, F.; Ahmad, A.; Abbas, S.; Mahdi, F. The role of vitamin e in human health and some diseases. Sultan Qaboos Univ. Med. J. 2014, 14, e157–e165. [Google Scholar] [CrossRef]
  17. Gammoh, N.; Rink, L. Zinc in Infection and Inflammation. Nutrients 2017, 9, 624. [Google Scholar] [CrossRef]
  18. Avery, J.; Hoffmann, P. Selenium, Selenoproteins, and Immunity. Nutrients 2018, 10, 1203. [Google Scholar] [CrossRef]
  19. Miazek, K.; Beton, K.; Śliwińska, A.; Brożek-Płuska, B. The Effect of β-Carotene, Tocopherols and Ascorbic Acid as Anti-Oxidant Molecules on Human and Animal In Vitro/In Vivo Studies: A Review of Research Design and Analytical Techniques Used. Biomolecules 2022, 12, 1087. [Google Scholar] [CrossRef] [PubMed]
  20. Cheng, J.; Balbuena, E.; Miller, B.; Eroglu, A. The Role of β-Carotene in Colonic Inflammation and Intestinal Barrier Integrity. Front. Nutr. 2021, 27, 723480. [Google Scholar] [CrossRef]
  21. Li, G.; Ding, K.; Qiao, Y.; Zhang, L.; Zheng, L.; Pan, T.; Zhang, L. Flavonoids Regulate Inflammation and Oxidative Stress in Cancer. Molecules 2020, 25, 5628. [Google Scholar] [CrossRef]
  22. Slika, H.; Mansour, H.; Wehbe, N.; Nasser, S.A.; Iratni, R.; Nasrallah, G.; Shaito, A.; Ghaddar, T.; Kobeissy, F.; Eid, A.H. Therapeutic potential of flavonoids in cancer: ROS-mediated mechanisms. Biomed. Pharmacother. 2022, 146, 112442. [Google Scholar] [CrossRef] [PubMed]
  23. Zhao, J.; Zhao, F.; Yuan, J.; Liu, H.; Wang, Y. Gut microbiota metabolites, redox status, and the related regulatory effects of probiotics. Heliyon 2023, 9, 11. [Google Scholar] [CrossRef] [PubMed]
  24. Loveikyte, R.; Bourgonje, A.R.; van Goor, H.; Dijkstra, G.; van der Meulen-de Jong, A.E. The effect of iron therapy on oxidative stress and intestinal microbiota in inflammatory bowel diseases: A review on the conundrum. Redox Biol. 2023, 68, 102950. [Google Scholar] [CrossRef] [PubMed]
  25. 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]
  26. Circu, M.L.; Aw, T.Y. Intestinal redox biology and oxidative stress. Semin. Cell Dev. Biol. 2012, 23, 729–737. [Google Scholar] [CrossRef]
  27. Bedard, K.; Krause, K.H. The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology. Physiol. Rev. 2007, 87, 245–313. [Google Scholar] [CrossRef]
  28. Campbell, E.L.; Colgan, S.P. Control and dysregulation of redox signalling in the gastrointestinal tract. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 106–120. [Google Scholar]
  29. Patlevič, P.; Vašková, J.; Švorc, P., Jr.; Vaško, L.; Švorc, P. Reactive oxygen species and antioxidant defense in human gastrointestinal diseases. Integr. Med. Res. 2016, 5, 250–258. [Google Scholar] [CrossRef]
  30. Xiang, X.; Wang, H.; Zhou, W.; Wang, C.; Guan, P.; Xu, G.; Zhao, Q.; He, L.; Yin, Y.; Li, T. Glutathione Protects against Paraquat-Induced Oxidative Stress by Regulating Intestinal Barrier, Antioxidant Capacity, and CAR Signaling Pathway in Weaned Piglets. Nutrients 2022, 15, 198. [Google Scholar] [CrossRef]
  31. Stojiljković, V.; Pejić, S.; Kasapović, J.; Gavrilović, L.; Stojiljković, S.; Nikolić, D.; Pajović, S.B. Glutathione redox cycle in small intestinal mucosa and peripheral blood of pediatric celiac disease patients. An. Acad. Bras. Cienc. 2012, 84, 175–184. [Google Scholar]
  32. Huang, C.; De Ravin, S.S.; Paul, A.R.; Heller, T.; Ho, N.; Wu Datta, L.; Zerbe, C.S.; Marciano, B.E.; Kuhns, D.B.; Kader, H.A.; et al. Genetic Risk for Inflammatory Bowel Disease Is a Determinant of Crohn’s Disease Development in Chronic Granulomatous Disease. Inflamm. Bowel Dis. 2016, 22, 2794–2801. [Google Scholar]
  33. Hebuterne, X.; Filippi, J.; Schneider, S.M. Nutrition in adult patients with inflammatory bowel disease. Curr. Drug Targets 2014, 15, 1030–1038. [Google Scholar] [CrossRef]
  34. Hering, N.A.; Fromm, M.; Schulzke, J.D. Determinants of colonic barrier function in inflammatory bowel disease and potential therapeutics. J. Physiol. 2012, 590, 1035–1044. [Google Scholar] [CrossRef] [PubMed]
  35. Capaldo, C.T.; Nusrat, A. Cytokine regulation of tight junctions. Biochim. Biophys. Acta 2009, 1788, 864–871. [Google Scholar]
  36. Tolstanova, G.; Deng, X.; French, S.W.; Lungo, W.; Paunovic, B.; Khomenko, T.; Ahluwalia, A.; Kaplan, T.; Dacosta-Iyer, M.; Tarnawski, A.; et al. Early endothelial damage and increased colonic vascular permeability in the development of experimental ulcerative colitis in rats and mice. Lab. Investig. 2012, 92, 9–21. [Google Scholar]
  37. Brown, I.A.; McClain, J.L.; Watson, R.E.; Patel, B.A.; Gulbransen, B.D. Enteric glia mediate neuron death in colitis through purinergic pathways that require connexin-43 and nitric oxide. Cell. Mol. Gastroenterol. Hepatol. 2016, 2, 77–91. [Google Scholar] [CrossRef]
  38. Kim, Y.J.; Kim, E.H.; Hahm, K.B. Oxidative stress in inflammation-based gastrointestinal tract diseases: Challenges and opportunities. J. Gastroenterol. Hepatol. 2012, 27, 1004–1010. [Google Scholar] [CrossRef]
  39. Stavely, R.; Ott, L.C.; Rashidi, N.; Sakkal, S.; Nurgali, K. The Oxidative Stress and Nervous Distress Connection in Gastrointestinal Disorders. Biomolecules 2023, 13, 1586. [Google Scholar] [CrossRef] [PubMed]
  40. Liu, R.; Bian, Y.; Liu, L.; Liu, L.; Liu, X.; Ma, S. Molecular pathways associated with oxidative stress and their potential applications in radiotherapy. Int. J. Mol. Med. 2022, 49, 65. [Google Scholar] [CrossRef] [PubMed]
  41. Galati, G.; Tafazoli, S.; Sabzevari, O.; Chan, T.S.; O’Brien, P.J. Idiosyncratic NSAID drug induced oxidative stress. Chem.-Biol. Interact. 2002, 142, 25–41. [Google Scholar] [CrossRef]
  42. De Vries, D.K.; Kortekaas, K.A.; Tsikas, D.; Wijermars, L.G.; van Noorden, C.J.; Suchy, M.T.; Cobbaert, C.M.; Klautz, R.J.; Schaapherder, A.F.; Lindeman, J.H. Oxidative damage in clinical ischemia/reperfusion injury: A reappraisal. Antioxid. Redox Signal. 2013, 19, 535–545. [Google Scholar] [CrossRef]
  43. Bauer, A.J.; Boeckxstaens, G.E. Mechanisms of postoperative ileus. Neurogastroenterol. Motil. 2004, 16, 54–60. [Google Scholar] [CrossRef]
  44. Prasad, R.; Kowalczyk, J.; Meimaridou, E.; Storr, H.; Metherell, L. Oxidative stress and adrenocortical insufficiency. J. Endocrinol. 2014, 221, 63. [Google Scholar] [CrossRef]
  45. Zhou, L.; Wang, B.; Xie, H.; Du, C.; Tang, J.; Tang, W. Intrauterine exposure to oxidative stress induces caspase-1-dependent enteric nerve cell pyroptosis. Pediatr. Surg. Int. 2022, 38, 1555–1567. [Google Scholar] [CrossRef]
  46. Torres, J.; Mehandru, S.; Colombel, J.-F.; Peyrin-Biroulet, L. Crohn’s Disease. Lancet 2017, 389, 1741–1755. [Google Scholar] [CrossRef]
  47. Ungaro, R.; Mehandru, S.; Allen, P.B.; Peyrin-Biroulet, L.; Colombel, J.-F. Ulcerative Colitis. Lancet 2017, 389, 1756–1770. [Google Scholar] [CrossRef]
  48. Hackam, D.J.; Sodhi, C.P.; Good, M. New insights into necrotizing enterocolitis: From laboratory observation to personalized prevention and treatment. J. Pediatr. Surg. 2019, 54, 398–404. [Google Scholar] [CrossRef]
  49. Silva, K.D.A.; Nunes, J.P.S.; Andrieux, P.; Brochet, P.; Almeida, R.R.; Kuramoto Takara, A.C.K.; Pereira, N.B.; Abel, L.; Cobat, A.; Zaniratto, R.C.F. Chagas Disease Megaesophagus Patients Carrying Variant MRPS18B P260A Display Nitro-Oxidative Stress and Mitochondrial Dysfunction in Response to IFN-γ; Stimulus. Biomedicines 2022, 10, 2215. [Google Scholar] [CrossRef]
  50. Weimers, P.; Munkholm, P. The Natural History of IBD: Lessons Learned. Curr. Treat. Options Gastroenterol. 2018, 16, 101–111. [Google Scholar] [CrossRef]
  51. Yalchin, M.; Baker, A.M.; Graham, T.A.; Hart, A. Predicting Colorectal Cancer Occurrence in IBD. Cancers 2021, 13, 2908. [Google Scholar] [CrossRef]
  52. Ullman, T.A.; Itzkowitz, S.H. Intestinal inflammation and cancer. Gastroenterology 2011, 140, 1807–1816. [Google Scholar] [CrossRef]
  53. Peng, D.; Zaika, A.; Que, J.; El-Rifai, W. The antioxidant response in Barrett’s tumorigenesis: A double-edged sword. Redox Biol. 2021, 41, 101894. [Google Scholar] [CrossRef]
  54. McQuade, R.M.; Carbone, S.E.; Stojanovska, V.; Rahman, A.; Gwynne, R.M.; Robinson, A.M.; Goodman, C.A.; Bornstein, J.C.; Nurgali, K. Role of oxidative stress in oxaliplatin-induced enteric neuropathy and colonic dysmotility in mice. Br. J. Pharmacol. 2016, 173, 3502–3521. [Google Scholar] [CrossRef]
  55. Al-Asmari, A.K.; Khan, A.Q.; Al-Asmari, S.A.; Al-Rawi, A.; Al-Omani, S. Alleviation of 5-fluorouracil-induced intestinal mucositis in rats by vitamin E via targeting oxidative stress and inflammatory markers. J. Complement. Integr. Med. 2016, 13, 377–385. [Google Scholar] [CrossRef]
  56. Musa, A.E.; Shabeeb, D.; Alhilfi, H.S.Q. Protective effect of melatonin against radiotherapy-induced small intestinal oxidative stress: Biochemical evaluation. Medicina 2019, 55, 308. [Google Scholar] [CrossRef]
  57. Nagano, Y.; Matsui, H.; Shimokawa, O.; Hirayama, A.; Tamura, M.; Nakamura, Y.; Kaneko, T.; Rai, K.; Indo, H.; Majima, H. Rebamipide attenuates nonsteroidal anti-inflammatory drugs (NSAID) induced lipid peroxidation by the manganese superoxide dismutase (MnSOD) overexpression in gastrointestinal epithelial cells. J. Physiol. Pharmacol. 2012, 63, 137. [Google Scholar]
  58. Samak, G.; Gangwar, R.; Meena, A.S.; Rao, R.G.; Shukla, P.K.; Manda, B.; Narayanan, D.; Jaggar, J.H.; Rao, R. Calcium Channels and Oxidative Stress Mediate a Synergistic Disruption of Tight Junctions by Ethanol and Acetaldehyde in Caco-2 Cell Monolayers. Sci. Rep. 2016, 6, 38899. [Google Scholar] [CrossRef]
  59. Sasaki, M.; Joh, T. Oxidative stress and ischemia-reperfusion injury in gastrointestinal tract and antioxidant, protective agents. J. Clin. Biochem. Nutr. 2007, 40, 1–12. [Google Scholar] [CrossRef]
  60. Matsumoto, K.; Kawanaka, H.; Hori, M.; Kusamori, K.; Utsumi, D.; Tsukahara, T.; Amagase, K.; Horie, S.; Yamamoto, A.; Ozaki, H. Role of transient receptor potential melastatin 2 in surgical inflammation and dysmotility in a mouse model of postoperative ileus. Am. J. Physiol.-Gastrointest. Liver Physiol. 2018, 315, G104–G116. [Google Scholar] [CrossRef]
  61. Sahakian, L.; Filippone, R.T.; Stavely, R.; Robinson, A.M.; Yan, X.S.; Abalo, R.; Eri, R.; Bornstein, J.C.; Kelley, M.R.; Nurgali, K. Inhibition of APE1/Ref-1 redox signaling alleviates intestinal dysfunction and damage to myenteric neurons in a mouse model of spontaneous chronic colitis. Inflamm. Bowel Dis. 2021, 27, 388–406. [Google Scholar]
  62. Aydemir, C.; Dilli, D.; Uras, N.; Ulu, H.O.; Oguz, S.S.; Erdeve, O.; Dilmen, U. Total oxidant status and oxidative stress are increased in infants with necrotizing enterocolitis. J. Pediatr. Surg. 2011, 46, 2096–2100. [Google Scholar] [CrossRef]
  63. Carini, F.; Mazzola, M.; Rappa, F.; Jurjus, A.; Geagea, A.G.; Al Kattar, S.; Bou-Assi, T.; Jurjus, R.; Damiani, P.; Leone, A. Colorectal carcinogenesis: Role of oxidative stress and antioxidants. Anticancer Res. 2017, 37, 4759–4766. [Google Scholar]
  64. Jiménez, P.; Piazuelo, E.; Sánchez, M.T.; Ortego, J.; Soteras, F.; Lanas, A. Free radicals and antioxidant systems in reflux esophagitis and Barrett’s esophagus. World J. Gastroenterol. 2005, 11, 2697–2703. [Google Scholar]
  65. Bao, S.; Carr, E.D.; Xu, Y.H.; Hunt, N.H. Gp91(phox) contributes to the development of experimental inflammatory bowel disease. Immunol. Cell Biol. 2011, 89, 853–860. [Google Scholar] [PubMed]
  66. Strober, W.; Fuss, I.J.; Blumberg, R.S. The immunology of mucosal models of inflammation. Annu. Rev. Immunol. 2002, 20, 495–549. [Google Scholar] [CrossRef]
  67. Baydi, Z.; Limami, Y.; Khalki, L.; Zaid, N.; Naya, A.; Mtairag, E.M.; Oudghiri, M.; Zaid, Y. An Update of Research Animal Models of Inflammatory Bowel Disease. Sci. World J. 2021, 2021, 7479540. [Google Scholar] [CrossRef]
  68. Powrie, F.; Mason, D. OX-22high CD4+ T cells induce wasting disease with multiple organ pathology: Prevention by the OX-22low subset. J. Exp. Med. 1990, 172, 1701–1708. [Google Scholar]
  69. Mombaerts, P.; Mizoguchi, E.; Grusby, M.J.; Glimcher, L.H.; Bhan, A.K.; Tonegawa, S. Spontaneous development of inflammatory bowel disease in T cell receptor mutant mice. Cell 1993, 75, 274–282. [Google Scholar] [CrossRef]
  70. Kühn, R.; Löhler, J.; Rennick, D.; Rajewsky, K.; Müller, W. Interleukin-10-deficient mice develop chronic enterocolitis. Cell 1993, 75, 263–274. [Google Scholar] [CrossRef]
  71. Khor, B.; Gardet, A.; Xavier, R.J. Genetics and pathogenesis of inflammatory bowel disease. Nature 2011, 474, 307–317. [Google Scholar] [CrossRef]
  72. Pizarro, T.T.; Arseneau, K.O.; Cominelli, F. Lessons from genetically engineered animal models XI. Novel mouse models to study pathogenic mechanisms of Crohn’s disease. Am. J. Physiol. Gastrointest. Liver Physiol. 2000, 278, G665–G669. [Google Scholar] [CrossRef] [PubMed]
  73. Chassaing, B.; Aitken, J.D.; Malleshappa, M.; Vijay-Kumar, M. Dextran sulfate sodium (DSS)-induced colitis in mice. Curr. Protoc. Immunol. 2014, 104, 15–25. [Google Scholar] [CrossRef]
  74. da Silva, M.S.; Sánchez-Fidalgo, S.; Talero, E.; Cárdeno, A.; da Silva, M.A.; Villegas, W.; Souza Brito, A.R.; de La Lastra, C.A. Anti-inflammatory intestinal activity of Abarema cochliacarpos (Gomes) Barneby & Grimes in TNBS colitis model. J. Ethnopharmacol. 2010, 128, 467–475. [Google Scholar] [CrossRef] [PubMed]
  75. Kojima, R.; Kuroda, S.; Ohkishi, T.; Nakamaru, K.; Hatakeyama, S. Oxazolone-induced colitis in BALB/C mice: A new method to evaluate the efficacy of therapeutic agents for ulcerative colitis. J. Pharmacol. Sci. 2004, 96, 307–313. [Google Scholar] [CrossRef]
  76. Corazza, N.; Eichenberger, S.; Eugster, H.P.; Mueller, C. Nonlymphocyte-derived tumor necrosis factor is required for induction of colitis in recombination activating gene (RAG)2(−/−) mice upon transfer of CD4(+)CD45RB(hi) T cells. J. Exp. Med. 1999, 190, 1479–1492. [Google Scholar]
  77. Datta, K.; Suman, S.; Kallakury, B.V.S.; Fornace, A.J., Jr. Exposure to Heavy Ion Radiation Induces Persistent Oxidative Stress in Mouse Intestine. PLoS ONE 2012, 7, e42224. [Google Scholar] [CrossRef]
  78. Kosiewicz, M.M.; Nast, C.C.; Krishnan, A.; Rivera-Nieves, J.; Moskaluk, C.A.; Matsumoto, S.; Kozaiwa, K.; Cominelli, F. Th1-type responses mediate spontaneous ileitis in a novel murine model of Crohn’s disease. J. Clin. Investig. 2001, 107, 695–702. [Google Scholar] [PubMed]
  79. Crespo, I.; San-Miguel, B.; Mauriz, J.L.; Ortiz de Urbina, J.J.; Almar, M.; Tuñón, M.J.; González-Gallego, J. Protective Effect of Protocatechuic Acid on TNBS-Induced Colitis in Mice Is Associated with Modulation of the SphK/S1P Signaling Pathway. Nutrients 2017, 9, 288. [Google Scholar] [CrossRef]
  80. Nemoto, Y.; Kanai, T.; Makita, S. Bone marrow retaining colitogenic CD4+ T cells may be a pathogenic reservoir for chronic colitis. Gastroenterology 2007, 132, 176–189. [Google Scholar] [CrossRef]
  81. Nemoto, Y.; Kanai, T.; Kameyama, K. Long-lived colitogenic CD4+ memory T cells residing outside the intestine participate in the perpetuation of chronic colitis. J. Immunol. 2009, 183, 5059–5068. [Google Scholar] [PubMed]
  82. Nemoto, Y.; Kanai, T.; Shinohara, T. Luminal CD4+ T cells penetrate gut epithelial monolayers and egress from lamina propria to blood circulation. Gastroenterology 2011, 141, 2130–2139. [Google Scholar] [CrossRef] [PubMed]
  83. Nemoto, Y.; Kanai, T.; Takahara, M. Bone marrow-mesenchymal stem cells are a major source of interleukin-7 and sustain colitis by forming the niche for colitogenic CD4 memory T cells. Gut 2013, 62, 1142–1152. [Google Scholar] [CrossRef]
  84. Pickert, G.; Neufert, C.; Leppkes, M. STAT3 links IL-22 signaling in intestinal epithelial cells to mucosal wound healing. J. Exp. Med. 2009, 206, 1465–1472. [Google Scholar] [CrossRef] [PubMed]
  85. Kaser, A.; Lee, A.H.; Franke, A. XBP1 links ER stress to intestinal inflammation and confers genetic risk for human inflammatory bowel disease. Cell 2008, 134, 743–756. [Google Scholar] [CrossRef]
  86. Adolph, T.E.; Tomczak, M.F.; Niederreiter, L. Paneth cells as a site of origin for intestinal inflammation. Nature 2013, 503, 272–276. [Google Scholar] [CrossRef]
  87. Garrett, W.S.; Lord, G.M.; Punit, S. Communicable ulcerative colitis induced by T-bet deficiency in the innate immune system. Cell 2007, 131, 33–45. [Google Scholar] [CrossRef]
  88. Panwala, C.M.; Jones, J.C.; Viney, J.L. A novel model of inflammatory bowel disease: Mice deficient for the multiple drug resistance gene, mdr1a, spontaneously develop colitis. J. Immunol. 1998, 161, 5733–5744. [Google Scholar] [CrossRef]
  89. Resta-Lenert, S.; Smitham, J.; Barrett, K.E. Epithelial dysfunction associated with the development of colitis in conventionally housed mdr1a−/− mice. Am. J. Physiol. Gastrointest. Liver Physiol. 2005, 289, G153–G162. [Google Scholar]
  90. Collett, A.; Tanianis-Hughes, J.; Carlson, G.L. Comparison of P-glycoprotein-mediated drug-digoxin interactions in Caco-2 with human and rodent intestine: Relevance to in vivo prediction. Eur. J. Pharm. Sci. 2005, 26, 386–393. [Google Scholar]
  91. Velcich, A.; Yang, W.; Heyer, J. Colorectal cancer in mice genetically deficient in the mucin Muc2. Science 2002, 295, 1726–1729. [Google Scholar] [CrossRef] [PubMed]
  92. Van der Sluis, M.; De Koning, B.A. Muc2-deficient mice spontaneously develop colitis, indicating that MUC2 is critical for colonic protection. Gastroenterology 2006, 131, 117–129. [Google Scholar] [CrossRef]
  93. Wen, C.; Chen, D.; Zhong, R.; Peng, X. Animal models of inflammatory bowel disease: Category and evaluation indexes. Gastroenterol. Rep. 2024, 12, goae021. [Google Scholar] [CrossRef] [PubMed]
  94. Vázquez-Baeza, Y.; Hyde, E.R.; Suchodolski, J.S.; Knight, R. Dog and human inflammatory bowel disease rely on overlapping yet distinct dysbiosis networks. Nat. Microbiol. 2016, 1, 16177. [Google Scholar] [CrossRef]
  95. Nielsen, T.S.; Fredborg, M.; Theil, P.K.; Yue, Y.; Bruhn, L.V.; Andersen, V.; Purup, S. Dietary Red Meat Adversely Affects Disease Severity in a Pig Model of DSS-Induced Colitis Despite Reduction in Colonic Pro-Inflammatory Gene Expression. Nutrients 2020, 9, 1728. [Google Scholar] [CrossRef]
  96. Pistol, G.C.; Bulgaru, C.V.; Marin, D.E.; Oancea, A.G.; Taranu, I. Dietary Grape Seed Meal Bioactive Compounds Alleviate Epithelial Dysfunctions and Attenuates Inflammation in Colon of DSS-Treated Piglets. Foods 2021, 10, 530. [Google Scholar] [CrossRef]
  97. Roediger, W.E.W. Causation of human ulcerative colitis: A lead from an animal model that mirrors human disease. JGH Open 2019, 3, 277–280. [Google Scholar] [CrossRef]
  98. Wang, J.; Lv, M.; He, L.; Wang, X.; Lan, Y.; Chen, J.; Chen, M.; Zhang, C.; Tang, R.; Zhou, D.; et al. Transcriptomic landscape of persistent diarrhoea in rhesus macaques and comparison with humans and mouse models with inflammatory bowel disease. Gene 2021, 800, 145837. [Google Scholar] [CrossRef]
  99. Flores, E.M.; Nguyen, A.T.; Odem, M.A.; Eisenhoffer, G.T.; Krachler, A.M. The zebrafish as a model for gastrointestinal tract-microbe interactions. Cell. Microbiol. 2020, 22, e13152. [Google Scholar] [CrossRef] [PubMed]
  100. Uyttebroek, L.; Pype, C.; Hubens, G.; Timmermans, J.P.; Van Nassauw, L. Effect of TNBS-induced colitis on enteric neuronal subpopulations in adult zebrafish. Eur. J. Histochem. 2020, 64, 3161. [Google Scholar] [CrossRef]
  101. Di Paola, D.; Natale, S.; Iaria, C.; Cordaro, M.; Crupi, R.; Siracusa, R.; D’Amico, R.; Fusco, R.; Impellizzeri, D.; Cuzzocrea, S.; et al. Intestinal Disorder in Zebrafish Larvae (Danio rerio): The Protective Action of N-Palmitoylethanolamide-oxazoline. Life 2022, 12, 125. [Google Scholar] [CrossRef] [PubMed]
  102. Hanyang, L.; Xuanzhe, L.; Xuyang, C.; Yujia, Q.; Jiarong, F.; Jun, S.; Zhihua, R. Application of Zebrafish Models in Inflammatory Bowel Disease. Front. Immunol. 2017, 8, 501. [Google Scholar] [CrossRef] [PubMed]
  103. Rastogi, A.; Clark, C.W.; Conlin, S.M.; Brown, S.E.; Timme-Laragy, A.R. Mapping glutathione utilization in the developing zebrafish (Danio rerio) embryo. Redox Biol. 2019, 26, 101235. [Google Scholar] [CrossRef]
  104. Massarsky, A.; Kozal, J.S.; Di Giulio, R.T. Glutathione and zebrafish: Old assays to address a current issue. Chemosphere 2017, 168, 707–715. [Google Scholar] [CrossRef]
  105. Lin, C.H.; Hwang, P.P. The Control of Calcium Metabolism in Zebrafish (Danio rerio). Int. J. Mol. Sci. 2016, 17, 1783. [Google Scholar] [CrossRef]
  106. Lin, J.; Hackam, D.J. Worms, flies and four-legged friends: The applicability of biological models to the understanding of intestinal inflammatory diseases. Dis. Model. Mech. 2011, 4, 447–456. [Google Scholar] [CrossRef]
  107. Buchon, N.; Broderick, N.A.; Poidevin, M.; Pradervand, S.; Lemaitre, B. Drosophila intestinal response to bacterial infection: Activation of host defense and stem cell proliferation. Cell Host Microbe 2009, 5, 200–211. [Google Scholar] [CrossRef] [PubMed]
  108. Madi, J.R.; Outa, A.A.; Ghannam, M.; Hussein, H.M.; Shehab, M.; Hasan, Z.A.K.H.; Fayad, A.A.; Shirinian, M.; Rahal, E.A. Drosophila melanogaster as a Model System to Assess the Effect of Epstein-Barr Virus DNA on Inflammatory Gut Diseases. Front. Immunol. 2021, 12, 586930. [Google Scholar] [CrossRef]
  109. Haerty, W.; Artieri, C.; Khezri, N.; Singh, R.S.; Gupta, B.P. Comparative analysis of function and interaction of transcription factors in nematodes: Extensive conservation of orthology coupled to rapid sequence evolution. BMC Genom. 2008, 9, 399. [Google Scholar] [CrossRef]
  110. Aboobaker, A.A.; Blaxter, M.L. Medical significance of Caenorhabditis elegans. Ann. Med. 2000, 32, 23–30. [Google Scholar] [CrossRef]
  111. Heylen, M.; Ruyssers, N.E.; Gielis, E.M.; Vanhomwegen, E.; Pelckmans, P.A.; Moreels, T.G.; De Man, J.G.; De Winter, B.Y. Of worms, mice and man: An overview of experimental and clinical helminth-based therapy for inflammatory bowel disease. Pharmacol. Ther. 2014, 143, 153–167. [Google Scholar] [CrossRef]
  112. Sharma, M.; Sharma, S.; Wadhwa, J. Improved uptake and therapeutic intervention of curcumin via designing binary lipid nanoparticulate formulation for oral delivery in inflammatory bowel disorder. Artif. Cells Nanomed. Biotechnol. 2019, 47, 45–55. [Google Scholar] [CrossRef] [PubMed]
  113. Li, S.; Wang, C.; Zhang, C.; Luo, Y.; Cheng, Q.; Yu, L.; Sun, Z. Evaluation of the Effects of Different Bacteroides vulgatus Strains against DSS-Induced Colitis. J. Immunol. Res. 2021, 2021, 9117805. [Google Scholar] [CrossRef]
  114. Lei, H.; Johnson, L.A.; Eaton, K.A.; Liu, S.; Ni, J.; Wang, X.; Higgins, P.D.R.; Xu, G. Characterizing intestinal strictures of Crohn’s disease in vivo by endoscopic photoacoustic imaging. Biomed. Opt. Express 2019, 10, 2542–2555. [Google Scholar] [CrossRef]
  115. Otoni, C.C.; Heilmann, R.M.; García-Sancho, M.; Sainz, A.; Ackermann, M.R.; Suchodolski, J.S.; Steiner, J.M.; Jergens, A.E. Serologic and fecal markers to predict response to induction therapy in dogs with idiopathic inflammatory bowel disease. J. Vet. Intern. Med. 2018, 32, 999–1008. [Google Scholar] [CrossRef] [PubMed]
  116. Li, Y.; Liu, X.J.; Su, S.L.; Yan, H.; Guo, S.; Qian, D.W.; Duan, J.A. Evaluation of Anti-Inflammatory and Antioxidant Effectsof Chrysanthemum Stem and Leaf Extract on Zebrafish Inflammatory Bowel Disease Model. Molecules 2022, 27, 2114. [Google Scholar] [CrossRef] [PubMed]
Figure 1. A schematic representation of the current study design.
Figure 1. A schematic representation of the current study design.
Jcm 14 06569 g001
Table 1. Published studies that analyze the mechanism of actions of endogenous and exogenous antioxidants.
Table 1. Published studies that analyze the mechanism of actions of endogenous and exogenous antioxidants.
SubstanceMechanism of ActionAuthor and Year
Superoxide dismutaseSuperoxide dismutase converts superoxide radicals to hydrogen peroxide and molecular oxygen, reducing oxidative stress.Rampon et al., 2018 [10]
Superoxide dismutase Superoxide dismutase inhibits the production of pro-inflammatory cytokines, reducing colitis symptoms.Liang et al., 2022 [11]
Glutathione (GSH)Glutathione protects against reactive oxygen and nitrogen species, playing a crucial role in maintaining redox homeostasis and regulating immune responses.Vašková et al., 2023 [12]
Glutathione peroxidase (GPx)Glutathione peroxidase regulates Th17 lymphocyte differentiation, promoting immune regulation and antioxidant defenses.Morris et al., 2022 [13]
CatalaseLow catalase levels reduce autophagy-dependent cell death, which may exacerbate conditions like Crohn’s disease.Iborra et al., 2022 [14]
Vitamin CVitamin C improves neutrophil chemokinesis and chemotaxis, enhancing immune response.Elste et al., 2017 [15]
Vitamin EVitamin E inhibits reactive oxygen species production during fat oxidation and free radical reactions.Rizvi et al., 2014 [16]
ZincZinc deficiency increases pro-inflammatory cytokines like TNF-α and IL-6, impacting immune function.Gammoh et al., 2017 [17]
SeleniumSelenium affects immune cells by influencing antioxidant functions and signaling pathways in cells like NK cells and T lymphocytes.Avery et al., 2018 [18]
BetacaroteneBetacarotene scavenges superoxide radicals and quenches singlet oxygen, reducing oxidative stress.Miazek et al., 2022 [19]
BetacaroteneBetacarotene reduces inflammation by downregulating the TLR4 signaling pathway.Cheng et al., 2021 [20]
FlavonoidsFlavonoids inhibit the NF-κB cascade, which is involved in inflammatory responses.Li et al., 2020 [21]
FlavonoidsFlavonoids chelate metal ions, uptake reactive oxygen species, and stimulate detoxification enzymes.Slika et al., 2022 [22]
Short-chain fatty acids (e.g., butyrate) produced by gut bacteriaCertain gut bacteria ferment dietary fiber to produce short-chain fatty acids; butyrate stimulates mitochondrial biogenesis, improves respiratory capacity, and activates antioxidant enzyme activity, enhancing antioxidant defenses and modulating redox signaling.Zhao et al., 2023 [23]
Metal-binding proteins (ceruloplasmin, ferritin)Ceruloplasmin oxidizes ferrous iron to ferric iron and binds it to transferrin for safe transport, while ferritin stores iron within enterocytes. By sequestering free iron, these proteins limit iron-mediated production of reactive oxygen species.Loveikyte et al., 2023 [24]
Table 2. Summarized table of information on oxidative stress in intestinal pathology.
Table 2. Summarized table of information on oxidative stress in intestinal pathology.
PathologyMechanism of ActionAuthor and Year
Drug/Toxin Exposure—ChemotherapyChemotherapy causes excessive production of ROS, leading to intestinal side effects such as mucositis. ROS and inflammatory mediators contribute to tissue damage in the gut.McQuade et al., 2016 [54]
Al-Asmari et al., 2016 [55]
Drug/Toxin Exposure—RadiotherapyRadiation reduces the body’s enzymatic antioxidant defenses, impairing the ability to neutralize oxidative stress, which may cause long-term damage to the gut.Musa et al., 2019 [56]
Drug/Toxin Exposure—NSAIDsNSAIDs like indomethacin increase mitochondrial superoxide and xanthine oxidase levels in colonic epithelial cells, leading to higher superoxide production and oxidative stress.Nagano et al., 2012 [57]
Drug/Toxin Exposure—AlcoholAlcohol metabolites like ethanol and acetaldehyde cause oxidative stress in epithelial cells, disrupting tight junction integrity and raising superoxide levels, leading to increased gut permeability.Samak et al., 2016 [58]
Ischemia–Reperfusion During intestinal ischemia–reperfusion, xanthine dehydrogenase converts to xanthine oxidase, which produces free radicals upon reoxygenation, leading to oxidative stress and tissue injury.Sasaki et al., 2007 [59]
Postoperative InjuryOxidative stress can occur independently of inflammation after tissue injury, possibly promoting further inflammation in postoperative injury scenarios.Matsumoto et al., 2018 [60]
Congenital Pathologies—Triple-A syndromeIn Triple-A syndrome, mutations in the AAAS gene disrupt the redox balance, increasing susceptibility to oxidative stress-induced cell death, particularly in tumors.Prasad et al., 2014 [44]
Congenital Pathologies—Hirschsprung’s diseaseOxidative stress during development impairs the formation of the enteric nervous system, contributing to congenital pathologies like Hirschsprung’s disease.Zhou et al., 2022 [45]
Inflammation—Inflammatory bowel diseaseOxidative stress plays a significant role in IBD, leading to chronic inflammation and tissue damage in colitis models.Sahakian et al., 2021 [61]
Inflammation—Necrotizing enterocolitisElevated oxidant levels are observed in patients with necrotizing enterocolitis, contributing to oxidative damage and inflammation in the gut.Aydemir et al., 2011 [62]
InfectionThe MRPS18B P260A variant in Chagas megaesophagus patients affects mitochondrial function and induces nitro-oxidative stress, exacerbating the disease.Silva et al., 2022 [49]
CancerIn colorectal cancer, free radicals from various sources induce oxidative stress, leading to genomic instability and transforming normal colon cells into dysplastic and neoplastic cells.Carini et al., 2017 [63]
CancerIn Barrett’s esophagus, oxidative stress is indicated by high levels of peroxynitrite, superoxide, and glutathione, which contribute to tissue damage and may lead to esophageal cancer.Jiménez et al., 2005 [64]
Table 3. Summarized table of evolution and diversity of experimental murine models of inflammatory bowel disease.
Table 3. Summarized table of evolution and diversity of experimental murine models of inflammatory bowel disease.
Species/StrainInduction/Intervention MethodOxidative Stress Markers AssessedMain OutcomesLimitationsAuthor and Year
Mouse (various; commonly C57BL/6/BALB/c)DSS-induced colitisNot reportedEpithelial denudation, barrier disruption, acute inflammation with increased ROS and lipid peroxidationModels acute injury; limited chronicity; dosing/time dependenceChassaing et al., 2014 [73]
Rat/MouseTNBS-induced colitisMPO (Myeloperoxidase) Th1-mediated colitis; oxidative injury with elevated ROS/MPO and mucosal damageSensitization-dependent; variability in severity; solvent effectsda Silva et al., 2010 [74]
Mouse (BALB/c)Oxazolone-induced colitisNot reported Th2/NKT-driven colitis mimicking UC features; mucosal inflammation, microulcerationsPrimarily distal colon; model reproducibility variesKojima et al., 2004 [75]
Mouse (Rag2−/− recipients)Adoptive CD4+ T-cell transferNot reported Chronic colitis via Th1 effector responses; sustained inflammationRequires immunodeficient hosts; labor-intensiveCorazza et al., 1999 [76]
Mouse (C57BL/6J; 6 to 8 weeks; female)Heavy-ion radiation-induced intestinal oxidative stress in miceIntracellular ROS, mitochondrial superoxide, NADPH oxidase activity, mitochondrial membrane potential, DNA oxidative damage, mitotic activity^56Fe radiation caused persistent increases in mitochondrial ROS, enhanced NADPH oxidase activity, mitochondrial dysfunction, and oxidative DNA damage in intestinal epithelial cells compared with γ-irradiationOnly one post-exposure time point (1 year); no assessment of early dynamic changes; applicability limited to radiation-induced oxidative stressDatta et al., 2012 [77]
Mouse (SAMP1/YitFc)Spontaneous mutation modelNot reportedSpontaneous CD-like ileitis; chronic inflammation; extraintestinal featuresStrain-specific; limited availabilityKosiewicz et al., 2001 [78]
Mice (TNBS model)Colitis induced with TNBS; treatment with Protocatechuic Acid (PCA)GSH, GSSG/GSH ratio, SOD, CAT, Nrf2 expressionTNBS colitis decreased antioxidant defenses (↓ GSH, ↓ SOD, ↓ CAT, ↓ Nrf2). PCA treatment restored antioxidant enzyme activity, improved GSH balance, and reduced oxidative stress and inflammationOnly one antioxidant compound tested; short-term study, no long-term outcomes assessedCrespo et al., 2017 [79]
Mouse (IL-7 transgenic)Genetic—IL-7 overexpressionNot reportedPersistence of colitogenic CD4+ memory T cells; chronic colitisImmune-niche specific; not a pure OS modelNemoto et al., 2007 [80]
Nemoto et al., 2009 [81]
Nemoto et al., 2011 [82]
Nemoto et al., 2013 [83]
Mouse (IEC STAT3/IL-22 axis)Genetic—IL-22 deficiency/impaired STAT3Not reportedDefective mucosal healing; worsened colitisFocused on repair pathways; OS not primary endpoint, but secondary effectPickert et al., 2009 [84]
Mouse (XBP1 in IECs)Genetic—XBP1 disruption (ER stress)Not reportedPaneth/goblet cell defects; inflammation driven by ER stressEmphasizes ER stress; OS not primary endpoint, but secondary effectKaser et al., 2008 [85]
Adolph et al., 2013 [86]
Mouse (T-bet−/−Rag2−/−; TRUC)Genetic—TRUC modelNot reportedCommunicable UC-like colitis; microbiota-immune interactionsComplex immune/microbiota interplay; OS not primary endpoint, but secondary effectGarrett et al., 2007 [87]
Mouse (Mdr1a−/−)Genetic—Mdr1a−/− colitisNot reportedBarrier dysfunction; bacterial translocation; chronic colitisTransporter-specific; strain housing effectsPanwala et al., 1998 [88]
Resta-Lenert et al., 2005 [89]
Collett et al., 2005 [90]
Mouse (Muc2−/−)Genetic—Muc2−/− deficiencyNot reportedMucus barrier loss; spontaneous colitis; tumor riskStrong barrier phenotype; OS readouts often indirectVelcich et al., 2002 [91]
Van der Sluis et al., 2006 [92]
Arrows (↓) indicate direction of change: ↓ = decrease.
Table 4. Summarized table of experimental models of inflammatory bowel disease.
Table 4. Summarized table of experimental models of inflammatory bowel disease.
Species/StrainInduction/Intervention MethodOxidative Stress Markers AssessedMain OutcomesLimitationsAuthor and Year
Guinea pigDSS colitis + curcumin C-SLNsLPO, protein carbonyl, MPO↓ leucocyte infiltration, ↓ OS, ↓ TNF-α; preserved colonic structureSpecies cost; limited resourcesSharma et al., 2019 [112]
Guinea pigDSS colitis + B. vulgatus 7K1Not reportedMicrobiota modulation; ↓ colitis severityProbiotic strain specificity; OS not a primary endpoint (secondary/indirect effect)Li et al., 2021 [113]
New Zealand White rabbitDSS colitis; endoscopic monitoringNot reportedEnables longitudinal endoscopic/bioptic assessmentOS markers not detailedLei et al., 2019 [114]
Dog (spontaneous IBD)Clinical evaluationNot OS; fecal canine calprotectin (cCP)cCP as non-invasive inflammation biomarkerOS markers not measuredOtoni et al., 2018 [115]
Pig / PigletDSS colitis; diet interventionsMostly inflammatory gene expression; OS not reportedUC-like features; dietary effects on mucosal immunityHigh cost; OS readouts variableNielsen et al., 2020 [95]
Rhesus macaque (spontaneous enteritis)Natural diseaseNot reportedImmune expression profiles overlap with human IBDLimited availability; ethics; OS not a primary endpoint (secondary/indirect effect)Wang et al., 2021 [98]
Zebrafish (larvae/juvenile)DSS/TNBS; toxicants; extractsNot reportedMirrors microbiota/immune shifts; neuronal changes in colitisDevelopmental stage-dependentUyttebroek et al., 2020 [100]
ZebrafishChemical; viral componentsNot reportedInnate immunity and epithelial repair pathways in GI inflammationInvertebrate; translational limitsHanyang et al., 2017 [102]
ZebrafishBacteria–epithelial interactionsSODConserved TFs/pathways; drug screening utilityNo adaptive immunity/circulationLi et al., 2022 [116]
Drosophila melanogasterChemical; viral componentsNot reportedInnate immunity and epithelial repair pathways in GI inflammationInvertebrate; translational limitsMadi et al., 2021 [108]
Caenorhabditis elegansBacteria–epithelial interactionsNot reportedConserved TFs/pathways; drug screening utilityNo adaptive immunity/circulationHaerty et al., 2008 [109]
Arrows (↓) indicate direction of change: ↓ = decrease.
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

Pavel, C.D.; Gales, C.; Pavel, I.A.; Zamfir, C.L. The Relevance of Experimental Models in Assessing the Impact of Oxidative Stress on Intestinal Pathology. J. Clin. Med. 2025, 14, 6569. https://doi.org/10.3390/jcm14186569

AMA Style

Pavel CD, Gales C, Pavel IA, Zamfir CL. The Relevance of Experimental Models in Assessing the Impact of Oxidative Stress on Intestinal Pathology. Journal of Clinical Medicine. 2025; 14(18):6569. https://doi.org/10.3390/jcm14186569

Chicago/Turabian Style

Pavel, Cristian Dan, Cristina Gales, Irina Andreea Pavel, and Carmen Lăcrămioara Zamfir. 2025. "The Relevance of Experimental Models in Assessing the Impact of Oxidative Stress on Intestinal Pathology" Journal of Clinical Medicine 14, no. 18: 6569. https://doi.org/10.3390/jcm14186569

APA Style

Pavel, C. D., Gales, C., Pavel, I. A., & Zamfir, C. L. (2025). The Relevance of Experimental Models in Assessing the Impact of Oxidative Stress on Intestinal Pathology. Journal of Clinical Medicine, 14(18), 6569. https://doi.org/10.3390/jcm14186569

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