Next Article in Journal
Soybean β-Conglycinin and Cowpea β-Vignin Peptides Inhibit Breast and Prostate Cancer Cell Growth: An In Silico and In Vitro Approach
Previous Article in Journal
Exploring the Potential of Anthocyanins for Repairing Photoaged Skin: A Comprehensive Review
Previous Article in Special Issue
Association between Gut Microbiota Profiles, Dietary Intake, and Inflammatory Markers in Overweight and Obese Women
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Effects of Interventions with Glucosinolates and Their Metabolites in Cruciferous Vegetables on Inflammatory Bowel Disease: A Review

1
College of Food Science, Southwest University, Chongqing 400715, China
2
Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China
3
Research Center for Fruits and Vegetables Logistics Preservation and Nutritional Quality Control, Southwest University, Chongqing 400715, China
4
College of Life Science, Sichuan Normal University, Chengdu 610101, China
5
Institute of Agro-products Processing Science and Technology, Sichuan Academy of Agricultural Sciences, Chengdu 610066, China
*
Authors to whom correspondence should be addressed.
Foods 2024, 13(21), 3507; https://doi.org/10.3390/foods13213507
Submission received: 30 September 2024 / Revised: 29 October 2024 / Accepted: 30 October 2024 / Published: 1 November 2024

Abstract

:
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract which affects millions of individuals worldwide. Despite advancements in treatment options, there is increasing interest in exploring natural interventions with minimal side effects. Cruciferous vegetables, such as broccoli, cabbage, and radishes, contain bioactive compounds known as glucosinolates (GLSs), which have shown promising effects in alleviating IBD symptoms. This review aims to provide a comprehensive overview of the physiological functions and mechanisms of cruciferous GLSs and their metabolites in the context of IBD. Reviewed studies demonstrated that GLSs attenuated all aspects of IBD, including regulating the intestinal microbiota composition, exerting antioxidant and anti-inflammatory effects, restoring intestinal barrier function, and regulating epigenetic mechanisms. In addition, a few interventions with GLS supplementation in clinical studies were also discussed. However, there are still several challenges and remaining knowledge gaps, including variations in animals’ experimental outcomes, the bioavailability of certain compounds, and few clinical trials to validate their effectiveness in human subjects. Addressing these issues will contribute to a better understanding of the therapeutic potential of cruciferous GLSs and their metabolites in the management of IBD.

1. Introduction

Inflammatory bowel disease (IBD) is a non-specific bowel disorder characterized by inflammation and disruption of the intestinal epithelial barrier, mainly including ulcerative colitis (UC) and Crohn’s disease (CD) [1]. The prevalence of IBD in developing countries was relatively low compared with North America and western European countries, but the incidence of IBD has been rising rapidly in newly industrialized countries in South America, Asia, and Africa in the past few decades due to their westernized lives [2,3,4]. IBD has been a public health challenge worldwide [5].
The pathophysiology of IBD is attributed to a complicated combination of genetic, microbial and environmental factors involving aberrant immunological responses to the intestinal microbiota, leading to chronic intestinal inflammation [6]. IBD not only damages the intestinal epithelial structure but also triggers more diseases through the gut-organ axis [7]. Epidemiological studies demonstrated that IBD patients have a higher risk of Alzheimer’s disease (AD), a neurodegenerative condition, and both diseases showed that the intestinal microbiota and metabolites changed in a similar way, suggesting that IBD may be one of the potential pathogenic factors of AD through bidirectional communication between intestinal microbes and the brain [8]. Clinically, drugs such as sulfasalazine, glucocorticoids, thioguanins and thiopurine can alleviate IBD symptoms. However, these medications do not alter the overall course of IBD and have many adverse effects, including gastrointestinal complaints (diarrhea, vomiting, abdominal pain, etc.), hepatotoxicity, pancreatitis and hyperglycemia [9]. Older adults are more susceptible to these adverse events than younger people [6].
Dietary therapy plays a critical part in IBD management, from prevention of the disease to active disease and complication treatment. Several diets have been investigated to treat active IBD, including the specific carbohydrate diet, Mediterranean diet, plant-based diet and low-emulsifier diet [10]. Moreover, studies have shown that dietary components can benefit the treatment of intestinal diseases at different levels through inhibiting intestinal inflammation, restoring gut microbiota and renewed intestinal epithelial and improved barrier function [11,12]. Dietary intakes of fruit and vegetables are correlated with a reduced risk of developing IBD (UC and CD) and are recommended for prevention of IBD [13,14]. Among them, cruciferous vegetables such as broccoli, cabbage and cauliflower are part of a balanced diet and rich in vitamins, minerals and dietary fiber as well as phytochemicals such as glucosinolates (GLSs). GLSs are a kind of secondary metabolite of cruciferous vegetables, which have attracted increasing attention recently. GLSs and their metabolites have anti-inflammatory, anticarcinogenic, antibacterial, antioxidant and other activities, and they also exert protective effects on IBD in mouse models and human intestinal cell models [15].
In this review, the metabolism in vivo and physiological functions of cruciferous GLSs are summarized, mainly focusing on the protective mechanism of GLSs and their metabolites on IBD, including gut microbiota modulation, antioxidant activity, anti-inflammatory activity and maintaining the intestinal barrier. Furthermore, the influences of processing methods on the content of total GLSs and isothiocyanates (ITCs) are also reviewed to provide perspectives and foundations for dietary prevention of IBD in the future.

2. Cruciferous Vegetables and Glucosinolates

Cruciferous vegetables, characterized by their four equal-sized petals [12], belong to the family Brassicaceae and include widely consumed vegetables such as broccoli, kale, mustard, radish and cabbage. These cruciferous vegetables possess a variety of health-promoting activities, which are ascribed to their secondary metabolites [16]. Of them, GLSs are a group of nitrogen- and sulfur-containing water-soluble secondary metabolites, which are synthesized by amino acid (especially methionine) chain extension, formation of the glucosinolate core structure and the secondary modification of side chains [17]. Therefore, all GLS compounds are structurally similar, including β-D-thioglucoside groups, sulfonated moieties and R groups of amino acid side chains of different origins [18]. According to the different R groups, it can be divided into aliphatic groups (side chains derived from methionine, alanine, valine, leucine and isoleucine, among other amino acids), aromatic (side chains derived from tyrosine and phenylalanine) and indole groups (side chains derived from tryptophan), of which aliphatic types account for the largest proportion [19].
The GLS content varies with cruciferous vegetables varieties, growth environments, different organs, and signal substances which induce responses [19,20]. For kale, broccoli, mustard and Chinese cabbage, their GLSs are mainly aliphatic and indole individuals (Table 1). There are differences in the types and amounts of GLSs among cruciferous vegetables. Variation in GLS content has also been observed among different organs in the same vegetable [19]. Therefore, the total GLS content of vegetables falls within a certain range, with the highest content found in seeds. In addition to the edible parts of vegetables, their roots and leaves are also rich in GLSs, and thus these parts can be reasonably utilized for their contained GLSs, aside from being used as feed or fertilizer or discarded as waste [19].

3. Physiological Functions of GLSs and Metabolites

3.1. Metabolism of GLSs In Vivo

In intact plants, GLSs are chemically stable, biologically inert and not highly active, but they can be degraded by myrosinase and influenced by environmental factors such as the temperature, light and pH [23]. GLSs and myrosinase are stored in different cells of cruciferous vegetables. Once the plant tissues are disrupted, they meet, react and produce ITCs, nitriles, epithionitriles (ETNs), etc.
When cruciferous vegetables are chewed, enzymatic degradation also occurs in the human gastrointestinal tract [24]. Thirteen GLS metabolites were found in human urine and plasma after broccoli consumption, including sulforaphane (SFN), sulforaphane-cysteine and sulforaphane-N-acetyl-cysteine from glucoraphanin (GRP), indole-3-carbinol (I3C), indole-3-carboxaldehyde and indole-3-carboxylic acid from glucobrassicin (GBS) and methoxyl indole-3-carbinol, methoxyl-indole-3-carboxaldehyde, methoxyl-indole-3-carboxylic acid and methoxyl-ascorbigen from methoxyl glucosinolates [25]. Oral administration of broccoli leaves demonstrated that low levels of GLSs were absorbed in mice. Only glucoerucin (GER), GBS, SFN and I3C were detected in plasma, and SFN and its derivatives mainly remained in the intestinal tract and then appeared in the feces [26].
The metabolism of GLSs in vivo is attributed to myrosinase derived from plant tissue or the human microbiota. Since the first bacterial myrosinase was detected, a large number of microorganisms which can metabolize GLSs have been isolated [24], including Lactobacillus, Bifidobacterium, Bacteroides, Enterococcus and Escherichia coli, which inhabit different parts of the mammalian intestine [27,28], as well as Aspergillus niger, Aspergillus syndowi and other fungi [23,29], but these bacterial myrosinases have lower hydrolyzing efficiencies than that of plant myrosinase, and the main metabolite of glucorabhanin is nitrile rather than SFN [30]. Lastly, consumption of cruciferous vegetables can induce myrosinase-like activity in intestinal microbiota. For example, four days of broccoli consumption enhanced the conversion of GRP to the corresponding ITCs by the cecal microbiota ex vivo [31].

3.2. Alleviating Effects of GLSs and Their Metabolites on Colitis

GLS metabolites possess several health benefits, such as antioxidant, antibacterial, anticancer and anti-inflammatory activities as well as regulation of intestinal flora and other effects [28,32,33]. The physiological functions of GLSs and their metabolites have been discussed in previous reviews [12,16]. Our review mainly focuses on the alleviating effects of GLSs and their metabolites on IBD.
GLSs and their metabolites are promising in the prevention of gastrointestinal diseases. For example, phenylethyl isothiocyanate (PEITC) exerts anti-inflammatory activities by inhibiting NF-κB activation in gastrointestinal inflammation diseases [34]. Benzyl isothiocyanate (BITC) can regulate inflammation, oxidative stress, and apoptosis through the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) and NF-kB signaling pathways, thus exerting anti-gastric ulcer effects [35].
Previous studies have found that cruciferous vegetables, such as GLS-rich broccoli, cabbage and mustard greens, can effectively alleviate the symptoms of IBD in human intestinal cell models and nematode and murine models (Table 2). For intestinal colitis study, dextran sodium sulfate (DSS, 2~5%)-induced mice colitis is one of the most widely used models [36], characterized by extensive damage to the intestinal epithelium, depletion of goblet cells, infiltration of inflammatory cells, and the presence of edema [37,38].
Cruciferous vegetables, GLSs and metabolites have demonstrated a potent effect in attenuating DSS-induced colitis symptoms. Both raw broccoli and lightly cooked broccoli can alleviate the DSS-induced disease activity index (DAI), relieve symptoms such as a 50% reduction body weight, stool bleeding, a shortened colon length and increased intestinal barrier permeability, and reduce colonic injury [15,39]. GRP supplementation improved body weights, stool blood, fecal consistency, and colon tissue morphology in DSS-induced colitis mice [40]. SFN can reverse body weight loss and shortened colon lengths, improve intestinal gland damage and bleeding and reduce the fibrotic area in colitis models [29,41,42]. I3C treatment in 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis mice maintained crypt formation and a normal colon tissue structure, and it showed signs of reduced cellular infiltration [43]. Meanwhile, 3,3′-Diindolylmethane (DIM), produced by the digestion of I3C, can repair IL-1β-induced differentiation of the human intestinal Caco-2 cell monolayer barrier and restore its permeability [44]. Allyl isothiocyanate (AITC)-treated intestinal epithelium showed a relatively intact structure and more goblet cells [45]. However, to our knowledge, there are few clinical trials for GLS and isothiocyanates and their effect on intestinal inflammation.
Table 2. Effect of cruciferous vegetables, GLSs and metabolites on alleviating the symptoms of IBD.
Table 2. Effect of cruciferous vegetables, GLSs and metabolites on alleviating the symptoms of IBD.
ModelsInducementInterventionEffectsReferences
C57BL6/J-Ahrb/b and Ahrd/d mice, 8–10 weeksdrinking 3.5% DSS water for 6 days15% broccoli diet for 14 days beforehandbroccoli diet significantly attenuated the clinical manifestation of splenomegaly and DAI in Ahrb/b and Ahrd/d mice[46]
C57BL/6 mice, male, 8–10 weeksdrinking 2.5% DSS for one week10% raw broccoli (RB), 10% lightly cooked broccoli (CB)both CB and RB effectively reduced DAI, extended colon length and induced less blood endotoxin and less severe colon lesions[39]
interleukin (IL)-10-knockout mice on C57BL/6 backgroundinoculation with Helicobacter hepaticusdiet with 10% raw broccoli sproutsbroccoli sprout diet reduced weight stagnation, fecal blood and diarrhea, enhanced gut microbiota richness and reduced the prevalence and abundance of pathobiont bacteria triggering inflammation[47]
C57BL/6J mice, female and maledrinking 2% DSS water for 5 days for SPF mice and 1% DSS for GF mice5% steamed broccoli sprout (SBS) diet for 4–6 weeksSBS decreased DSS-induced colitis via the gut microbiota converting cruciferous vegetables into bioactive metabolites, promoting anti-inflammatory effects[15]
C57BL/6J mice. male and female, 7–8 weeksaater with 3% DSS for 1 week, followed 1 week of recovery and then 1 week 3% DSSred cabbage juice (RCJ)RCJ significantly improved body weight and survival of mice, decreased DAI scores, improved intestinal barrier integrity by enhancing the expression of colonic mucins and tight junction (TJ) proteins and the abundance of SCFA-producing bacteria and increased PPAR-γ activation[48]
Wistar rats, maledrinking 4% DSS water for 6 days, mild colitisdiet with 8750 mg/kg broccoli
sprout extract (BSE)
diets with BE reduced the DSS-induced rise in the expression of pro-inflammatory mediators NFκB, MCP-1, COX2 and VCAM-1[49]
C57BL/6J mice male, 6 weeks old25 g/L DSS water370 mg/kg·day BSE dissolved in 0.2 mL of skim milkBSE administration increased body weight, improved antioxidant activities and restored the intestinal barrier through enhancing TJ protein expression[50]
C57BL/6J mice, male, 7 weeksdrinking 3% DSS water for 5 days (acute UC) and cyclic rotations of 2.5% DSS water for 30 dmoringa seed extract (MSE)MSE decreased DAI scores and colon weight/length ratios, increased colon lengths, reduced colonic inflammation and damage in acute UC, decreased colonic pro-inflammatory expression and downregulated gene expression of pro-inflammatory activity[51]
Sprague Dawley rats, male, 3 monthsintrarectal injection with 2,4-dinitrobenzenesulfonic acid (DNBS, 20 mg in 0.25 mL of 50% ethanol)Eruca sativa defatted seed meal (0.1~1 g/kg p.o)administration of E. sativa seed (1 g/kg) promoted colon recovery from injury and decreased enteric gliosis[52]
C57BL/6J mice, male, 6 weekswater consisting of 2.5% (w/v) DSSgavage with BSE (370 mg/kg·day) or with Bifidobacterium longum CCFM1206 (109 CFU/mL)combined treatment of B. longum CCFM1206 and BSE ameliorated DSS-induced colitis symptoms, mitigated colonic inflammatory levels and oxidative injury and restored the intestinal barrier[53]
C57BL/6 mice, 8 weeks olddrinking 2.5% DSS for 9 daysdiet supplemented with GRP (600 ppm) for 4 weeksGRP attenuated body weight loss, DAI and colon shortening, maintained the colonic structure, inhibited inflammatory reactions and reduced colonic macrophage infiltration[40]
C57BL/6 micedrinking 4% DSS water for 5 days, acute colitispretreatment with 25 mg/kg·b.w. SFN per os for 7 daysSFN pretreatment significantly minimized body weight loss and DAI, extended colon lengths and relieved colon inflammation[37]
C57BL/6 mice, male, 8 weeks olddrinking 3% DSS water AITC in corn oilAITC intervention showed less body weight loss, fewer colitis symptoms and longer colons, lessened the disruption of colonic histological structure and decreased mucosal inflammation[54]
C57BL/6 mice, male, 8 weeks olddrinking 3% DSS water for 7 daysDIMDIM significantly ameliorated the clinical symptoms and histological features, reduced inflammatory cell infiltration and suppressed the expression of pro-inflammatory cytokines and vascular endothelial growth factors[55]
C57BL/6 mice, female, 6 weeks olddrinking 2.5% DSS water for 7 daysAITC (10 mg/kg/day) for 7 daysAITC could attenuate the severity of colitis through enhancing the intestinal barrier, including both TJ protein and mucin expression[45]
BALB/cJ and C57BL/6 mice, female50 μL intrarectal injections of 1 mg of TNBS in 50% ethanolI3C (40 mg/kg in 0.05% DMSO/corn oil)I3C repressed colonic inflammation and prevented microbial dysbiosis, increasing a group of butyrate-producing gram-positive bacteria, which was correlated with an increase in IL-22[43]
C57BL/6 (18~22 g), maledrinking 2.5% DSS waterSFN intragastric administration 20 mg/kg·days for 2 weeksSFN treatment increased body weight and colon length, decreased the colon damage scores and myeloperoxidase (MPO) activity, reversed DSS-induced gut microbiota dysbiosis and restored the abundance of Butyricicoccus[29]
Sprague Dawley ratsintracolonic single administration of 2 mL of 4% acetic acidSFN (15 mg/kg) by oral gavage daily for 2 weeksSFN maintained the length and weight of the colon and improved morphological changes by improving antioxidant ability, elevating mitochondrial biogenesis and suppressing DNA polymerization[42]
C57BL/6 mice, maledrinking 2% DSS water for 7 daysSFN (2.5, 5, 10 and 20 mg/kg body weight)SFN treatment alleviated the changes in colon length, DAI scores and pathological damages, partially recovered gut microbiota disorder and enhanced the content of volatile fatty acids[41]
C57BL/6JNifdc mice, male, 6–8 weeks old2.5% DSS was gavaged for 7 daysSFN (20, 40, 10 mg/kg·days)SFN effectively attenuated intestinal inflammation through skewing the switching from classically (M1) to alternatively (M2) activated phenotypes both in intestinal and bone marrow-derived macrophages, leading to changes in the inflammatory mediators[38]
human colonic cancer cell lines HT-29 and
Caco-2
treated with IL-1β (1 ng/mL) for 5 hDIMDIM mainly recovered the intestinal permeability of differentiated Caco-2 cells through increasing TJ protein expression and significantly enhanced the transepithelial electrical resistance of the cell monolayer[44]
Caenorhabditis elegans, wild-type strain N2 and the mutant strain SS104feeding with Pseudomonas aeruginosa PAO1DIMDIM relieved the damaged intestinal permeability and prolonged the lifespan of C. elegans fed P. aeruginosa[44]
In conclusion, GLSs and their metabolites can effectively alleviate the IBD symptoms in in vivo and in vitro experiments. The intervention mechanisms are mainly intestinal microbiota regulation, antioxidant and anti-inflammatory effects, maintenance of the intestinal barrier and regulation of epigenetic mechanisms.

4. Mechanism of GLSs and Their Metabolites in Alleviating IBD

4.1. Regulation of Gut Microbiota

The dysbiosis of the gut microbiota is associated with the pathogenesis of IBD [32,56]. Individuals with intestinal inflammation have an imbalance in their gut microbiota, characterized by a decrease in bacterial diversity and alterations in the relative abundance of certain genera and species. A comprehensive reanalysis of fecal 16S rRNA amplicon sequencing data from 934 IBD patients and 1584 healthy individuals confirmed reduced alpha and beta diversity in IBD patients compared with healthy controls and identified 38 novel differential genera in CD and 28 novel genera in UC [57]. These lower-abundance genera in IBD patients included the Lachnospiraceae NK4A136 group, Akkermansia, Faecalibacterium, Roseburia, Alistipes and Parabacteroides, while these genera Streptococcus, Enterococcus and (Ruminococcus) gnavus group were more abundant in both CD and UC. The relative abundance of Lactobacillus, (Eubacterium) xylanophilum group and other Muribaculaceae also decreased in the DSS-treated mice [50].
It was observed that there is a decrease in the relative abundance of bacteria with anti-inflammatory activities belonging to Clostridium cluster IV and an increase in the proportion of bacteria with pro-inflammatory activities in IBD patients [56]. The increased pathogenic bacteria, particularly Escherichia coli, invade the intestinal epithelial cells and induces inflammatory responses [42].

4.1.1. Regulation of Gut Microbiota Composition

The gut microorganisms with myrosinase-like activities can hydrolyze GLSs, and the GLS metabolites in turn affect the gut microbiota [24]. Broccoli extract, which mainly contains polyphenols and GLSs, increases the proportion of beneficial bacteria such as Bifidobacterium, Blautia, Coprococcus and Phascolarctobacterium, and it reduces the harmful bacteria Escherichia in vitro, while GLS hydrolysis is remarkably associated with Bilophila, Alistipes, Bifidobacterium, etc. [58]. Red cabbage juice intervention improved the abundance of Butyrivibrio, Roseburia, Ruminococcaceae, Acetatifactor muris, Rosburia Sp. CAG:303 and Dorea Sp. 5–2 compared with the DSS group. In the case of Clostridia, it has been reported to produce butyrate [48]. Administration of steamed broccoli sprouts increased the richness and diversity of mice gut microbiota and altered the microbial community structure, resulting in increasing the relative abundance of Firmicutes and Bifidobacterium and the beneficial bacteria Akkermansia muciniphila (Akkermansia) while reducing Bacteroides acidifaciens [15,41]. Bifidobacterium produced short-chain fatty acids (SCFAs) to maintain the gut barrier, while Bacteroides acidifaciens degraded mucin and compromised the protective layer of intestinal epithelial cells, leading to colonic inflammation (Figure 1).
It has been demonstrated that GLSs and their metabolites can alter intestinal microbiota compositions in colitis models. SFN partially restored gut microbiota dysbiosis induced by DSS administration and increased the relative abundance of Butyricicoccus, synergistically exerting anti-inflammatory effects with Butyricicoccus [29]. I3C prevented TNBS-induced microbial dysbiosis, characterized by an increase in gram-negative bacteria, and selectively increased the proportion of the butyrate-producing gram-positive bacteria, especially Roseburia [43].

4.1.2. Promoting the Production of SCFAs

SCFAs, a class of crucial metabolites of intestinal microbiota, show anti-inflammatory activity by binding and activating endogenous receptors, such as GPR41 and GPR43, to prevent immune reactions, improve the level of IL-10 and inhibit the production of IL17, thus protecting the intestinal barrier, suppressing excessive signaling of TLR and alleviating IBD symptoms [59,60,61].
Jaworska et al. demonstrated that there was an increase in the blood-to-stool ratio of SCFAs both in rats and pediatric patients with IBD, which was attributed to increased intestinal barrier permeability [62]. Generally, SCFA contents were decreased in IBD-related microbial dysbiosis, which is consistent with the depletion of SCFA producers such as Faecalibacterium prausnitzii and Roseburia hominis [63]. Although the levels of lactic acid, a SCFA precursor, were increased in CD and UC adult patients, microbial dysbiosis may prevent the conversion of lactic acid to butyric acid, leading to a decrease in SCFAs [63,64].
SFN administration promoted the production of SCFAs [41]. Treatment with I3C can increase the abundance of Roseburia (which produces butyrate) and the concentration of butyric acid [43]. Increased levels of butyric acid and isobutyric acid in mice can enhance the production of anti-inflammatory cytokines and regulate inflammation by reducing pro-inflammatory cytokines [65]. Butyric acid can be absorbed and utilized by intestinal epithelial cells, inhibiting neutrophil function, improving barrier function and reducing inflammatory responses [66]. Treatment with butyrate salts significantly improved intestinal inflammation in mice and protected the intestinal epithelial barrier [41]. Sodium butyrate can activate AMPK to induce mitophagy, alleviate hydrogen peroxide-induced oxidative stress and maintain the intestinal epithelial barrier’s integrity [67].

4.2. Antioxidant Activity

Although the underlying pathomechanisms of IBD have not yet been completely clarified, accumulating clinical and experimental evidence demonstrates that oxidative stress plays a critical role in the initiation and development of IBD [68]. Under physiological conditions, reactive oxygen species (ROS) and reactive nitrogen species (RNS) are produced in the intracellular organelles of intestinal cells. Once the antioxidant defense system is overwhelmed by high levels of ROS or RNS generation, excessive pro-oxidants attack the gastrointestinal mucosal layer and alter the inflammatory response [68], resulting in more cellular impairments, including DNA damage, protein aggregation and membrane dysfunction [69]. Certain gut microorganisms can also produce ROS directly, which further worsens intestinal inflammation [70].
GLSs and their metabolites have no direct antioxidant activity, but they can induce cell-protective enzymes, including various antioxidant enzymes, to exert indirect antioxidant effects [71]. SFN attenuates LPS-induced oxidative stress in Caco2 cells through improving the levels of superoxide dismutase, glutathione peroxidase, catalase and total antioxidative capacity, and it activates the AMPK/ SIRT1/PGC-1α pathway, which helps control the levels of ROS in mitochondria [72]. Cedrowski et al. found that SFN and erucin thermally decompose into sulfenic acids and methylsul finyl radicals, thus exhibiting antioxidant activity at high temperatures above 100 °C [73]. SFN strongly activates transcription factor EB (TFEB) to promote autophagic flux and lysosomal biogenesis, relieving oxidative stress [74].
GRP supplementation activated Nrf2, increased the levels of heme oxygenase-1 (HO-1) and xanthine oxidase and reduced the 8-hydroxydeoxyguanosine levels in DSS-induced colitis mice [40]. Nrf2 can reduce intestinal mucosal damage by inhibiting ROS production and enhancing the transcription of antioxidant target genes [75]. SFN treatment increased the Nrf2 levels in UC rats [42]. Nrf2 is negatively regulated by kelch-like ECH-associated protein 1 (Keap1), which promotes its degradation through the ubiquitin-proteasome pathway. Naturally occurring ITCs such as AITC, BITC and SFN can promote Nrf2 dissociating from Keap-1 [76,77]. Although there is little evidence that SFN can activate Nrf2 in humans [78], as an electrophile, SFN could react with the cysteine residues in Keap1 [79,80], forming a sulfenic acid-based adduct with Keap1 [81] and thereby rendering Nrf2 binding ineffective and inducing Nrf2 expression [82].

4.3. Anti-Inflammatory Activity

4.3.1. Downregulating Inflammatory Mediators

Inhibiting pro-inflammatory cytokines and maintaining anti-inflammatory cytokines. Pro-inflammatory cytokines (tumor necrosis factor-alpha (TNF-α), IL-1, IL-6, IL-12, IL-17, IL-18, IL-21 and IL-23), especially IL-6, TNF-α and IL-1β, are associated with the initiation and progression of IBD [7,40,83], which result from recruited monocytes and activated macrophages. IL-10, transforming growth factor-beta (TGF-β) and other anti-inflammatory cytokines also contribute to the pathogenesis of IBD by reducing the inflammatory response [84].
GLSs and their metabolites could alleviate IBD symptoms by inhibiting the levels of pro-inflammatory cytokines and promoting the production of anti-inflammatory cytokines. GRP pretreatment downregulates inflammatory cytokines such as IL-1β, IL-18 and TNF-α [40]. I3C, the hydrolysate of GBS, is a natural ligand of the aryl hydrocarbon receptor (AHR) [85]. It has been reported that IBD patients have reduced levels of endogenous AHR ligands compared with healthy individuals. AHR ligands can alleviate colonic inflammation through AHR activation, making AHRs a potential target for IBD treatment [86]. Peng et al. demonstrated that AHR gene knockouts promote intestinal epithelial cell death and inflammation [87]. AHR activation can interact with inhibitors of apoptosis proteins (IAPs), namely ubiquitin ligase, to upregulate its expression, and IAPs can accelerate the degradation of receptor-interacting protein kinase 1 (R1PK1). R1PK1 is a key mediator of programmed cell death and inflammation [87]. AHRs activated by I3C reduce the expression of other pro-inflammatory cytokines, including IL-8, IL6-, IL-1β and TNF-α [87,88] while maintaining the level of anti-inflammatory cytokine IL-22 mRNA [89]. Philip B. Busbe et al. pointed out that I3C mainly induces IL-22 to alleviate inflammation [43]. DIM is also an AHR ligand which can reduce the IL-8 levels in human colon cells treated with IL-1β [44].
Lower concentrations (10~20 μM) of SFN suppress the expression of pro-inflammatory cytokines [90,91]. Treatment with SFN significantly reduced the concentrations of pro-inflammatory cytokines IL-6, IFN-γ and TNF-α in mice with DSS-induced colitis [15,41,90]. SFN also increased the content of the mediator STAT3, which is involved in colitis and colitis-associated colon cancer epithelial repair as well as inflammation processes [41], and it weakens the ability of LPS to induce the production of pro-inflammatory cytokines (IL-1β, IL-6, IL-8 and TNF-α) and pro-apoptotic caspases-3 and 9 [72]. The inhibition of IL-6 by SFN depends on the interference of Nrf2 with the binding of IL-6 gene transcription [39]. SFN can enhance the expression of Nrf2 and HO-1. The activation of Nrf2 and HO-1 enhances the production of anti-inflammatory cytokines, especially IL-4 and IL-10 [91,92]. Additionally, SFN can induce Nrf2 to stimulate immune responses and exert anti-inflammatory effects [93] as well as regulate the inflammatory enzyme iNOS to inhibit the production of NO [91].
Interfering with M1 macrophage polarization. Activated specific immature macrophages in an inflamed alimentary tract secrete pro-inflammatory cytokines to aggravate severe reactions and barrier damage. Reconstruction of the barrier and elimination of inflammation are driven by the switch in polarization from the classically activated phenotype (M1) macrophages to the alternatingly activated phenotype (M2) macrophages [94]. The balance of M1 and M2 macrophages is associated with intestinal inflammation, which becomes more severe with an increase in M1 macrophages. LPS-activated mouse macrophages polarize to the M1 type by inducing pro-inflammatory marker proteins to alter their energy metabolism and reduce respiration [95]. SFN pretreatment successfully interferes with M1 polarization of LPS- and IFNγ-mediated THP-1-derived macrophages, producing high-energy cells with high glycolysis and high respiration and decreasing the expression levels of M1 (IL-23, CCR7, IL-1β, IL-6 and TNF-α) markers [94].
Co-crystallization with MIF. Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine which acts as a mediator of inflammation and innate immune response and has been associated with several conditions, including IBD [96,97]. It has been found that ITCs could co-crystallize with seven molecular targets, one of which is MIF [98]. ITCs inhibit the tautomerase activity of MIF through covalently binding its N-terminal cysteine, suggesting a potential pathway against inflammatory colitis.
Attenuation of inflammasomes. Inflammasomes are cytosolic multiprotein complexes of the innate immune system which assemble in response to invading pathogens or endogenous stress [99]. Activation of the inflammasome generates active caspase-1, which cleaves the precursor cytokines pro-IL-1β and pro-IL-18 into active IL-1β and IL-18 and induces pyroptosis [100]. The NLR family pyrin domain-containing 3 (NLRP3) inflammasome is composed of three parts: NLRP3, apoptosis-associated speck-like protein containing a CARD (ASC) and pro-caspase-1 [101].
Inflammasome activation was causatively linked to the development of chronic intestinal inflammation [102]. The expression of NLRP3, ASC and caspase-1 was significantly increased in a DSS-induced mouse colitis model, resulting in activation of related proteins and increases in the levels of IL-18 and IL-1β [103]. SFN administration reversed these changes to varying degrees, inhibiting NLRP3 activation and restoring IL-18 and IL-1β, thus decreasing intestinal inflammation. BITC could also inhibit inflammasome activation and IL-1β production in LPS- and ATP-stimulated THP-1 cells by suppressing the NF-κB pathway [104]. As a potential target for IBD treatment, the NLRP3 inflammasome needs more attention in the future.

4.3.2. Inhibiting NF-κB

Nuclear factor-κB (NF-κB) is a key transcription activator factor in inflammation and plays a crucial role in inflammatory responses. It has been observed that constitutive activation of NF-κB inflames colon tissue in IBD patients [105]. Normally, NF-κB is bound to IκB proteins in the cytoplasm. Upon stimulation by various factors (including cytokines, growth factors, mitogens, microbial components or stress factors), IκB is phosphorylated, ubiquitinated and degraded, leading to the translocation of NF-κB into the nucleus, where it controls the transcription of pro-inflammatory biomarkers such as inducible nitric oxide synthase (iNOS), TNF-α and IL-1β, among others [35]. SFN could inhibit the phosphorylation of IκB, thereby suppressing the transcription of pro-inflammatory genes controlled by NF-κB. LPS activates NF-κB upon TLR4 signaling, resulting in the production of pro-inflammatory cytokines. SFN can inhibit TLR4 signaling [106], attenuate cell stimulation by LPS and significantly suppress the transcription and translation of iNOS, TNF-α and IL-1β.
I3C and DIM are effective inhibitors of NF-κB [107] and reduce T cell activation and pro-inflammatory cytokine production [108,109]. AHR, activated by I3C and DIM, inhibits the activation of NF-κB by directly binding to NF-κB, as well as promoting IAP activity and inhibiting the activation of R1PK1 [87]. Meanwhile, 6-MITC, a compound of Japanese horseradish, competitively inhibits glycogen synthase kinase-3β (GSK-3β) and NF-κB. GSK-3β is an important positive regulator of various pro-inflammatory cytokines and mediators, and its inhibition helps alleviate colonic inflammation [110]. In addition, Chang et al. pointed out that regulating the balance between Th17 and Treg is an effective strategy for the treatment and prevention of IBD. Th17 cells have pro-inflammatory effects, while Treg cells have anti-inflammatory effects, and I3C and DIM can inhibit Th17 cells and increase Treg activity [111].

4.4. Maintaining the Intestinal Barrier

The intestinal barrier protects the human body from external harmful factors. IBD is characterized by symptoms including decreased expression of tight junction (TJ) proteins and mucins and thus disruption of the intestinal epithelial barrier’s integrity [41,45]. The intestinal epithelium works as a barrier with a layer of physical and immunological defense which not only prevents the invasion of pathogens and microorganisms but also serves as a selective permeability membrane [45,50]. TJ proteins are non-classical transmembrane proteins which connect intestinal epithelial cells and thus maintain the semi-permeable properties of these cells [45].
DIM treatment enhanced the expression of TJ proteins (including occludin and ZO-1) and significantly restored intestinal permeability in differentiated Caco-2 cells treated with IL-1β [44]. AITC-regulated TJ proteins and mucin 2 alleviated DSS-induced intestinal injury and failure [45]. The administration of broccoli seed extract can upregulate mRNA levels and TJ protein expression, including claudin-1, occludin and ZO-1, being beneficial to the intestinal barrier [50]. Apart from countering oxidative stress, Nrf2-Keap1 has been reported as one of pathways which regulates TJ protein expression [75], and GLS can activate Nrf2, as we noted above.

4.5. Other Mechanisms

Epigenetic modifications include major DNA methylation, histone modifications and miRNA expression. Many studies have shown that GLS compounds could exert anticancer effects by modulating epigenetic mechanisms [112,113,114,115]. However, there is limited research on the alleviation of IBD through the modulation of epigenetic mechanisms.
MicroRNAs (miRNAs) are a group of single-stranded non-coding RNAs which regulate gene expression through targeting mRNA post-transcriptionally [116]. Imbalances in miRNAs in IBD patients mediate inflammation through various pathways [117], and miRNAs are also considered biomarkers for the diagnosis and prevention of IBD [118]. SFN and DIM can regulate different miRNA expressions through various pathways to protect the intestines [119]. For example, Saleh et al. found that SFN significantly downregulates the expression of miRNA-155 and miRNA-146a, which are key regulators of TLR4-mediated inflammatory response, at the epigenetic level [106].

5. Processing Strategy of GLS-Containing Cruciferous Vegetable

The metabolites of GLSs, including ITCs, indoles, ETNs and nitriles, vary greatly as they are influenced by several factors, which was well reviewed in previous articles [120]. According to our review on the effects of GLSs and metabolites on colitis, GLS metabolites such as SFN, AITC and I3C exert a beneficial effect on the colitis model. Studies demonstrated that ITCs are health-promoting GLS metabolites, whereas ETNs and nitriles have less beneficial health effects and even exert adverse effects [121]. The functional properties of ITCs can be attributed to the special structure of a —N—C—S group, whose highly electrophilic carbon atom can react with strong nucleophiles such as thiols or protein disulfide bonds in physiological conditions [122]. Therefore, the consumption of cruciferous vegetables with high bioactive compound contents is desirable.

5.1. Pretreatment

Processing treatments for cruciferous vegetables, such as cutting, blanching, freezing, heating and drying, affect GLS stability and metabolism in vitro and in the body, as well as their health benefits [123] (Table 3). Cutting vegetables leads to the enzymatic degradation of GLSs into ITC or nitriles at low pH levels or ETNs in the presence of epithiospecifier proteins (ESPs). Blanching pretreatment (30 s) followed by cooling treatment had no significant influence on the total GLS content [124].

5.2. Heat Treatment

Low-intensity heat treatment can increase ITC formation. For example, high-temperature conditions inhibit the activity of myrosinase and ESPs to an extent at 70 °C and 50 °C, respectively [30]. When heating above 60 °C, ESP activity decreases significantly, while myrosinase remains active, and thus bioactive ITC metabolites increase [121]. Once food processing raises the temperature of vegetables above 70 °C, this results in a low utilization rate of GLSs and a low SFN content [125]. Long-term heating led to thermally induced GLS breakdown and an increase in the formation of nitriles [121,126]. The chemical degradation product thioglucose reacted with the ITCs to further generate new degradation products, namely 4-hydroxy-3-(4-(methylsulfinyl) butyl) thiazolidine-2-thione and 3-alkyl-4-hydroxythiazolidine-2-thiones, during aqueous heating [127,128]. Neither health benefits nor toxic effects were found with 3-alkyl-4-hydroxythiazolidine-2-thiones up until now. Therefore, controlling the thermal intensity (temperature and time) can decrease the losses of GLSs.
Table 3. Changes in GLSs and their metabolites after different processing.
Table 3. Changes in GLSs and their metabolites after different processing.
MaterialsGLSs Before TreatmentProcessing MethodDetection MethodGLSs and Their Metabolites After TreatmentRef.
broccoli
cauliflower, white cabbages, red cabbages, Chinese cabbages, baby cabbages,
white radish roots and red radish roots
Total GLS contents varied among vegetable type and could be
ordered as follows: red radish root > broccoli > white cabbage > red
cabbage > white radish root > baby cabbage > Chinese cabbage >cauliflower. The dominant GLS also depended on each vegetable type. For example, GRP accounted for 58.77% and 47.33% of the total GLS contents of broccoli and red cabbage, respectively.
blanching (30 s) and cooling at 2–4 °C for 5 minHPLC quadrupole time of flight (QTOF)blanching had little influence on the total GLS contents[124]
QF −/+ boiling (8 min)Total GLS contents of QF groups were higher than other groups for each species. High treatment temperature in VD resulted in a low GLS content. Blanching VFD is suitable for GLS preservation. Boiling led to a further decrease in the GLS content. The order of total GLS contents for each species was QF-B > VFD-B > VD-B > OD-B. There were significant differences in the stability among different GLSs or cruciferous vegetables.
OD −/+boiling (8 min)
VD −/+ boiling (8 min)
VFD −/+ boiling (8 min)
red cabbage0.21 µmol/g FW of ITCs but 0.62 µmol/g FW ETNs and 0.17 µmol/g FW nitriles formed after homogenizationaqueous heat treatment at 100 °CUHPLC, GC-MS4-pentenenitrile ↓, 3-butenyl isothiocyanate ↑ and 1-cyano-3,4epithiobutane ↓ after short heat treatment (2–3 min), where nitriles accounted for 92% after 120 min of heat treatment[121]
white cabbage0.72 µmol/g FW of ITCs, 0.50 µmol/g FW of ETNs and 0.16 µmol/g of nitrilesaqueous heat treatment at 100 °CUHPLC, GC-MS3-butenenitrile ↓, 2-propenyl isothiocyanate ↑ and 1-cyano-2,3-epithiopropane ↓ after 3 min of heating, where nitriles accounted for 99.5% after 120 min of heat treatment[121]
kohlrabi0.85 µmol/g FW of nitriles, 0.34 µmol/g FW ITCs and no ETNsaqueous heat treatment at 100 °CUHPLC, GC-MS4-(methylthio)pentanenitrile ↓ and 4-(methylthio)butyl isothiocyanate ↑ after 3 min of heating, where nitriles accounted for 99% after 120 min of heat treatment[121]
red cabbagethe main GLSs were GRP, PRO, GIB and GBSheating in boiling waterHPLC-DAD-ToF-MS, GC-MSGLS degradation products included 3-butenenitrile, 5-(methylsulfinyl) pentanenitrile, indole-3-acetonitrile, 4-pentenenitrile, 3-phenylpropanenitrile and 1-cyano-2,3-epithiopropane. Formation of the corresponding nitriles increased over time, and ITCs did not accumulate in broths during boiling.[126]
kohlrabithe main GLSs were GER followed by glucoiberverin and smaller amounts of GIB, GRP and several indolic GLSsheating in boiling waterHPLC-DAD-ToF-MS, GC-MSDegradation products included 5-(methylthio) pentanenitrile, 4-(methylthio)butanenitrile, 4-(methylsulfinyl)butanenitrile, 3-(methylthio) propyl ITC and 4-(methylthio) butyl ITC. The relative ITC concentration steadily declined, and the corresponding nitriles increased over heating time.[126]
red cabbagehigh content of SIN, GIB and GRP as well as GBS and low amounts of GNA, glucoiberverin, GER and gluconasturtiin (GNS)freshly prepared homogenates incubated for 1 hUHPLC-DAD-TOF-MS, GC-MSGLS hydrolysis product differed depending on the structure, mainly including corresponding ETNs, nitrile, amine and ITCs. SIN yielded high amounts of ETNs and amine, followed by ITC.[122]
white cabbagemainly SIN and GIB as well as GBS and lower amounts of GNA, glucoiberverin, GRP and GNS.freshly prepared homogenates incubated for 1 hUHPLC-DAD-TOF-MS, GC-MSGLS hydrolysis product differed depending on their structure, mainly included corresponding ETN, nitrile, amine and ITCs. GIB and GRP yielded high amounts of nitrile, and ITC yielded and low levels of amine.[122]
broccoli seedsAliphatic GLS content was 54.5−218.7 μmol/g fresh weight, accounting for >90% of the total GLS. The major GLSs were GRP and GER in 27 samples and PRO in 7 samples.enzymatic degradation (ground and incubated at 25 °C for 2 h)HPLC, GC-FIDITC, nitrile and ETNs of SIN, GNA, GIB, GER and PRO, such as glucomesonitrile, SFN and butenylsulfuroside cyclonitrile[129]
leaf mustardcontent of SIN, GNA, PRO, GBS, 4-methoxyglucobrassicin, neoglucobrassicin and GNS ranges among the varietiesfermentation (20 °C for 4 days)HPLC, GC-MSthree ITCs, three EPNs and two CNs, including SIN-ITC, GNA-ITC, GNS-ITC, SIN-EPN, GNA-EPN, Pro-EPN, SIN-CN and GNA-CN[130]
broccoli seed extractGRP24 h of anaerobic fermentation with B. longumHPLC, UHPLC Q Exactive MSSFN, SFN−L-cysteine and erucin[53]
Note: GC-MS = gas chromatograph mass spectrometer; LC-MS = liquid chromatograph mass spectrometer; HPLC = high-performance liquid chromatography; UHPLC-DAD-TOF-MS = ultra high-performance liquid chromatography diode array detection time-of-flight mass spectrometry; ↓ = decreasing; ↑ = increasing.

5.3. Drying and Freezing

The influence of the drying treatment on the GLS content varied widely, depending on whether the high temperature environment was adopted. Luo et al. found that the total GLS content was lower in the oven-drying (OD) and vacuum-drying (VD) groups than vacuum freeze-drying (VFD). The high temperature employed in OD and VD could facilitate the thermal degradation and hydrolysis of GLS by myrosinase [124]. Quick-freezing (QF) and VFD maintained both the GLS content and the activity of myrosinase well.

5.4. Fermentation

Apart from the plant myrosinase, microorganisms also can degrade GLSs. After fermenting at 20 °C for 4 days, the GLS metabolites included three ITCs, three EPNs, and two CNs in leaf mustard [130]. Fermentation of broccoli seed extract (BSE) by B. longum released SFN, sulforaphane−L-cysteine and erucin [53]. Consequently, the combined treatment of B. longum and BSE relieved the colitis symptoms and colonic inflammatory activity of the mice model, which could be attributed to the combined intervention improving the contents of SFN and sulforaphane−N-acetylcysteine in mice colons compared with the BSE intervention alone.

5.5. Other Observations

Moreover, the extent of GLS degradation during processing depends on the concentration of the vegetable broth and the vegetable source [121,126]. GLS degradation declined with a declining vegetable concentration after long boiling treatment [126]. According to the GLS stability, white and red cabbages are more suitable for processing than other cruciferous vegetables [124]. It was suggested that short cooking times are more appropriate for broccoli and red radish root for fresh eating.

6. Conclusions and Perspectives

This review focused on the physiological functions of GLSs and their metabolites in cruciferous vegetables, highlighting their roles in modulating microbiota, antioxidation, anti-inflammation, maintaining the intestinal barrier, and regulating epigenetics to alleviate IBD symptoms. Dietary preparations mainly composed of cruciferous vegetables appear to be a promising intervention strategy for IBD, especially UC.
However, some issues still need to be addressed in the future. The main aspects include the following. (1) There were differences in intestinal microbiota among the colitis experiments, resulting in difficulty in the analysis of colitis-related microbiota and suffering bias due to technical differences. Therefore, high-quality animal experiments and even clinical trials can be employed to obtain more consistent conclusions. (2) Due to the low bioavailability and poor stability of GLSs and their metabolites, micro- and nanocapsules could be applied to improve their stability and bioavailability, but there is limited research in this area thus far. (3) SFN, DIM, and other substances have been shown to regulate epigenetic mechanisms, and further studies are necessary. (4) Long-term intake of glucosinolates may cause gastrointestinal discomfort, and metabolites of glucosinolates such as nitriles have certain levels of toxicity. Thus, safety in consuming cruciferous vegetables needs to be emphasized. (5) Most studies on the intervention of IBD using GLSs and their metabolites have been conducted for animal models, and there are limited clinical trial data on human intestinal inflammation. More clinical trials are needed to obtain clinical scientific data and expand the application of GLSs and their metabolites to human health in the future.

Author Contributions

Conceptualization, N.Z. and J.M.; literature search, X.Z. and F.L.; writing—original draft preparation, J.Z. and X.Z.; writing—review and editing, J.Z. and L.G.; software, X.L.; visualization, H.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was financially supported by the Natural Science Foundation of Chongqing (grant number CSTB2022NSCQ-MSX0386) and the Scientific and Technological Research Program of Chongqing Municipal Education Commission (grant number KJQN202300216).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in the article. Further inquiries can be directed toward the corresponding authors.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Abraham, C.; Cho, J.H. Inflammatory Bowel Disease. N. Engl. J. Med. 2009, 361, 2066–2078. [Google Scholar] [CrossRef] [PubMed]
  2. Ng, S.C.; Shi, H.Y.; Hamidi, N.; Underwood, F.E.; Tang, W.; Benchimol, E.I.; Panaccione, R.; Ghosh, S.; Wu, J.C.Y.; Chan, F.K.L.; et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: A systematic review of population-based studies. Lancet 2017, 390, 2769–2778. [Google Scholar] [CrossRef]
  3. Kotze, P.G.; Underwood, F.E.; Damião, A.O.M.C.; Ferraz, J.G.P.; Saad-Hossne, R.; Toro, M.; Iade, B.; Bosques-Padilla, F.; Teixeira, F.V.; Juliao-Banos, F.; et al. Progression of Inflammatory Bowel Diseases Throughout Latin America and the Caribbean: A Systematic Review. Clin. Gastroenterol. Hepatol. 2020, 18, 304–312. [Google Scholar] [CrossRef] [PubMed]
  4. Park, J.; Cheon, J.H. Incidence and Prevalence of Inflammatory Bowel Disease across Asia. Yonsei Med. J. 2021, 62, 99–108. [Google Scholar] [CrossRef] [PubMed]
  5. Alatab, S.; Sepanlou, S.G.; Ikuta, K.; Vahedi, H.; Bisignano, C.; Safiri, S.; Sadeghi, A.; Nixon, M.R.; Abdoli, A.; Abolhassani, H. The global, regional, and national burden of inflammatory bowel disease in 195 countries and territories, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol. Hepatol. 2020, 5, 17–30. [Google Scholar] [CrossRef]
  6. Singh, S.; Boland, B.S.; Jess, T.; Moore, A.A. Management of inflammatory bowel diseases in older adults. Lancet Gastroenterol. Hepatol. 2023, 8, 368–382. [Google Scholar] [CrossRef]
  7. Wei, L.-Y.; Zhang, J.-K.; Zheng, L.; Chen, Y. The functional role of sulforaphane in intestinal inflammation: A review. Food Funct. 2022, 13, 514–529. [Google Scholar] [CrossRef]
  8. Wang, D.; Zhang, X.; Du, H.; Psychiatry, B. Inflammatory bowel disease: A potential pathogenic factor of Alzheimer’s disease. Prog. Neuro-Psychopharmacol. 2022, 119, 110610. [Google Scholar] [CrossRef]
  9. Rogler, G. Gastrointestinal and liver adverse effects of drugs used for treating IBD. Best Pract. Res. Clin. Gastroenterol. 2010, 24, 157–165. [Google Scholar] [CrossRef]
  10. Fitzpatrick, J.A.; Melton, S.L.; Yao, C.K.; Gibson, P.R.; Halmos, E.P. Dietary management of adults with IBD—The emerging role of dietary therapy. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 652–669. [Google Scholar] [CrossRef]
  11. Chen, G.; Li, Y.; Li, X.; Zhou, D.; Wang, Y.; Wen, X.; Wang, C.; Liu, X.; Feng, Y.; Li, B.; et al. Functional foods and intestinal homeostasis: The perspective of in vivo evidence. Trends Food Sci. Technol. 2021, 111, 475–482. [Google Scholar] [CrossRef]
  12. Li, Y.-Z.; Yang, Z.-Y.; Gong, T.-T.; Liu, Y.-S.; Liu, F.-H.; Wen, Z.-Y.; Li, X.-Y.; Gao, C.; Luan, M.; Zhao, Y.-H.; et al. Cruciferous vegetable consumption and multiple health outcomes: An umbrella review of 41 systematic reviews and meta-analyses of 303 observational studies. Food Funct. 2022, 13, 4247–4259. [Google Scholar] [CrossRef] [PubMed]
  13. Bischoff, S.C.; Bager, P.; Escher, J.; Forbes, A.; Hebuterne, X.; Hvas, C.L.; Joly, F.; Klek, S.; Krznaric, Z.; Ockenga, J.; et al. ESPEN guideline on Clinical Nutrition in inflammatory bowel disease. Clin. Nutr. 2023, 42, 352–379. [Google Scholar] [CrossRef] [PubMed]
  14. Hou, J.K.; Abraham, B.; El-Serag, H. Dietary intake and risk of developing inflammatory bowel disease: A systematic review of the literature. Am. J. Gastroenterol. 2011, 106, 563–573. [Google Scholar] [CrossRef]
  15. Zhang, T.; Holman, J.; McKinstry, D.; Trindade, B.C.; Eaton, K.A.; Mendoza-Castrejon, J.; Ho, S.; Wells, E.; Yuan, H.; Wen, B.; et al. A steamed broccoli sprout diet preparation that reduces colitis via the gut microbiota. J. Nutr. Biochem. 2023, 112, 109215. [Google Scholar] [CrossRef]
  16. Li, N.; Wu, X.; Zhuang, W.; Wu, C.; Rao, Z.; Du, L.; Zhou, Y. Cruciferous vegetable and isothiocyanate intake and multiple health outcomes. Food Chem. 2022, 375, 131816. [Google Scholar] [CrossRef]
  17. Lv, Q.; Li, X.; Fan, B.; Zhu, C.; Chen, Z. The Cellular and Subcellular Organization of the Glucosinolate-Myrosinase System against Herbivores and Pathogens. Int. J. Mol. Sci. 2022, 23, 1577. [Google Scholar] [CrossRef]
  18. Suzuki, C.; Ohnishi-Kameyama, M.; Sasaki, K.; Murata, T.; Yoshida, M. Behavior of Glucosinolates in Pickling Cruciferous Vegetables. J. Agric. Food Chem. 2006, 54, 9430–9436. [Google Scholar] [CrossRef]
  19. Li, Z.; Zheng, S.; Liu, Y.; Fang, Z.; Yang, L.; Zhuang, M.; Zhang, Y.; Lv, H.; Wang, Y.; Xu, D. Characterization of glucosinolates in 80 broccoli genotypes and different organs using UHPLC-Triple-TOF-MS method. Food Chem. 2021, 334, 127519. [Google Scholar] [CrossRef]
  20. Hwang, I.M.; Park, B.; Dang, Y.M.; Kim, S.-Y.; Seo, H.Y. Simultaneous direct determination of 15 glucosinolates in eight Brassica species by UHPLC-Q-Orbitrap-MSN. Food Chem. 2019, 282, 127–133. [Google Scholar] [CrossRef]
  21. Park, C.H.; Park, S.-Y.; Park, Y.J.; Kim, J.K.; Park, S.U. Metabolite Profiling and Comparative Analysis of Secondary Metabolites in Chinese Cabbage, Radish, and Hybrid xBrassicoraphanus. J. Agric. Food Chem. 2020, 68, 13711–13719. [Google Scholar] [CrossRef] [PubMed]
  22. Sun, B.; Tian, Y.-X.; Jiang, M.; Yuan, Q.; Chen, Q.; Zhang, Y.; Luo, Y.; Zhang, F.; Tang, H.-R. Variation in the main health-promoting compounds and antioxidant activity of whole and individual edible parts of baby mustard (Brassica juncea var. gemmifera). RSC Adv. 2018, 8, 33845–33854. [Google Scholar] [CrossRef] [PubMed]
  23. Bhat, R.; Vyas, D. Myrosinase: Insights on structural, catalytic, regulatory, and environmental interactions. Crit. Rev. Biotechnol. 2019, 39, 508–523. [Google Scholar] [CrossRef] [PubMed]
  24. Sikorska-Zimny, K.; Beneduce, L. The Metabolism of Glucosinolates by Gut Microbiota. Nutrients 2021, 13, 2750. [Google Scholar] [CrossRef] [PubMed]
  25. Sun, J.; Charron, C.S.; Novotny, J.A.; Peng, B.; Yu, L.; Chen, P. Profiling glucosinolate metabolites in human urine and plasma after broccoli consumption using non-targeted and targeted metabolomic analyses. Food Chem. 2020, 309, 125660. [Google Scholar] [CrossRef]
  26. Martins, T.; Ferreira, T.; Colaço, B.; Medeiros-Fonseca, B.; Pinto, M.d.L.; Barros, A.N.; Venâncio, C.; Rosa, E.; Antunes, L.M.; Oliveira, P.A.; et al. Absorption and Excretion of Glucosinolates and Isothiocyanates after Ingestion of Broccoli (Brassica oleracea L. var italica) Leaf Flour in Mice: A Preliminary Study. Nutraceuticals 2023, 3, 540–555. [Google Scholar] [CrossRef]
  27. Liou, C.S.; Sirk, S.J.; Diaz, C.A.C.; Klein, A.P.; Fischer, C.R.; Higginbottom, S.K.; Erez, A.; Donia, M.S.; Sonnenburg, J.L.; Sattely, E.S. A Metabolic Pathway for Activation of Dietary Glucosinolates by a Human Gut Symbiont. Cell 2020, 180, 717–728.e719. [Google Scholar] [CrossRef]
  28. Shakour, Z.T.; Shehab, N.G.; Gomaa, A.S.; Wessjohann, L.A.; Farag, M.A. Metabolic and biotransformation effects on dietary glucosinolates, their bioavailability, catabolism and biological effects in different organisms. Biotechnol. Adv. 2022, 54, 107784. [Google Scholar] [CrossRef]
  29. Zhang, Y.; Tan, L.; Li, C.; Wu, H.; Ran, D.; Zhang, Z. Sulforaphane alter the microbiota and mitigate colitis severity on mice ulcerative colitis induced by DSS. AMB Express 2020, 10, 119. [Google Scholar] [CrossRef]
  30. Yuanfeng, W.; Chengzhi, L.; Ligen, Z.; Juan, S.; Xinjie, S.; Yao, Z.; Jianwei, M. Approaches for enhancing the stability and formation of sulforaphane. Food Chem. 2021, 345, 128771. [Google Scholar] [CrossRef]
  31. Liu, X.; Wang, Y.; Hoeflinger, J.L.; Neme, B.P.; Jeffery, E.H.; Miller, M.J. Dietary Broccoli Alters Rat Cecal Microbiota to Improve Glucoraphanin Hydrolysis to Bioactive Isothiocyanates. Nutrients 2017, 9, 262. [Google Scholar] [CrossRef] [PubMed]
  32. Pei, L.-y.; Ke, Y.-s.; Zhao, H.-h.; Wang, L.; Jia, C.; Liu, W.-z.; Fu, Q.-h.; Shi, M.-n.; Cui, J.; Li, S.-c. Role of colonic microbiota in the pathogenesis of ulcerative colitis. BMC Gastroenterol. 2019, 19, 10. [Google Scholar] [CrossRef] [PubMed]
  33. Kaczmarek, J.L.; Liu, X.; Charron, C.S.; Novotny, J.A.; Jeffery, E.H.; Seifried, H.E.; Ross, S.A.; Miller, M.J.; Swanson, K.S.; Holscher, H.D. Broccoli consumption affects the human gastrointestinal microbiota. J. Nutr. Biochem. 2019, 63, 27–34. [Google Scholar] [CrossRef]
  34. Coscueta, E.R.; Sousa, A.S.; Reis, C.A.; Pintado, M.M. Phenylethyl Isothiocyanate: A Bioactive Agent for Gastrointestinal Health. Molecules 2022, 27, 794. [Google Scholar] [CrossRef]
  35. El Badawy, S.A.; Ogaly, H.A.; Abd-Elsalam, R.M.; Azouz, A.A. Benzyl isothiocyanates modulate inflammation, oxidative stress, and apoptosis via Nrf2/HO-1 and NF-κB signaling pathways on indomethacin-induced gastric injury in rats. Food Funct. 2021, 12, 6001–6013. [Google Scholar] [CrossRef]
  36. Eichele, D.D.; Kharbanda, K.K. Dextran sodium sulfate colitis murine model: An indispensable tool for advancing our understanding of inflammatory bowel diseases pathogenesis. World J. Gastroenterol. 2017, 23, 6016. [Google Scholar] [CrossRef]
  37. Wagner, A.E.; Will, O.; Sturm, C.; Lipinski, S.; Rosenstiel, P.; Rimbach, G. DSS-induced acute colitis in C57BL/6 mice is mitigated by sulforaphane pre-treatment. J. Nutr. Biochem. 2013, 24, 2085–2091. [Google Scholar] [CrossRef] [PubMed]
  38. Sun, Y.; Tang, J.; Li, C.; Liu, J.; Liu, H. Sulforaphane attenuates dextran sodium sulphate induced intestinal inflammation via IL-10/STAT3 signaling mediated macrophage phenotype switching. Food Sci. Hum. Wellness 2022, 11, 129–142. [Google Scholar] [CrossRef]
  39. Wang, Y.; Jeffery, E.H.; Miller, M.J.; Wallig, M.A.; Wu, Y. Lightly cooked broccoli is as effective as raw broccoli in mitigating dextran sulfate sodium-induced colitis in mice. Nutrients 2018, 10, 748. [Google Scholar] [CrossRef]
  40. Tian, Q.; Xu, Z.; Sun, Q.; Iniguez, A.B.; Du, M.; Zhu, M.J. Broccoli-Derived Glucoraphanin Activates AMPK/PGC1α/NRF2 Pathway and Ameliorates Dextran-Sulphate-Sodium-Induced Colitis in Mice. Antioxidants 2022, 11, 2404. [Google Scholar] [CrossRef]
  41. He, C.; Gao, M.; Zhang, X.; Lei, P.; Yang, H.; Qing, Y.; Zhang, L. The Protective Effect of Sulforaphane on Dextran Sulfate Sodium-Induced Colitis Depends on Gut Microbial and Nrf2-Related Mechanism. Front. Nutr. 2022, 9, 893344. [Google Scholar] [CrossRef] [PubMed]
  42. Alattar, A.; Alshaman, R.; Al-Gayyar, M.M.H. Therapeutic effects of sulforaphane in ulcerative colitis: Effect on antioxidant activity, mitochondrial biogenesis and DNA polymerization. Redox Rep. 2022, 27, 128–138. [Google Scholar] [CrossRef] [PubMed]
  43. Busbee, P.B.; Menzel, L.; Alrafas, H.R.; Dopkins, N.; Becker, W.; Miranda, K.; Tang, C.; Chatterjee, S.; Singh, U.P.; Nagarkatti, M.; et al. Indole-3-carbinol prevents colitis and associated microbial dysbiosis in an IL-22-dependent manner. JCI Insight 2020, 5, 127551. [Google Scholar] [CrossRef] [PubMed]
  44. Kim, J.Y.; Le, T.A.N.; Lee, S.Y.; Song, D.G.; Hong, S.C.; Cha, K.H.; Lee, J.W.; Pan, C.H.; Kang, K.e. 3,3′-Diindolylmethane Improves Intestinal Permeability Dysfunction in Cultured Human Intestinal Cells and the Model Animal Caenorhabditis elegans. J. Agric. Food Chem. 2019, 67, 9277–9285. [Google Scholar] [CrossRef]
  45. Kim, M.W.; Choi, S.; Kim, S.Y.; Yoon, Y.S.; Kang, J.-H.; Oh, S.H. Allyl Isothiocyanate Ameliorates Dextran Sodium Sulfate-Induced Colitis in Mouse by Enhancing Tight Junction and Mucin Expression. Int. J. Mol. Sci. 2018, 19, 2025. [Google Scholar] [CrossRef]
  46. Hubbard, T.D.; Murray, I.A.; Nichols, R.G.; Cassel, K.; Podolsky, M.; Kuzu, G.; Tian, Y.; Smith, P.; Kennett, M.J.; Patterson, A.D.; et al. Dietary broccoli impacts microbial community structure and attenuates chemically induced colitis in mice in an Ah receptor dependent manner. J. Funct. Foods 2017, 37, 685–698. [Google Scholar] [CrossRef]
  47. Holcomb, L.; Holman, J.M.; Hurd, M.; Lavoie, B.; Colucci, L.; Hunt, B.; Hunt, T.; Kinney, M.; Pathak, J.; Mawe, G.M.; et al. Early life exposure to broccoli sprouts confers stronger protection against enterocolitis development in an immunological mouse model of inflammatory bowel disease. bioRxiv Prepr. Serv. Biol. 2023, 8, e00688-23. [Google Scholar] [CrossRef]
  48. Wilson, E.J.; Natesh, N.S.; Ghadermazi, P.; Pothuraju, R.; Prajapati, D.R.; Pandey, S.; Kaifi, J.T.; Dodam, J.R.; Bryan, J.N.; Lorson, C.L.; et al. Red Cabbage Juice-Mediated Gut Microbiota Modulation Improves Intestinal Epithelial Homeostasis and Ameliorates Colitis. Int. J. Mol. Sci. 2024, 25, 539. [Google Scholar] [CrossRef]
  49. Mueller, K.; Blum, N.M.; Mueller, A.S. Examination of the Anti-Inflammatory, Antioxidant, and Xenobiotic-Inducing Potential of Broccoli Extract and Various Essential Oils during a Mild DSS-Induced Colitis in Rats. ISRN Gastroenterol. 2013, 2013, 710856. [Google Scholar] [CrossRef]
  50. Wu, J.; Guo, W.; Cui, S.; Tang, X.; Zhang, Q.; Lu, W.; Jin, Y.; Zhao, J.; Mao, B.; Chen, W. Broccoli seed extract rich in polysaccharides and glucoraphanin ameliorates DSS-induced colitis via intestinal barrier protection and gut microbiota modulation in mice. J. Sci. Food Agric. 2023, 103, 1749–1760. [Google Scholar] [CrossRef]
  51. Kim, Y.; Wu, A.G.; Jaja-Chimedza, A.; Graf, B.L.; Waterman, C.; Verzi, M.P.; Raskin, I. Isothiocyanate-enriched moringa seed extract alleviates ulcerative colitis symptoms in mice. PLoS ONE 2017, 12, e0184709. [Google Scholar] [CrossRef] [PubMed]
  52. Lucarini, E.; Micheli, L.; Pagnotta, E.; Matteo, R.; Parisio, C.; Toti, A.; Ferrara, V.; Ciampi, C.; Martelli, A.; Testai, L.; et al. Beneficial Effects of Eruca sativa Defatted Seed Meal on Visceral Pain and Intestinal Damage Resulting from Colitis in Rats. Foods 2022, 11, 580. [Google Scholar] [CrossRef] [PubMed]
  53. Wu, J.; Cui, S.; Tang, X.; Zhang, Q.; Jin, Y.; Zhao, J.; Mao, B.; Zhang, H. Bifidobacterium longum CCFM1206 Promotes the Biotransformation of Glucoraphanin to Sulforaphane That Contributes to Amelioration of Dextran-Sulfate-Sodium-Induced Colitis in Mice. J. Agric. Food Chem. 2023, 71, 1100–1112. [Google Scholar] [CrossRef] [PubMed]
  54. Davaatseren, M.; Hwang, J.-T.; Park, J.H.; Kim, M.-S.; Wang, S.; Sung, M.J. Allyl Isothiocyanate Ameliorates Angiogenesis and Inflammation in Dextran Sulfate Sodium-Induced Acute Colitis. PLoS ONE 2014, 9, e102975. [Google Scholar] [CrossRef]
  55. Jeon, E.J.; Davaatseren, M.; Hwang, J.T.; Park, J.H.; Hur, H.J.; Lee, A.S.; Sung, M.J. Effect of Oral Administration of 3,3′-Diindolylmethane on Dextran Sodium Sulfate-Induced Acute Colitis in Mice. J. Agric. Food Chem. 2016, 64, 7702–7709. [Google Scholar] [CrossRef]
  56. Nishida, A.; Inoue, R.; Inatomi, O.; Bamba, S.; Naito, Y.; Andoh, A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin. J. Gastroenterol. 2018, 11, 1–10. [Google Scholar] [CrossRef]
  57. Vestergaard, M.V.; Allin, K.H.; Eriksen, C.; Zakerska-Banaszak, O.; Arasaradnam, R.P.; Alam, M.T.; Kristiansen, K.; Brix, S.; Jess, T. Gut microbiota signatures in inflammatory bowel disease. United Eur. Gastroent. 2024, 12, 22–33. [Google Scholar] [CrossRef]
  58. Zhang, Y.; Jiang, C.; Huang, S.; Sun, J.; Song, X.; Nishanbaev, S.Z.; Benito, M.J.; Wu, Y. Effects of Polyphenols and Glucosinolates in Broccoli Extract on Human Gut Microorganisms Based on Simulation In Vitro. ACS Omega 2022, 7, 45096–45106. [Google Scholar] [CrossRef]
  59. He, C.; Huang, L.; Lei, P.; Liu, X.; Li, B.; Shan, Y. Sulforaphane normalizes intestinal flora and enhances gut barrier in mice with BBN-induced bladder cancer. Mol. Nutr. Food Res. 2018, 62, 1800427. [Google Scholar] [CrossRef]
  60. Zhang, Z.; Zhang, H.; Chen, T.; Shi, L.; Wang, D.; Tang, D. Regulatory role of short-chain fatty acids in inflammatory bowel disease. Cell Commun. Signal. 2022, 20, 1–10. [Google Scholar] [CrossRef]
  61. Peng, K.; Xia, S.; Xiao, S.; Yu, Q. Short-chain fatty acids affect the development of inflammatory bowel disease through intestinal barrier, immunology, and microbiota: A promising therapy? J. Gastroenterol. Hepatol. 2022, 37, 1710–1718. [Google Scholar] [CrossRef] [PubMed]
  62. Jaworska, K.; Konop, M.; Bielinska, K.; Hutsch, T.; Dziekiewicz, M.; Banaszkiewicz, A.; Ufnal, M. Inflammatory bowel disease is associated with increased gut-to-blood penetration of short-chain fatty acids: A new, non-invasive marker of a functional intestinal lesion. Exp. Physiol. 2019, 104, 1226–1236. [Google Scholar] [CrossRef]
  63. Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A.N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T.W.; Andrews, E.; Ajami, N.J.; Bonham, K.S.; Brislawn, C.J.; et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569, 655–662. [Google Scholar] [CrossRef]
  64. Kaczmarczyk, O.; Dąbek-Drobny, A.; Woźniakiewicz, M.; Paśko, P.; Dobrowolska-Iwanek, J.; Woźniakiewicz, A.; Piątek-Guziewicz, A.; Zagrodzki, P.; Mach, T.; Zwolińska-Wcisło, M. Fecal levels of lactic, succinic and short-chain fatty acids in patients with ulcerative colitis and crohn disease: A pilot study. J. Clin. Med. 2021, 10, 4701. [Google Scholar] [CrossRef] [PubMed]
  65. Rhimi, S.; Kriaa, A.; Mariaule, V.; Saidi, A.; Drut, A.; Jablaoui, A.; Akermi, N.; Maguin, E.; Hernandez, J.; Rhimi, M. The Nexus of Diet, Gut Microbiota and Inflammatory Bowel Diseases in Dogs. Metabolites 2022, 12, 1176. [Google Scholar] [CrossRef]
  66. Li, G.; Lin, J.; Zhang, C.; Gao, H.; Lu, H.; Gao, X.; Zhu, R.; Li, Z.; Li, M.; Liu, Z. Microbiota metabolite butyrate constrains neutrophil functions and ameliorates mucosal inflammation in inflammatory bowel disease. Gut Microbes 2021, 13, 1968257. [Google Scholar] [CrossRef] [PubMed]
  67. Li, X.; Wang, C.; Zhu, J.; Lin, Q.; Yu, M.; Wen, J.; Feng, J.; Hu, C. Sodium butyrate ameliorates oxidative stress-induced intestinal epithelium barrier injury and mitochondrial damage through AMPK-mitophagy pathway. Oxid. Med. Cell. Longev. 2022, 2022, 3745135. [Google Scholar] [CrossRef] [PubMed]
  68. Tian, T.; Wang, Z.; Zhang, J. Pathomechanisms of Oxidative Stress in Inflammatory Bowel Disease and Potential Antioxidant Therapies. Oxid. Med. Cell. Longev. 2017, 2017, 4535194. [Google Scholar] [CrossRef]
  69. Guan, G.; Lan, S. Implications of Antioxidant Systems in Inflammatory Bowel Disease. BioMed Res. Int. 2018, 2018, 1290179. [Google Scholar] [CrossRef]
  70. Collins, D.; Hogan, A.M.; Winter, D.C. Microbial and viral pathogens in colorectal cancer. Lancet Oncol. 2011, 12, 504–512. [Google Scholar] [CrossRef]
  71. Chodur, G.M.; Olson, M.E.; Wade, K.L.; Stephenson, K.K.; Nouman, W.; Garima; Fahey, J.W. Wild and domesticated Moringa oleifera differ in taste, glucosinolate composition, and antioxidant potential, but not myrosinase activity or protein content. Sci. Rep. 2018, 8, 7995. [Google Scholar] [CrossRef]
  72. Zhang, Y.-J.; Wu, Q. Sulforaphane protects intestinal epithelial cells against lipopolysaccharide-induced injury by activating the AMPK/SIRT1/PGC-1ɑ pathway. Bioengineered 2021, 12, 4349–4360. [Google Scholar] [CrossRef] [PubMed]
  73. Cedrowski, J.; Dąbrowa, K.; Przybylski, P.; Krogul-Sobczak, A.; Litwinienko, G. Antioxidant activity of two edible isothiocyanates: Sulforaphane and erucin is due to their thermal decomposition to sulfenic acids and methylsulfinyl radicals. Food Chem. 2021, 353, 129213. [Google Scholar] [CrossRef] [PubMed]
  74. Li, D.; Shao, R.; Wang, N.; Zhou, N.; Du, K.; Shi, J.; Wang, Y.; Zhao, Z.; Ye, X.; Zhang, X. Sulforaphane activates a lysosome-dependent transcriptional program to mitigate oxidative stress. Autophagy 2021, 17, 872–887. [Google Scholar] [CrossRef] [PubMed]
  75. Wen, Z.; Liu, W.; Li, X.; Chen, W.; Liu, Z.; Wen, J.; Liu, Z. A Protective Role of the NRF2-Keap1 Pathway in Maintaining Intestinal Barrier Function. Oxid. Med. Cell. Longev. 2019, 2019, 1759149. [Google Scholar] [CrossRef] [PubMed]
  76. Egbujor, M.C.; Petrosino, M.; Zuhra, K.; Saso, L. The role of organosulfur compounds as Nrf2 activators and their antioxidant effects. Antioxidants 2022, 11, 1255. [Google Scholar] [CrossRef]
  77. Petkovic, M.; Leal, E.C.; Alves, I.; Bose, C.; Palade, P.T.; Singh, P.; Awasthi, S.; Børsheim, E.; Dalgaard, L.T.; Singh, S.P. Dietary supplementation with sulforaphane ameliorates skin aging through activation of the Keap1-Nrf2 pathway. J. Nutr. Biochem. 2021, 98, 108817. [Google Scholar] [CrossRef]
  78. Clifford, T.; Acton, J.P.; Cocksedge, S.P.; Davies, K.A.B.; Bailey, S.J. The effect of dietary phytochemicals on nuclear factor erythroid 2-related factor 2 (Nrf2) activation: A systematic review of human intervention trials. Mol. Biol. Rep. 2021, 48, 1745–1761. [Google Scholar] [CrossRef]
  79. Bourgonje, A.R.; Kloska, D.; Grochot-Przęczek, A.; Feelisch, M.; Cuadrado, A.; van Goor, H. Personalized redox medicine in inflammatory bowel diseases: An emerging role for HIF-1α and NRF2 as therapeutic targets. Redox Biol. 2023, 60, 102603. [Google Scholar] [CrossRef]
  80. Cheng, X.-R.; Yu, B.-T.; Song, J.; Ma, J.-H.; Chen, Y.-Y.; Zhang, C.-X.; Tu, P.-H.; Muskat, M.N.; Zhu, Z.-G. The Alleviation of Dextran Sulfate Sodium (DSS)-Induced Colitis Correlate with the log P Values of Food-Derived Electrophilic Compounds. Antioxidants 2022, 11, 2406. [Google Scholar] [CrossRef]
  81. Melim, C.; Lauro, M.R.; Pires, I.M.; Oliveira, P.J.; Cabral, C. The role of glucosinolates from cruciferous vegetables (Brassicaceae) in gastrointestinal cancers: From prevention to therapeutics. Pharmaceutics 2022, 14, 190. [Google Scholar] [CrossRef] [PubMed]
  82. Ruhee, R.T.; Suzuki, K. The integrative role of sulforaphane in preventing inflammation, oxidative stress and fatigue: A review of a potential protective phytochemical. Antioxidants 2020, 9, 521. [Google Scholar] [CrossRef] [PubMed]
  83. Holman, J.; Hurd, M.; Moses, P.L.; Mawe, G.M.; Zhang, T.; Ishaq, S.L.; Li, Y. Interplay of broccoli/broccoli sprout bioactives with gut microbiota in reducing inflammation in inflammatory bowel diseases. J. Nutr. Biochem. 2023, 113, 109238. [Google Scholar] [CrossRef] [PubMed]
  84. Polinska, B.; Matowicka-Karna, J.; Kemona, H. The cytokines in inflammatory bowel disease. Postep. Hig. Med. Dosw. 2009, 63, 389–394. [Google Scholar]
  85. Miller, C.A. Expression of the human aryl hydrocarbon receptor complex in yeast—Activation of transcription by indole compounds. J. Biol. Chem. 1997, 272, 32824–32829. [Google Scholar] [CrossRef]
  86. Neavin, D.R.; Liu, D.; Ray, B.; Weinshilboum, R.M. The role of the aryl hydrocarbon receptor (AHR) in immune and inflammatory diseases. Int. J. Mol. Sci. 2018, 19, 3851. [Google Scholar] [CrossRef]
  87. Peng, C.; Wu, C.; Xu, X.; Pan, L.; Lou, Z.; Zhao, Y.; Jiang, H.; He, Z.; Ruan, B. Indole-3-carbinol ameliorates necroptosis and inflammation of intestinal epithelial cells in mice with ulcerative colitis by activating aryl hydrocarbon receptor. Exp. Cell Res. 2021, 404, 112638. [Google Scholar] [CrossRef]
  88. Garcia-Ibañez, P.; Núñez-Sánchez, M.A.; Oliva-Bolarín, A.; Martínez-Sánchez, M.A.; Ramos-Molina, B.; Ruiz-Alcaraz, A.J.; Moreno, D.A. Anti-inflammatory potential of digested Brassica sprout extracts in human macrophage-like HL-60 cells. Food Funct. 2023, 14, 112–121. [Google Scholar] [CrossRef]
  89. Wu, Y.; Wang, J.; He, Q.; Yu, L.; Pham, Q.; Cheung, L.; Zhang, Z.; Kim, Y.S.; Smith, A.D.; Wang, T.T. Dietary indole-3-carbinol alleviated spleen Enlargement, enhanced IgG response in C3H/HeN mice infected with Citrobacter rodentium. Nutrients 2020, 12, 3148. [Google Scholar] [CrossRef]
  90. Abu-Qatouseh, L. Sulforaphane from broccoli attenuates inflammatory hepcidin by reducing IL-6 secretion in human HepG2 cells. J. Funct. Foods 2020, 75, 104210. [Google Scholar]
  91. Ruhee, R.T.; Ma, S.; Suzuki, K. Sulforaphane protects cells against lipopolysaccharide-stimulated inflammation in murine macrophages. Antioxidants 2019, 8, 577. [Google Scholar] [CrossRef] [PubMed]
  92. Subedi, L.; Lee, J.H.; Yumnam, S.; Ji, E.; Kim, S.Y. Anti-Inflammatory Effect of Sulforaphane on LPS-Activated Microglia Potentially through JNK/AP-1/NF-kappa B Inhibition and Nrf2/HO-1 Activation. Cells 2019, 8, 194. [Google Scholar] [CrossRef] [PubMed]
  93. Mahn, A.; Castillo, A. Potential of sulforaphane as a natural immune system enhancer: A review. Molecules 2021, 26, 752. [Google Scholar] [CrossRef] [PubMed]
  94. Ali, M.; Bonay, M.; Vanhee, V.; Vinit, S.; Deramaudt, T.B. Comparative effectiveness of 4 natural and chemical activators of Nrf2 on inflammation, oxidative stress, macrophage polarization, and bactericidal activity in an in vitro macrophage infection model. PLoS ONE 2020, 15, e0234484. [Google Scholar] [CrossRef]
  95. Bahiraii, S.; Brenner, M.; Yan, F.; Weckwerth, W.; Heiss, E.H. Sulforaphane diminishes moonlighting of pyruvate kinase M2 and interleukin 1β expression in M1 (LPS) macrophages. Front. Immunol. 2022, 13, 935692. [Google Scholar] [CrossRef] [PubMed]
  96. Trivedi-Parmar, V.; Jorgensen, W.L. Advances and insights for small molecule inhibition of macrophage migration inhibitory factor. J. Med. Chem. 2018, 61, 8104–8119. [Google Scholar] [CrossRef]
  97. Spencer, E.S.; Dale, E.J.; Gommans, A.L.; Rutledge, M.T.; Vo, C.T.; Nakatani, Y.; Gamble, A.B.; Smith, R.A.; Wilbanks, S.M.; Hampton, M.B. Multiple binding modes of isothiocyanates that inhibit macrophage migration inhibitory factor. Eur. J. Med. Chem. 2015, 93, 501–510. [Google Scholar] [CrossRef]
  98. Guerrero-Alonso, A.; Antunez-Mojica, M.; Medina-Franco, J.L. Chemoinformatic Analysis of Isothiocyanates: Their Impact in Nature and Medicine. Mol. Inf. 2021, 40, 2100172. [Google Scholar] [CrossRef]
  99. Li, Y.; Huang, H.; Liu, B.; Zhang, Y.; Pan, X.; Yu, X.-Y.; Shen, Z.; Song, Y.-H. Inflammasomes as therapeutic targets in human diseases. Signal Transduct. Target. Ther. 2021, 6, 247. [Google Scholar] [CrossRef]
  100. Guo, H.; Callaway, J.B.; Ting, J.P.Y. Inflammasomes: Mechanism of action, role in disease, and therapeutics. Nat. Med. 2015, 21, 677–687. [Google Scholar] [CrossRef]
  101. Kiser, C.; Gonul, C.P.; Olcum, M.; Genc, S. Inhibitory effects of sulforaphane on NLRP3 inflammasome activation. Mol. Immunol. 2021, 140, 175–185. [Google Scholar] [CrossRef] [PubMed]
  102. Zhang, J.; Fu, S.; Sun, S.; Li, Z.; Guo, B. Inflammasome activation has an important role in the development of spontaneous colitis. Mucosal Immunol. 2014, 7, 1139–1150. [Google Scholar] [CrossRef] [PubMed]
  103. Zhou, Z.-J.; Dong, J.-Y.; Qiu, Y.; Zhang, G.-L.; Wei, K.; He, L.-H.; Sun, Y.-N.; Jiang, H.-Z.; Zhang, S.-S.; Guo, X.-R.; et al. Sulforaphane decreases oxidative stress and inhibits NLRP3 inflammasome activation in a mouse model of ulcerative colitis. Biomed. Pharmacother. 2024, 175, 116706. [Google Scholar] [CrossRef] [PubMed]
  104. Park, W.S.; Lee, J.; Na, G.; Park, S.; Seo, S.-K.; Choi, J.S.; Jung, W.-K.; Choi, I.-W. Benzyl Isothiocyanate Attenuates Inflammasome Activation in Pseudomonas aeruginosa LPS-Stimulated THP-1 Cells and Exerts Regulation through the MAPKs/NF-κB Pathway. Int. J. Mol. Sci. 2022, 23, 1228. [Google Scholar] [CrossRef]
  105. Liu, T.; Zhang, L.; Joo, D.; Sun, S.-C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef]
  106. Saleh, H.A.; Ramdan, E.; Elmazar, M.M.; Azzazy, H.M.; Abdelnaser, A. Comparing the protective effects of resveratrol, curcumin and sulforaphane against LPS/IFN-γ-mediated inflammation in doxorubicin-treated macrophages. Sci. Rep. 2021, 11, 545. [Google Scholar] [CrossRef]
  107. Aggarwal, B.B.; Shishodia, S. Molecular targets of dietary agents for prevention and therapy of cancer. Biochem. Pharmacol. 2006, 71, 1397–1421. [Google Scholar] [CrossRef]
  108. Busbee, P.B.; Nagarkatti, M.; Nagarkatti, P.S. Natural indoles, indole-3-carbinol and 3, 3′-diindolymethane, inhibit T cell activation by staphylococcal enterotoxin B through epigenetic regulation involving HDAC expression. Toxicol. Appl. Pharm. 2014, 274, 7–16. [Google Scholar] [CrossRef] [PubMed]
  109. López-Chillón, M.T.; Carazo-Díaz, C.; Prieto-Merino, D.; Zafrilla, P.; Moreno, D.A.; Villaño, D. Effects of long-term consumption of broccoli sprouts on inflammatory markers in overweight subjects. Clin. Nutr. 2019, 38, 745–752. [Google Scholar] [CrossRef]
  110. Lohning, A.; Kidachi, Y.; Kamiie, K.; Sasaki, K.; Ryoyama, K.; Yamaguchi, H. 6-(methylsulfinyl) hexyl isothiocyanate (6-MITC) from Wasabia japonica alleviates inflammatory bowel disease (IBD) by potential inhibition of glycogen synthase kinase 3 beta (GSK-3β). Eur. J. Med. Chem. 2021, 216, 113250. [Google Scholar] [CrossRef]
  111. Chang, Y.; Zhai, L.; Peng, J.; Wu, H.; Bian, Z.; Xiao, H. Phytochemicals as regulators of Th17/Treg balance in inflammatory bowel diseases. Biomed. Pharmacother. 2021, 141, 111931. [Google Scholar] [CrossRef] [PubMed]
  112. Zhang, T.; Lei, F.; Jiang, T.; Xie, L.; Huang, P.; Li, P.; Huang, Y.; Tang, X.; Gong, J.; Lin, Y. H19/miR-675-5p targeting SFN enhances the invasion and metastasis of nasalpharyngeal cancer cells. Curr. Molec. Pharmacol. 2019, 12, 324–333. [Google Scholar] [CrossRef] [PubMed]
  113. Martin, S.L.; Kala, R.; Tollefsbol, T.O. Mechanisms for the inhibition of colon cancer cells by sulforaphane through epigenetic modulation of microRNA-21 and human telomerase reverse transcriptase (hTERT) down-regulation. Curr. Cancer Drug Targets 2018, 18, 97–106. [Google Scholar] [CrossRef]
  114. Gao, L.; Cheng, D.; Yang, J.; Wu, R.; Li, W.; Kong, A.-N. Sulforaphane epigenetically demethylates the CpG sites of the miR-9-3 promoter and reactivates miR-9-3 expression in human lung cancer A549 cells. J. Nutr. Biochem. 2018, 56, 109–115. [Google Scholar] [CrossRef]
  115. Nandini, D.; Rao, R.S.; Deepak, B.; Reddy, P.B. Sulforaphane in broccoli: The green chemoprevention!! Role in cancer prevention and therapy. J. Oral Maxillofac. Pathol. JOMFP 2020, 24, 405. [Google Scholar] [CrossRef]
  116. Li, Z.; Rana, T.M. Therapeutic targeting of microRNAs: Current status and future challenges. Nat. Rev. Drug Discov. 2014, 13, 622–638. [Google Scholar] [CrossRef] [PubMed]
  117. Krishnachaitanya, S.S.; Liu, M.; Fujise, K.; Li, Q. MicroRNAs in inflammatory bowel disease and its complications. Int. J. Mol. Sci. 2022, 23, 8751. [Google Scholar] [CrossRef]
  118. Jung, H.; Kim, J.S.; Lee, K.H.; Tizaoui, K.; Terrazzino, S.; Cargnin, S.; Smith, L.; Koyanagi, A.; Jacob, L.; Li, H. Roles of microRNAs in inflammatory bowel disease. Int. J. Biol. Sci. 2021, 17, 2112. [Google Scholar] [CrossRef]
  119. Dacosta, C.; Bao, Y. The role of MicroRNAs in the chemopreventive activity of sulforaphane from cruciferous vegetables. Nutrients 2017, 9, 902. [Google Scholar] [CrossRef]
  120. Wu, J.; Cui, S.; Liu, J.; Tang, X.; Zhao, J.; Zhang, H.; Mao, B.; Chen, W. The recent advances of glucosinolates and their metabolites: Metabolism, physiological functions and potential application strategies. Crit. Rev. Food Sci. 2023, 63, 4217–4234. [Google Scholar] [CrossRef]
  121. Hanschen, F.S.; Kuehn, C.; Nickel, M.; Rohn, S.; Dekker, M. Leaching and degradation kinetics of glucosinolates during boiling of Brassica oleracea vegetables and the formation of their breakdown products. Food Chem. 2018, 263, 240–250. [Google Scholar] [CrossRef] [PubMed]
  122. Andernach, L.; Witzel, K.; Hanschen, F.S. Glucosinolate-derived amine formation in Brassica oleracea vegetables. Food Chem. 2023, 405, 134907. [Google Scholar] [CrossRef] [PubMed]
  123. Sikorska-Zimny, K.; Beneduce, L. The glucosinolates and their bioactive derivatives in Brassica: A review on classification, biosynthesis and content in plant tissues, fate during and after processing, effect on the human organism and interaction with the gut microbiota. Crit. Rev. Food Sci. 2021, 61, 2544–2571. [Google Scholar] [CrossRef]
  124. Luo, S.; An, R.; Zhou, H.; Zhang, Y.; Ling, J.; Hu, H.; Li, P. The glucosinolate profiles of Brassicaceae vegetables responded differently to quick-freezing and drying methods. Food Chem. 2022, 383, 132624. [Google Scholar] [CrossRef]
  125. Bello, C.; Maldini, M.; Baima, S.; Scaccini, C.; Natella, F. Glucoraphanin and sulforaphane evolution during juice preparation from broccoli sprouts. Food Chem. 2018, 268, 249–256. [Google Scholar] [CrossRef]
  126. Renz, M.; Dekker, M.; Rohn, S.; Hanschen, F.S. Plant matrix concentration and redox status influence thermal glucosinolate stability and formation of nitriles in selected Brassica vegetable broths. Food Chem. 2023, 404, 134594. [Google Scholar] [CrossRef] [PubMed]
  127. Hoffmann, H.; Andernach, L.; Kanzler, C.; Hanschen, F.S. Novel transformation products from glucosinolate-derived thioglucose and isothiocyanates formed during cooking. Food Res. Int. 2022, 157, 111237. [Google Scholar] [CrossRef]
  128. Hoffmann, H.; Ott, C.; Raupbach, J.; Andernach, L.; Renz, M.; Grune, T.; Hanschen, F.S. Assessing Bioavailability and Bioactivity of 4-Hydroxythiazolidine-2-Thiones, Newly Discovered Glucosinolate Degradation Products Formed During Domestic Boiling of Cabbage. Front. Nutr. 2022, 9, 941286. [Google Scholar] [CrossRef]
  129. Wang, J.; Yu, H.; Zhao, Z.; Sheng, X.; Shen, Y.; Gu, H. Natural Variation of Glucosinolates and Their Breakdown Products in Broccoli (Brassica oleracea var. italica) Seeds. J. Agric. Food Chem. 2019, 67, 12528–12537. [Google Scholar] [CrossRef]
  130. Di, H.; Ma, J.; Zhang, Y.; Wei, J.; Yang, J.; Ma, J.; Bian, J.; Xu, J.; Huang, Z.; Tang, Y.; et al. Correlations between flavor and glucosinolates and changes in quality-related physiochemical characteristics of Guizhou suancai during the fermentation process. Food Chem. 2023, 405, 134965. [Google Scholar] [CrossRef]
Figure 1. The attenuating mechanism of GLSs and metabolites on IBD through anti-inflammatory and antioxidant activity, maintaining the intestinal barrier, gut microbiota modulation and modulating epigenetics.
Figure 1. The attenuating mechanism of GLSs and metabolites on IBD through anti-inflammatory and antioxidant activity, maintaining the intestinal barrier, gut microbiota modulation and modulating epigenetics.
Foods 13 03507 g001
Table 1. GLS composition and contents in common brassica vegetables.
Table 1. GLS composition and contents in common brassica vegetables.
Classification of GLSCompound NameGlucosinolate Content (μmol/g)
Kale [20]
Brassica oleracea L.
Broccoli [19]
B. oleracea L. var. Italic)
Chinese Cabbage [21]
B. rapa var. Glabra Rule)
Mustard [22]
B. juncea
aliphatic GLSglucoiberin (GIB)4.78~5.420~0.878N/AN/A
glucoberteroinN/A0.008~6.2730.35~0.38N/A
glucoraphanin (GRP)N/A0.136~14.973N/AN/A
progoitrin (PRO)0.08~0.700~4.5370.71~0.87N/A
sinigrin (SIN)1.64~1.780~3.161N/A13.95~17.67
glucoalyssinN/A0~2.7281.11~1.190.45~0.53
gluconapin (GNA)N/AN/A0.23~0.270.12~0.18
percentage (%) 84.40~85.08%27.78~48%18.93~23.20%71.56~90.69%
indole GLSglucobrasscin (GBS)0.98~1.040.103~27.6903.94~4.360.43~0.59
neoglucobrasscin0.07~0.330.018~45.9543.54~3.56N/A
glucobrassicanapinN/AN/A0.85~0.910.20~0.28
4-hydroxyglucobrassicinN/A0.014~3.2890.08~0.120.23~0.35
4-methoxyglucobrassicinN/A0.014~3.9150.96~1.120.28~0.4
percentage (%) 13.74~14.64%43.67~70.99%73.90~86.22%3.50~6.47%
aromatic GLSgluconasturtiin (GNS)N/A0.004–0.441N/A0.22~0.34
sinalbinN/AN/AN/AN/A
glucotropaeolinN/A0–0.040N/AN/A
percentage (%) 00.42~1.33%01.08~2.13%
total (μmol/g) 7.64~9.360.30~113.8811.68~12.6815.93~20.29
Note: Data in the table are measured by dry weight. N/A = undetected.
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

Zhao, J.; Zhang, X.; Li, F.; Lei, X.; Ge, L.; Li, H.; Zhao, N.; Ming, J. The Effects of Interventions with Glucosinolates and Their Metabolites in Cruciferous Vegetables on Inflammatory Bowel Disease: A Review. Foods 2024, 13, 3507. https://doi.org/10.3390/foods13213507

AMA Style

Zhao J, Zhang X, Li F, Lei X, Ge L, Li H, Zhao N, Ming J. The Effects of Interventions with Glucosinolates and Their Metabolites in Cruciferous Vegetables on Inflammatory Bowel Disease: A Review. Foods. 2024; 13(21):3507. https://doi.org/10.3390/foods13213507

Chicago/Turabian Style

Zhao, Jichun, Xiaoqin Zhang, Fuhua Li, Xiaojuan Lei, Lihong Ge, Honghai Li, Nan Zhao, and Jian Ming. 2024. "The Effects of Interventions with Glucosinolates and Their Metabolites in Cruciferous Vegetables on Inflammatory Bowel Disease: A Review" Foods 13, no. 21: 3507. https://doi.org/10.3390/foods13213507

APA Style

Zhao, J., Zhang, X., Li, F., Lei, X., Ge, L., Li, H., Zhao, N., & Ming, J. (2024). The Effects of Interventions with Glucosinolates and Their Metabolites in Cruciferous Vegetables on Inflammatory Bowel Disease: A Review. Foods, 13(21), 3507. https://doi.org/10.3390/foods13213507

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