Next Article in Journal
A High-Throughput Behavioral Assay for Screening Novel Anxiolytics in Larval Zebrafish
Previous Article in Journal
Food–Drug Interactions: Effect of Propolis on the Pharmacokinetics of Enrofloxacin and Its Active Metabolite Ciprofloxacin in Rabbits
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Garlic Peel-Derived Phytochemicals Using GC-MS: Antioxidant, Anti-Inflammatory, and Anti-Apoptotic Effects in Ulcerative Colitis Rat Model

by
Duaa A. Althumairy
1,
Rasha Abu-Khudir
2,*,
Afnan I. Alandanoosi
3 and
Gehan M. Badr
4
1
Department of Biological Sciences, College of Science, King Faisal University, P.O. Box 380, Al-Ahsa 31982, Saudi Arabia
2
Department of Chemistry, College of Science, King Faisal University, P.O. Box 380, Al-Ahsa 31982, Saudi Arabia
3
Department of Food and Nutrition Sciences, College of Agricultural and Food Sciences, King Faisal University, P.O. Box 380, Al-Ahsa 31982, Saudi Arabia
4
Department of Zoology, Faculty of Science, Ain Shams University, Cairo 11566, Egypt
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2025, 18(7), 969; https://doi.org/10.3390/ph18070969
Submission received: 3 June 2025 / Revised: 21 June 2025 / Accepted: 24 June 2025 / Published: 27 June 2025
(This article belongs to the Section Natural Products)

Abstract

Background/Objectives: Ulcerative colitis (UC) is a chronic, relapsing inflammatory bowel disease (IBD) that poses a significant gastroenterological challenge. Methods: This study investigates the protective effects of garlic peel extract (GPE) in a rat model of acetic acid (AA)-induced colitis. Rats received oral GPE (100 mg/kg) for 14 days prior to AA administration, and this continued for 14 days post-induction. Results: GC-MS analysis of GPE identified several key phytochemicals, primarily methyl esters of fatty acids (62.47%), fatty acids (10.36%), fatty acid derivatives (6.75%), and vitamins (4.86%) as the major constituents. Other notable compounds included steroids, natural alcohols, organosulfur compounds, fatty aldehydes, carotenoids, sugars, and glucosinolates. GPE treatment significantly improved body weight and colon length. Biochemical analysis showed that GPE downregulated the levels of the pro-inflammatory cytokines interleukin-1 (IL-1), IL-6, IL-17, tumor necrosis factor-alpha (TNF-α), and nuclear factor-kappa B (NF-κB), compared to the colitis (AA) group. Additionally, GPE reduced the oxidative stress (OS) biomarkers, including myeloperoxidase (MPO) and malondialdehyde (MDA), as well as caspase-3, a marker for apoptosis. Furthermore, GPE treatment resulted in enhanced activities of the enzymatic antioxidants catalase (CAT) and superoxide dismutase (SOD), along with increased levels of the anti-inflammatory cytokine IL-10. These findings were supported by histological evidence. Conclusions: Collectively, GPE holds promise as a therapeutic strategy for UC, owing to its natural bioactive compounds and their potential synergistic anti-inflammatory, antioxidant, and anti-apoptotic effects.

Graphical Abstract

1. Introduction

Ulcerative colitis (UC) is a chronic, nonspecific inflammatory bowel disease (IBD) that primarily affects the rectum and colon. It typically involves only the mucosal layer, manifesting as continuous ulcerations accompanied by an inflammatory cascade driven by dysregulated innate immune responses and an imbalance between pro- and anti-inflammatory activities [1,2]. The severity of ulceration correlates with immune activation and elevated levels of pro-inflammatory cytokines. Chronic inflammation disrupts the mucosal barrier and activates the nuclear factor-kappa B (NF-κB) signaling pathway, a central regulator of inflammatory responses. NF-κB promotes the expression of cytokines such as IL-1, IL-6, IL-17, and TNF-α, which contribute to tissue damage and mucosal injury [3,4,5,6]. In addition, infiltration of neutrophils and macrophages enhances the production of pro-inflammatory mediators, whereas it downregulates the expression of anti-inflammatory cytokines, including IL-10 [7,8,9]. Moreover, it promotes myeloperoxidase (MPO) release by increasing reactive oxygen species (ROS) levels, which are central to UC development [10,11]. In addition, the observed activation of macrophages and neutrophils in UC leads to the generation of nitric oxide (NO) that triggers lipid peroxidation (LPO) and excessive production of malondialdehyde (MDA) [7]. Oxidative stress (OS) is known to be an important trigger of apoptosis that plays a crucial role in the initiation as well as the progression of UC. This is accompanied by upregulation of apoptotic proteinssuch as caspase-3, caspase-9, and Bax, as well as downregulation of Bcl-2 [12,13,14].
The exact etiology of UC remains unclear, and current therapeutic options are limited. Conventional treatments that primarily target symptom reduction are associated with adverse side effects as well as challenging long-term safety and efficacy. These include 5-aminosalicylates (5-ASAs), corticosteroids (CSs), immunomodulators (IMMs), and biologics, which encompass inhibitors of pro-inflammatory cytokines and antagonists of integrin [15,16,17,18]. Hence, identifying alternative therapeutic modalities that are both efficient and have fewer side effects is of growing interest.
Naturally derived compounds with antioxidant and anti-inflammatory activities have gained attention as plausible alternatives to conventional therapy. Garlic (Allium sativum) possesses diverse bioactivities, including antioxidant, anti-inflammatory, antidiabetic, antihypertensive, antimicrobial, and anticancer properties. These biological activities are primarily attributed to organosulfur-containing compounds (OSCs) such as S-allyl-cysteine sulfoxide (alliin), diallyl thiosulfonate (allicin), S-allyl-L-cysteine (SAC), and diallyl sulfide (DAS) [19,20,21,22]. While much attention has been focused on the medicinal properties of garlic bulbs, less is known about the potential benefits of garlic peel, which is often discarded as waste.
Recent studies suggest that garlic peel contains bioactive compounds that may offer significant health benefits, including the modulation of immune responses and the reduction in OS, both of which are crucial in the management of UC [23,24,25,26]. Garlic peel extract (GPE) contains abundant phenolic compounds and exhibits strong antioxidant and antimicrobial activities, comparable or superior to fresh garlic extracts [25,27]. Studies have identified multiple bioactive phenols (phenylpropanoids) in garlic peels that contribute to their antioxidant potential and anticancer effects [23,27,28,29]. Given that UC is associated with altered gut microbiota, immune dysregulation, and compromised mucosal integrity [30,31], GPE’s antimicrobial and immunomodulatory effects may offer therapeutic benefits. Garlic peels also show antifungal properties and potential in modulating fungal microbiota, which are increasingly recognized in the pathology of UC [25,32]. Furthermore, garlic peels are rich in polyphenols, including flavonoids and phenolic acids that suppress COX-2 expression, reduce ROS levels, and exert anti-apoptotic effects by downregulating NF-κB and caspase-3 transcription [23,33,34].
In summary, the derived phytochemicals in GPE, analyzed using GC-MS, show promise as a natural alternative therapeutic strategy for UC due to its synergistic antioxidant, anti-inflammatory, and anti-apoptotic properties. These effects are attained via the modulation of NF-κB signaling and OS, suggesting a potential role in improving UC outcomes.

2. Results

2.1. Identification of Bioactive Compounds of GPE by GC-MS Analysis

The GC–MS analysis of garlic peel extract (GPE), as presented in Table 1 and Figure 1, identified a diverse range of phytochemical constituents. The major detected components, based on peak area percentages, were fatty acid esters (62.47%), followed by fatty acids (10.36%), fatty acid derivatives (6.75%), vitamins (4.86%), steroids (3.39%), natural alcohols (2.72%), organosulfur compounds (2.64%), fatty aldehydes (2.51%), carotenoids (2.43%), sugars (0.99%), and glucosinolates (0.80%). In addition, several minor compounds were detected, including ketone derivatives, alkanes, oximes, pyranone derivatives, lactones, benzofuran derivatives, aromatic esters, and pyrrolopyridazine derivatives. A few other compounds were excluded due to unreliable identification by the database.

2.2. Effects of Inner Garlic Peel Ethanolic Extract (GPE) on Final Body Weight and Colon Length in AA-Induced UC Rats

The final body weight (Figure 2A) and colon length (Figure 2B,C) in AA-induced UC rats (AA group) showed a highly significant decrease compared to the control (Ctrl) and the GPE-treated (GPE) groups. However, treatment of the AA-induced UC rats with GPE (GPE + AA group) resulted in a significant increase in final body weight and a highly significant increase in colon length. In spite of this, both the final body weight and colon length in the GPE + AA group were not restored completely back to normal, where a highly significant (p < 0.0001) decrease was observed in comparison with the Ctrl group.

2.3. Effects of Inner Garlic Peel Ethanolic Extract (GPE) on Pro- and Anti-Inflammatory Mediators and Transcription Factor NF-kB in AA-Induced UC Rats

The levels of pro-inflammatory mediators IL-1 (Figure 3A), IL-6 (Figure 3B), IL-17 (Figure 3C), and TNF-α (Figure 3D) in the colon tissues of the AA group were highly significantly increased compared to the Ctrl group. However, treatment with GPE resulted in a significant (IL-1) to a highly significant (IL-6, IL-17, and TNF-α) decrease in the GPE + AA group. In addition to the pro-inflammatory mediators, a highly significant increase in the levels of the anti-inflammatory mediator IL-10 was observed in the GPE + AA group in comparison with the AA group (Figure 3E). Regarding NF-κB, known to be a pivotal mediator of inflammatory response, there was a highly significant increase in its level in the AA group compared to the Ctrl group. However, it was decreased in the GPE + AA group in comparison with the AA group with a statistically highly significant difference (Figure 3F). With the exception of IL-6, the observed change in the pro- and anti-inflammatory mediators as well as NF-kB levels upon treatment of the AA rats with GPE was not sufficient to completely restore the inflammatory mediators back to normal levels, where a significant difference (p < 0.001) was observed compared to the Crtl group.

2.4. Effects of Inner Garlic Peel Ethanolic Extract (GPE) on Oxidative Stress Biomarkers in AA-Induced UC Rats

In comparison with the Ctrl group, the activities of CAT (Figure 4A) and SOD (Figure 4B) in the AA group demonstrated a highly significant decrease. On the contrary, treatment with GPE resulted in a highly significant increase in CAT and SOD activities in the GPE + AA group compared to the AA group. In addition, a highly significant increase was observed in the MDA level in the AA group compared to the Ctrl group, whereas a highly significant decrease in the MDA level was observed in the GPE + AA group in comparison with the AA group (Figure 4C). Regarding the myeloperoxidase (MPO) level, a highly significant increase was observed in the AA group compared to the Ctrl group. However, the MPO level was reduced in the GPE + AA group in comparison with the AA group, with a statistically highly significant difference (Figure 4D). The detected change in CAT and SOD activities as well as MDA and MPO levels upon treatment of the AA rats with GPE was not sufficient to completely restore the biomarkers of OS back to normal levels, where a significant difference (p < 0.0001) was observed in comparison with the Ctrl group.

2.5. Effects of Inner Garlic Peel Ethanolic Extract (GPE) on Caspase-3 Levels in AA-Induced UC Rats

In comparison with the Ctrl group, the level of caspase-3 in AA group showed a highly significant increase. On the contrary, treatment with GPE resulted in a highly significant decrease in the level of caspase-3 in the GPE + AA group compared to the AA group (Figure 5). However, the level of caspase-3 was not completely recovered back to normal levels in the GPE + AA group, where a significant difference (p < 0.001) was observed in comparison with the Ctrl group.

2.6. Effects of Inner Garlic Peel Ethanolic Extract (GPE) on Colon Histopathology in AA-Induced UC Rats

As shown in Figure 6A, the H&E-stained sections of colonic tissues of the Ctrl group showed normal histological architecture of the mucosa with intact mucosal epithelium, an abundance of goblet cells, and no signs of inflammation. The colon tissues of the rats in the AA group, on the other hand, featured an altered histology evidenced by loss of surface columnar epithelial cells, necrosis of mucosa associated with severe infiltration of mononuclear inflammatory cells, and a reduced number of goblet cells (Figure 6B). On the contrary, sections of colon tissues of the GPE group showed nearly normal structures of the mucosa, muscular layer, and submucosa (Figure 6C). The histopathological examination of the sections of colon tissues of the GPE + AA group exhibited greater healing of ulcerous areas. Additionally, slight loss of some surface columnar epithelial cells as well as minimal necrosis of the mucosa, crypts, and goblet cells were recovered to the extent that they closely resemble those in the colon tissues of the Ctrl group (Figure 6D). When the GPE + AA group was compared to the AA group, the microscopic scores of the colon tissues’ histological sections revealed a highly significant decrease (Figure 6E).

3. Discussion

Ulcerative colitis (UC) is a chronic IBD that is characterized by relapsing episodes of mucosal inflammation and epithelial injury, often leading to severe long-term complications. Although conventional therapies, including 5-ASAs, CSs, IMMs, biologics, and Janus kinase (JAK) inhibitors have proven efficacy, their use is frequently limited by adverse effects, cytotoxicity, or diminished long-term effectiveness [63,64,65,66]. Consequently, there is growing interest in exploring safer, naturally derived therapeutic alternatives.
The present study provides compelling evidence for the therapeutic potential of GPE in ameliorating the pathophysiological features of UC in a rat model. GC-MS profiling revealed that GPE is rich in bioactive compounds, particularly fatty acid methyl esters (62.47%). The major esterified fatty acids identified, including 11-octadecenoic acid, linolenic acid, oleic acid, and palmitic acid methyl esters, have well-documented anti-inflammatory and antioxidant properties [67,68,69,70]. These esters can be hydrolyzed into free fatty acids (FFAs) such as polyunsaturated (PUFAs) and monounsaturated fatty acids (MUFAs), which are known to suppress the production of pro-inflammatory cytokines and promote mucosal healing [71,72,73]. Also, GC-MS analysis revealed that the GPE is rich in omega-3 fatty acids, particularly docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), which are well known for their anti-inflammatory properties and may offer therapeutic benefits in inflammatory conditions such as IBD [48].
Moreover, vitamins identified in the extract, particularly ascorbic acid (vitamin C), have demonstrated the ability to inhibit the expression of NF-κB, COX-2, and iNOS, thereby alleviating OS and inflammation in DSS-induced colitis. The therapeutic potential of ascorbic acid in IBD is attributed not only to its strong antioxidant properties but also to its diverse biological activities [54]. As a powerful scavenger of ROS, vitamin C plays a key role in protecting the intestinal mucosa from damage [74,75].
A hallmark of UC is persistent inflammation driven by immune dysregulation and OS. In accordance with previous findings [26,33,76], our study showed that GPE treatment significantly downregulated the pro-inflammatory cytokines IL-1, IL-6, IL-17, and TNF-α, as well as NF-κB, a key player in inflammatory signaling. These cytokines are integral to both the initiation and perpetuation of the inflammatory response in IBDs, including UC [77,78]. The NF-κB signaling pathway plays a pivotal role in modulating immunological and inflammatory responses, rendering it one of the most prominent signaling pathways associated with UC [79,80,81]. Accordingly, activation of NF-kB and its downstream pro-inflammatory targets has been previously reported in AA-induced UC [82,83]. GPE treatment also led to a significant increase in IL-10, a key anti-inflammatory cytokine involved in limiting immune responses and maintaining epithelial homeostasis [84,85,86]. This suggests that GPE not only suppresses inflammation but may also promote immune resolution through the induction of regulatory cytokines.
Oxidative stress (OS) is another critical factor in UC pathogenesis [87,88]. Similar to our findings, several studies revealed signs of oxidative damage, demonstrated by decreased activities of the endogenous antioxidant enzymes CAT and SOD, as well as elevated markers of oxidative damage, including MPO activity and MDA levels, in AA-induced UC [6,83,89]. Moreover, it has been reported that the development of OS might be attributed to the excessive secretion of MPO and pro-inflammatory cytokines, such as TNF-α [90]. In the current study, GPE treatment significantly increased the activity of both CAT and SOD. On the other hand, MPO activity and MDA levels, representing LPO markers, as well as neutrophil infiltration, were significantly decreased. These findings underscore the antioxidant capacity of GPE in mitigating oxidative damage in colonic tissue. Indeed, the antioxidant activity of GPE has been previously reported [23,25,29].
In addition to the anti-inflammatory and antioxidant effects of GPE, it exhibited anti-apoptotic activity. Caspase-3, a key executioner enzyme in apoptosis, was significantly downregulated in GPE-treated groups. Apoptosis of colonic epithelial cells, frequently caused by OS, contributes to mucosal barrier disruption and the subsequent release of damage-associated molecular patterns (DAMPs), which trigger inflammation via pattern recognition receptors, including toll-like receptors (TLRs) [91,92,93]. The suppression of caspase-3 activity suggests that GPE helps preserve epithelial integrity and limits inflammation-induced tissue damage. This effect may be attributed to sulfur-containing compounds present in GPE, which possess known antioxidant and cytoprotective properties [33,34].
In the current study, histological analysis supported the biochemical findings. Colonic tissues from GPE-treated rats with colitis (GPE + AA group) showed significant healing of ulcerated mucosa, reduced inflammatory cell infiltration, and restored epithelial architecture, with only minimal residual necrosis. These results collectively demonstrate that GPE facilitates tissue regeneration by attenuating inflammation, OS, and epithelial apoptosis.
Compared to conventional UC treatments, GPE appears to offer several advantages. Commonly used for mild-to-moderate UC, 5-ASAs primarily reduce inflammation in the colonic epithelium [94] but lack significant antioxidant or anti-apoptotic properties. Corticosteroids (CSs) are effective anti-inflammatories but are generally restricted to short-term use due to severe systemic side effects that hinder their utilization for maintained remission [95,96]. GPE, with its favorable safety profile, may be more appropriate for long-term application. Immunomodulators (IMMs) suppress lymphocyte proliferation through cytotoxic mechanisms [97], while GPE appears to achieve immune modulation through natural, non-cytotoxic pathways. Moreover, it has been proposed that the use of IMMs should be limited to patients with milder UC who cannot maintain corticosteroid-free remission on 5-ASAs alone or in combination with TNF antagonists in patients with moderate-to-severe UC [98]. Biologics, such as anti-TNF-α agents, target specific inflammatory mediators [99,100], and GPE’s downregulation of TNF-α suggests it may function through a comparable, albeit broader, mechanism. Likewise, JAK inhibitors disrupt cytokine signaling at the intracellular level [101,102], whereas GPE appears to exert a more generalized anti-inflammatory effect.
Importantly, GPE’s ability to act directly on colonic tissue and reduce caspase-3 expression via localized delivery of bioactive compounds distinguishes it from conventional drugs, which often have systemic effects and broader immune suppression. This targeted action, along with its antioxidant, anti-inflammatory, and anti-apoptotic properties, highlights GPE’s potential as a safe and multifaceted therapeutic agent. A thorough comparison of the clinical characteristics and mechanisms of action of GPE with conventional UC treatments reveals significant distinctions that warrant attention (Table 2). Unlike conventional therapies, which often exhibit limited efficacy and potential toxicity, GPE presents a novel approach with the potential for more sustained therapeutic benefits.

4. Materials and Methods

4.1. Plant Material

Inner garlic peels were purchased from a private market at Al-Ahsa, Saudi Arabia. Peels were cleaned with water, dried in the shade, and then dried in an oven at no more than 50 °C until a constant weight was achieved, thus protecting sensitive active ingredients, as previously reported [103]. They were then ground to a fine powder (80-mesh sieves) and prepared to be used for ethanolic extraction.

4.2. Preparation of Garlic Peel Ethanolic Extract (GPE)

Garlic peel ethanolic extract (GPE) was prepared by soaking 400 g of garlic peel powder in ethyl alcohol (70%) overnight in the dark and then filtered through Whatman No. 1 filter paper. The filtrate was collected, dried by evaporation under reduced pressure, and then weighed [104]. The dried extract was stored in a dry sterilized airtight screw-cap tube at room temperature until further investigation. The dry mass of the concentrated extract served as the basis for all subsequent experiments.

4.3. Gas Chromatography–Mass Spectrometry (GC-MS) Analysis of GPE

The identification of the chemical composition of GPE was performed using a Trace GC1310-ISQ mass spectrometer (Thermo Scientific, Austin, TX, USA) endowed with a direct capillary column TG–5MS (30 m × 0.25 mm × 0.25 µm film thickness). Initially, the temperature of the column oven was set at 50 °C; this was followed by 5 °C/min increments until 230 °C, where it was held for 2 min. Finally, the temperature was raised to 290 °C at a rate of 30 °C/min and held for another 2 min. The temperature of both the injector and MS transfer line was retained at 250 °C and 260 °C, respectively. Helium (He), flowing at a constant rate of 1.0 mL/min, served as the carrier gas. The GC-MS setup involved the injection of 1.0 µL of diluted samples using an AS1300 autosampler in split injection mode with a 3 min solvent delay [105]. Collection of electron ionization (EI) mass spectra was carried out in full scan mode at a 70 eV ionization voltage, over a mass-to-charge ratio (m/z) range of 40–1000, and using an EI ion source set at 200 °C. The retention times and mass spectra of the identified components were compared with those of WILEY 09 and NIST 11 mass spectral libraries.

4.4. Animals

The 6–7-week-old male albino rats (150–160 g) utilized in the current study were obtained from an animal facility at King Faisal University, Saudi Arabia. The rats were maintained under environmentally controlled conditions, including a temperature of 24 ± 1 °C and a light/dark cycle of 12 h. In addition, a standard rodent diet and water were provided to the animals ad libitum. Acclimatization to such conditions was carried out over a period of 2 weeks before starting the experiment. All experimental procedures were performed in accordance with the protocol approved by the Research Ethics Committee at King Faisal University (Ref. No. KFU-REC-2025-FEB-ETHICS3047).

4.5. Ulcerative Colitis (UC) Induction

Ulcerative colitis (UC) was induced as previously reported [106]. In brief, animals were allowed access to water throughout the induction but were deprived of food 24 h before it started. Under light ether anesthesia, a medical-grade polyethylene tube was introduced 4.5 cm proximally to the anus verge and used to inject 2 mL of acid acetic solution (AA; 3% in normal saline—Sigma-Aldrich, St. Louis, MO, USA) into the colon’s lumen. The rats were maintained in a head-down position both during and for 30 s after instillation to prevent leakage of the solution. The rats in the control (Ctrl) group were exposed to the same treatment using 0.9% saline as a substitute for AA solution.

4.6. Experimental Design

Twenty-four rats were randomly divided into four groups (6 rats/group): Group I (control; Ctrl group): animals received normal saline rectally and orally for 4 weeks; Group II (Colitis; AA group): animals received a single rectal dose of acetic acid (AA) instillation; Group III (GPE group): animals received oral GPE (100 mg/kg), and at day 14, the rats were subjected to saline instillation and continued the treatment for 14 more days; and Group IV (GPE + AA group): animals received oral GPE, and at day 14, the rats were subjected to UC induction as in Group II and continued the treatment for 14 more days. Upon completion of the experiment, rats from all experimental groups were euthanized using an overdose of ether. For enzyme-linked immunosorbent analysis (ELISA), the proximal colonic segments were promptly kept at −80 °C, while the distal colonic portions were prepared for histopathological investigation.

4.7. Preparation of Colonic Tissue Homogenates

The excised colon tissues were thoroughly rinsed with ice-cold phosphate-buffered saline (PBS; 0.01 mol/L, pH 7.0–7.2—Thermo Fisher Scientific, Waltham, MA, USA) to remove any residual blood and then weighed prior to homogenization. The weighed tissues were minced into small pieces; this was followed by their homogenization on ice in 5–10 mL PBS using a glass homogenizer. The obtained suspensions were sonicated using an ultrasonic cell disrupter that further broke the cell membranes. Next, the homogenates were centrifuged at 5000× g for 15 min at 4 °C using a Thermo Scientific™ Heraeus™ Multifuge X3R centrifuge (Thermo Fisher Scientific, Waltham, MA, USA), and the resulting supernatants were further used in ELISA assays.

4.8. Immunological and Molecular Assays

ELISA kits (My Bio-Source, Inc., San Diego, CA, USA) were used to assess the levels of IL-1 (Cat. No.: MBS264984), IL-6 (Cat. No.: MBS495243), IL-10 (Cat. No.: MBS2503826), IL-17 (Cat. No.: MBS723014), TNF-α (Cat. No.: MBS267737), NF-κB (Cat. No.: MBS453975), and Caspase 3 (Cat. No.: MBS4539) according to the manufacturer’s manual.

4.9. Oxidative Stress Assays

ELISA kits (My Bio-Source, Inc., San Diego, CA, USA) were used to assess the OS biomarkers, including the activity of the endogenous antioxidant enzymes CAT (Cat. No.: MBS006963) and SOD (Cat. No.: MBS036924) as well as the levels of MPO (Cat. No.: MBS450065) as described by the manufacturer’s manual. Levels of malondialdehyde (MDA), a marker of LPO, were determined by the thiobarbituric acid (TBA) test at 532 nm as previously described [107].

4.10. Histopathological Examination

The distal portions obtained from the excised colons were processed for histopathological examination as follows: First, the tissues were fixed in a 10% formaldehyde solution (Merck, Darmstadt, Germany) for 24 h at room temperature. They were then embedded in paraffin wax and sectioned to obtain 6 µm thick paraffin sections. Staining was carried out with hematoxylin and eosin (H&E—Abcam, Cambridge, UK), and sections were subsequently examined under a light microscope (Nikon Corporation, Tokyo, Japan).
The extent of colon ulceration was assessed on the basis of the observed inflammatory response. Accordingly, the inflammatory response was quantified using a scoring system that ranged from 0 to 4, as indicated: 0, no observed inflammation; 1, a low degree of inflammation with scattered infiltrating mononuclear cells; 2, a moderate degree of inflammation with multiple infiltrating mononuclear cells and a slight loss of some surface columnar epithelial cells; 3, a high degree of inflammation together with a high degree of mononuclear cell infiltration and loss of crypt structure; 4, optimal severity of inflammation with loss of some surface columnar epithelial cells and mucosal necrosis [4].

4.11. Statistical Analysis

Data were represented as mean values ± standard deviation (SD). One-way analysis of variance (ANOVA), followed by Tukey’s multiple comparisons post hoc test, was used to analyze the statistical difference (p < 0.05) among groups using GraphPad Prism 8 Software, version 8.0.2 (GraphPad Software, San Diego, CA, USA).

5. Conclusions

To our knowledge, the findings from this study suggest for the first time that GPE could serve as a valuable adjunct or alternative to conventional therapies for UC. Its broad-spectrum biological activity, coupled with a favorable safety profile, warrants further investigation in clinical trials to confirm its efficacy in human patients.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph18070969/s1, Supplementary File S1: Zip-Document (Raw Data).

Author Contributions

Conceptualization, D.A.A., R.A.-K., A.I.A., and G.M.B.; methodology, D.A.A., R.A.-K., A.I.A., and G.M.B.; validation, D.A.A., R.A.-K., A.I.A., and G.M.B.; formal analysis, D.A.A., R.A.-K., A.I.A., and G.M.B.; resources, D.A.A., R.A.-K., A.I.A., and G.M.B.; data curation, D.A.A., R.A.-K., A.I.A., and G.M.B.; writing—original draft preparation, D.A.A., R.A.-K., and G.M.B.; writing—R.A.-K. and G.M.B.; visualization, D.A.A., R.A.-K., A.I.A., and G.M.B.; supervision, R.A.-K. and G.M.B.; project administration, D.A.A., R.A.-K., and G.M.B.; funding acquisition, D.A.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Deanship of Scientific Research, Vice Presidency for Graduate Studies and Scientific Research, King Faisal University, Saudi Arabia [Grant No. KFU252385].

Institutional Review Board Statement

This study was reviewed and approved by the Research Ethics Committee at King Faisal University (Ref. No. KFU-REC-2025-FEB-ETHICS3047; approved on 17 February 2025).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data supporting the results of the present study are included within this article and its Supplementary Materials.

Acknowledgments

The authors acknowledge the Deanship of Scientific Research, Vice Presidency for Graduate Studies and Scientific Research, King Faisal University, Saudi Arabia for providing financial support [Grant No. KFU252385].

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AAAcetic acid
5-ASAs5-aminosalicylates
CATCatalase
COX-2Cyclooxygenase-2
CSsCorticosteroids
DAMPsDamage-associated molecular patterns
DASDiallyl sulfide
DHADocosahexaenoic acid
DSSDextran sulfate sodium
EPAEicosapentaenoic acid
FFAsFree fatty acids
GPEGarlic peel extract
GC-MSGas chromatography–mass spectrometry
IMMsImmunomodulators
IBDInflammatory bowel disease
IL-1Interleukin 1
IL-6Interleukin 6
IL-10Interleukin 10
IL-17Interleukin 17
iNOSInducible nitric oxide synthase
JAKJanus kinase
LPOLipid peroxidation
MDAMalondialdehyde
MPOMyeloperoxidase
MUFAsMonounsaturated fatty acids
NF-κBNuclear factor kappa B
OSCsOrganosulfur-containing compounds
OS Oxidative stress
PUFAsPolyunsaturated fatty acids
ROSReactive oxygen species
SACS-allyl-L-cysteine
SODSuperoxide dismutase
TLRsToll-like receptors
TNF-αTumor necrosis factor-alpha
UCUlcerative colitis

References

  1. Dakhli, N.; Rtibi, K.; Arrari, F.; Ayari, A.; Sebai, H. Prophylactic coloprotective effect of Urtica dioica leaves against dextran sulfate sodium (DSS)-induced ulcerative colitis in rats. Medicina 2023, 59, 1990. [Google Scholar] [CrossRef]
  2. Li, S.; Zhuge, A.; Chen, H.; Han, S.; Shen, J.; Wang, K.; Xia, J.; Xia, H.; Jiang, S.; Wu, Y.; et al. Sedanolide alleviates DSS-induced colitis by modulating the intestinal FXR-SMPD3 pathway in mice. J. Adv. Res. 2025, 69, 413–426. [Google Scholar] [CrossRef]
  3. Feng, J.; Liu, Y.; Zhang, C.; Ji, M.; Li, C. Phytic acid regulates proliferation of colorectal cancer cells by downregulating NF-kB and β-catenin signalling. Eur. J. Inflamm. 2023, 21, 1–14. [Google Scholar] [CrossRef]
  4. Alfwuaires, M.A.; Algefare, A.I.; Afkar, E.; Salam, S.A.; El-Moaty, H.I.A.; Badr, G.M. Immunomodulatory assessment of Portulaca oleracea L. extract in a mouse model of colitis. Biomed. Pharmacother. 2021, 143, 112148. [Google Scholar] [CrossRef] [PubMed]
  5. El Mahdy, R.N.; Nader, M.A.; Helal, M.G.; Abu-Risha, S.E.; Abdelmageed, M.E. Eicosapentaenoic acid mitigates ulcerative colitis-induced by acetic acid through modulation of NF-κB and TGF-β/EGFR signaling pathways. Life Sci. 2023, 327, 121820. [Google Scholar] [CrossRef]
  6. Ghasemi-Dehnoo, M.; Lorigooini, Z.; Amini-Khoei, H.; Sabzevary-Ghahfarokhi, M.; Rafieian-Kopaei, M. Quinic acid ameliorates ulcerative colitis in rats, through the inhibition of two TLR4-NF-κB and NF-κB-INOS-NO signaling pathways. Immun. Inflamm. Dis. 2023, 11, e926. [Google Scholar] [CrossRef]
  7. Rehman, I.U.; Saleem, M.; Raza, S.A.; Bashir, S.; Muhammad, T.; Asghar, S.; Qamar, M.U.; Shah, T.A.; Bin Jardan, Y.A.; Mekonnen, A.B.; et al. Anti-ulcerative colitis effects of chemically characterized extracts from Calliandra haematocephala in acetic acid-induced ulcerative colitis. Front. Chem. 2024, 12, 1291230. [Google Scholar] [CrossRef]
  8. Long, D.; Mao, C.; Xu, Y.; Zhu, Y. The emerging role of neutrophil extracellular traps in ulcerative colitis. Front. Immunol. 2024, 15, 1425251. [Google Scholar] [CrossRef] [PubMed]
  9. Chen, Z.; Yi, L.; Pan, Y.; Long, X.; Mu, J.; Yi, R.; Zhao, X. Lactobacillus fermentum ZS40 ameliorates inflammation in mice with ulcerative colitis induced by dextran sulfate sodium. Front. Pharmacol. 2021, 12, 700217. [Google Scholar] [CrossRef]
  10. Zhang, C.; Zhang, J.; Zhang, Y.; Song, Z.; Bian, J.; Yi, H.; Ma, Z. Identifying neutrophil-associated subtypes in ulcerative colitis and confirming neutrophils promote colitis-associated colorectal cancer. Front. Immunol. 2023, 14, 1095098. [Google Scholar] [CrossRef]
  11. Sun, W.; Hang, L.; Chen, L.; Qu, Y.; Zhang, Y.; Miao, Z.; Wang, X. Fraxin ameliorates ulcerative colitis by modulating oxidative stress, inflammation and TLR4/NF-κB and MAPK signaling pathways. Altern. Ther. Health Med. 2024, 30, 117–125. [Google Scholar] [PubMed]
  12. Ozbeyli, D.; Sen, A.; Yenal, N.O.; Yavuz, A.N.H.; Turet, D.M.; Yuksel, M.; Senkardes, I.; Aykac, A. Thymus longicaulis subsp. chaubardii ethanolic extract ameliorates acetic acid–induced rat ulcerative colitis model by inhibiting oxidative sress, inflammation, and apoptosis. J. Food Biochem. 2025, 2025, 6245901. [Google Scholar] [CrossRef]
  13. Chen, B.; Dong, X.; Zhang, J.L.; Sun, X.; Zhou, L.; Zhao, K.; Deng, H.; Sun, Z. Natural compounds target programmed cell death (PCD) signaling mechanism to treat ulcerative colitis: A review. Front. Pharmacol. 2024, 15, 1333657. [Google Scholar] [CrossRef]
  14. Albalawi, G.A.; Albalawi, M.Z.; Alsubaie, K.T.; Albalawi, A.Z.; Elewa, M.A.F.; Hashem, K.S.; Al-Gayyar, M.M.H. Curative effects of crocin in ulcerative colitis via modulating apoptosis and inflammation. Int. Immunopharmacol. 2023, 118, 110138. [Google Scholar] [CrossRef]
  15. Sebastian, S.A.; Kaiwan, O.; Co, E.L.; Mehendale, M.; Mohan, B.P. Current pharmacologic options and emerging therapeutic approaches for the management of ulcerative colitis: A narrative review. Spartan Med. Res. J. 2024, 9, 123397. [Google Scholar] [CrossRef] [PubMed]
  16. AlAmeel, T.; AlMutairdi, A.; Al-Bawardy, B. Emerging therapies for ulcerative colitis: Updates from recent clinical trials. Clin. Exp. Gastroenterol. 2023, 16, 147–167. [Google Scholar] [CrossRef]
  17. Chen, Y.; Wang, P.; Zhang, Y.; Du, X.Y.; Zhang, Y.J. Comparison of effects of aminosalicylic acid, glucocorticoids and immunosuppressive agents on the expression of multidrug-resistant genes in ulcerative colitis. Sci. Rep. 2022, 12, 20656. [Google Scholar] [CrossRef] [PubMed]
  18. Badr, G.; Elsawy, H.; Amalki, M.A.; Alfwuaires, M.; El-Gerbed, M.S.; Abdel-Moneim, A.M. Protective effects of myristicin against ulcerative colitis induced by acetic acid in male mice. Food Agric. Immunol. 2020, 31, 435–446. [Google Scholar] [CrossRef]
  19. Serrano, J.C.E.; Castro-Boque, E.; Garcia-Carrasco, A.; Moran-Valero, M.I.; Gonzalez-Hedstrom, D.; Bermudez-Lopez, M.; Valdivielso, J.M.; Espinel, A.E.; Portero-Otin, M. Antihypertensive effects of an optimized aged garlic extract in subjects with grade I hypertension and antihypertensive drug therapy: A randomized, triple-blind controlled trial. Nutrients 2023, 15, 3691. [Google Scholar] [CrossRef]
  20. Farhat, Z.; Scheving, T.; Aga, D.S.; Hershberger, P.A.; Freudenheim, J.L.; Hageman Blair, R.; Mammen, M.J.; Mu, L. Antioxidant and antiproliferative activities of several garlic forms. Nutrients 2023, 15, 4099. [Google Scholar] [CrossRef]
  21. Pandey, P.; Khan, F.; Alshammari, N.; Saeed, A.; Aqil, F.; Saeed, M. Updates on the anticancer potential of garlic organosulfur compounds and their nanoformulations: Plant therapeutics in cancer management. Front. Pharmacol. 2023, 14, 1154034. [Google Scholar] [CrossRef] [PubMed]
  22. Furdak, P.; Bartosz, G.; Sadowska-Bartosz, I. Which constituents determine the antioxidant activity and cytotoxicity of garlic? Role of organosulfur compounds and phenolics. Int. J. Mol. Sci. 2024, 25, 8391. [Google Scholar] [CrossRef]
  23. Dong, Y.; Zhang, J.; Xie, A.; Yue, X.; Li, M.; Zhou, Q. Garlic peel extract as an antioxidant inhibits triple-negative breast tumor growth and angiogenesis by inhibiting cyclooxygenase-2 expression. Food Sci. Nutr. 2024, 12, 6886–6895. [Google Scholar] [CrossRef] [PubMed]
  24. Kotenkova, E.A.; Kupaeva, N.V. Comparative antioxidant study of onion and garlic waste and bulbs. IOP Conf. Ser. Earth Environ. Sci. 2019, 333, 012031. [Google Scholar] [CrossRef]
  25. Carreon-Delgado, D.F.; Hernandez-Montesinos, I.Y.; Rivera-Hernandez, K.N.; Del Sugeyrol Villa-Ramirez, M.; Ochoa-Velasco, C.E.; Ramirez-Lopez, C. Evaluation of pretreatments and extraction conditions on the antifungal and antioxidant effects of garlic (Allium sativum) peel extracts. Plants 2023, 12, 217. [Google Scholar] [CrossRef]
  26. Linoj, J.; Ramadoss, R.; Selvam, S.P.; Sundar, S.; Ramani, P.; Shanmugam, R.K. Anti inflammatory activity of aqueous extract of garlic peel. J. Popul. Ther. Clin. Pharmacol. 2023, 30, 454–458. [Google Scholar]
  27. Hernandez-Montesinos, I.Y.; Carreon-Delgado, D.F.; Lazo-Zamalloa, O.; Tapia-Lopez, L.; Rosas-Morales, M.; Ochoa-Velasco, C.E.; Hernandez-Carranza, P.; Cruz-Narvaez, Y.; Ramirez-Lopez, C. Exploring agro-industrial by-products: Phenolic content, antioxidant capacity, and phytochemical profiling via FI-ESI-FTICR-MS untargeted analysis. Antioxidants 2024, 13, 925. [Google Scholar] [CrossRef]
  28. Ichikawa, M.; Ryu, K.; Yoshida, J.; Ide, N.; Kodera, Y.; Sasaoka, T.; Rosen, R.T. Identification of six phenylpropanoids from garlic skin as major antioxidants. J. Agric. Food Chem. 2003, 51, 7313–7317. [Google Scholar] [CrossRef] [PubMed]
  29. dos Santos, P.C.M.; da Silva, L.M.R.; Magalhaes, F.E.A.; Cunha, F.E.T.; Ferreira, M.J.G.; de Figueiredo, E.A.T. Garlic (Allium sativum L.) peel extracts: From industrial by-product to food additive. Appl. Food Res. 2022, 2, 100186. [Google Scholar] [CrossRef]
  30. Swirkosz, G.; Szczygiel, A.; Logon, K.; Wrzesniewska, M.; Gomulka, K. The role of the microbiome in the pathogenesis and treatment of ulcerative colitis-A literature review. Biomedicines 2023, 11, 3144. [Google Scholar] [CrossRef]
  31. Gyriki, D.; Nikolaidis, C.; Stavropoulou, E.; Bezirtzoglou, I.; Tsigalou, C.; Vradelis, S.; Bezirtzoglou, E. Exploring the gut microbiome’s role in inflammatory bowel disease: Insights and interventions. J. Pers. Med. 2024, 14, 507. [Google Scholar] [CrossRef] [PubMed]
  32. Zhang, Y.; Wang, L.; Peng, L. The role of intestinal fungi in the pathogenesis and treatment of ulcerative colitis. Microorganisms 2025, 13, 794. [Google Scholar] [CrossRef] [PubMed]
  33. Azmat, F.; Imran, A.; Islam, F.; Afzaal, M.; Zahoor, T.; Akram, R.; Aggarwale, S.; Rehman, M.; Naaz, S.; Ashraf, S.; et al. Valorization of the phytochemical profile, nutritional composition, and therapeutic potentials of garlic peel: A concurrent review. Int. J. Food Prop. 2023, 26, 2642–2655. [Google Scholar] [CrossRef]
  34. Mounithaa, N.; Selvam, S.P.; Shanmugam, R.K.; Ramadoss, R.; Sundar, S.; Ramani, P. Preparation of garlic peel extract and its free radical scavenging activity. J. Popul. Ther. Clin. Pharmacol. 2023, 30, 405–410. [Google Scholar]
  35. Wu, C.Y.; Couto, E.S.A.; Citadin, C.T.; Clemons, G.A.; Acosta, C.H.; Knox, B.A.; Grames, M.S.; Rodgers, K.M.; Lee, R.H.; Lin, H.W. Palmitic acid methyl ester inhibits cardiac arrest-induced neuroinflammation and mitochondrial dysfunction. Prostaglandins Leukot. Essent. Fat. Acids 2021, 165, 102227. [Google Scholar] [CrossRef]
  36. Salinas-Nolasco, C.; Perez-Hernandez, E.; Garza, S.; Park, H.G.; Brenna, J.T.; Castaneda-Hernandez, G.; Reyes-Lopez, C.A.S.; Perez-Hernandez, N.; Chavez-Pina, A.E. Antioxidative action of alpha-linolenic acid during its gastroprotective effect in an indomethacin-induced gastric injury model. Prev. Nutr. Food Sci. 2025, 30, 132–140. [Google Scholar] [CrossRef]
  37. Anand, R.; Kaithwas, G. Anti-inflammatory potential of alpha-linolenic acid mediated through selective COX inhibition: Computational and experimental data. Inflammation 2014, 37, 1297–1306. [Google Scholar] [CrossRef]
  38. Shoge, M.; Amusan, T. Phytochemical, antidiarrhoeal activity, isolation and characterisation of 11-Octadecenoic acid, methyl ester isolated from the seeds of Acacia nilotica Linn. J. Biotechnol. Immunol. 2020, 2, 1–12. [Google Scholar]
  39. Youssef, A.M.M.; Maaty, D.A.M.; Al-Saraireh, Y.M. Phytochemical analysis and profiling of antioxidants and anticancer compounds from Tephrosia purpurea (L.) subsp. apollinea family Fabaceae. Molecules 2023, 28, 3939. [Google Scholar]
  40. Ibrahim, O.H.M.; Al-Qurashi, A.D.; Asiry, K.A.; Mousa, M.A.A.; Alhakamy, N.A.; Abo-Elyousr, K.A.M. Investigation of potential in vitro anticancer and antimicrobial activities of Balanites aegyptiaca (L.) delile fruit extract and its phytochemical components. Plants 2022, 11, 2621. [Google Scholar] [CrossRef]
  41. Pinto, M.E.A.; Araujo, S.G.; Morais, M.I.; Sa, N.P.; Lima, C.M.; Rosa, C.A.; Siqueira, E.P.; Johann, S.; Lima, L. Antifungal and antioxidant activity of fatty acid methyl esters from vegetable oils. An. Acad. Bras. Ciên. 2017, 89, 1671–1681. [Google Scholar] [CrossRef]
  42. Al-Ani, W.M.; Almahdawy, S.S. GC/MS analysis of the peels of Annona muricata L. Res. J. Pharm. Technol. 2022, 15, 3137–3140. [Google Scholar]
  43. Efiong, E.E.; Nwaedozie, J.M.; Ubhenin, A.E.; Kwon-Ndung, E.H. GC-MS characterization of unsweetened and sweetened “Kunun-zaki” beverage and pulp. Food Sci. J. 2020, 19, 25–39. [Google Scholar] [CrossRef]
  44. Ezeduru, V.; Shao, A.R.Q.; Venegas, F.A.; McKay, G.; Rich, J.; Nguyen, D.; Thibodeaux, C.J. Defining the functional properties of cyclopropane fatty acid synthase from Pseudomonas aeruginosa PAO1. J. Biol. Chem. 2024, 300, 107618. [Google Scholar] [CrossRef] [PubMed]
  45. Fafal, T.; Tüzün, B.S.; Kıvçak, B. Fatty acid compositions and antioxidant activities of Ranunculus isthmicus subsp. tenuifolius and Ranunculus rumelicus. Int. J. Nat. Sci. 2022, 6, 151–159. [Google Scholar]
  46. Matsunaga, H.; Hokari, R.; Kurihara, C.; Okada, Y.; Takebayashi, K.; Okudaira, K.; Watanabe, C.; Komoto, S.; Nakamura, M.; Tsuzuki, Y.; et al. Omega-3 polyunsaturated fatty acids ameliorate the severity of ileitis in the senescence accelerated mice (SAM)P1/Yit mice model. Clin. Exp. Immunol. 2009, 158, 325–333. [Google Scholar] [CrossRef] [PubMed]
  47. Heshmati, J.; Morvaridzadeh, M.; Maroufizadeh, S.; Akbari, A.; Yavari, M.; Amirinejad, A.; Maleki-Hajiagha, A.; Sepidarkish, M. Omega-3 fatty acids supplementation and oxidative stress parameters: A systematic review and meta-analysis of clinical trials. Pharmacol. Res. 2019, 149, 104462. [Google Scholar] [CrossRef]
  48. Banaszak, M.; Dobrzynska, M.; Kawka, A.; Gorna, I.; Wozniak, D.; Przyslawski, J.; Drzymala-Czyz, S. Role of Omega-3 fatty acids eicosapentaenoic (EPA) and docosahexaenoic (DHA) as modulatory and anti-inflammatory agents in noncommunicable diet-related diseases—Reports from the last 10 years. Clin. Nutr. ESPEN 2024, 63, 240–258. [Google Scholar] [CrossRef]
  49. Ariturk, L.A.; Cilingir, S.; Kolgazi, M.; Elmas, M.; Arbak, S.; Yapislar, H. Docosahexaenoic acid (DHA) alleviates inflammation and damage induced by experimental colitis. Eur. J. Nutr. 2024, 63, 2801–2813. [Google Scholar] [CrossRef]
  50. Kusumah, D.; Wakui, M.; Murakami, M.; Xie, X.; Yukihito, K.; Maeda, I. Linoleic acid, alpha-linolenic acid, and monolinolenins as antibacterial substances in the heat-processed soybean fermented with Rhizopus oligosporus. Biosci. Biotechnol. Biochem. 2020, 84, 1285–1290. [Google Scholar] [CrossRef]
  51. Parang, K.; Knaus, E.E.; Wiebe, L.I.; Sardari, S.; Daneshtalab, M.; Csizmadia, F. Synthesis and antifungal activities of myristic acid analogs. Arch. Pharm. 1996, 329, 475–482. [Google Scholar] [CrossRef]
  52. Sasatsuki, H.; Nakazaki, A.; Uchida, K.; Shibata, T. Quantitative analysis of oxidized vitamin B1 metabolites generated by hypochlorous acid. Free Radic. Biol. Med. 2020, 152, 197–206. [Google Scholar] [CrossRef] [PubMed]
  53. Bose, R.; Kumar, M.S.; Manivel, A.; Mohan, S.C. Chemical constituents of Sauropus androgynus and evaluation of its antioxidant activity. Res. J. Phytochem. 2018, 12, 7–13. [Google Scholar]
  54. Andriolo, I.R.L.; Venzon, L.; da Silva, L.M. Perspectives about ascorbic acid to treat inflammatory bowel diseases. Drug Res. 2024, 74, 149–155. [Google Scholar] [CrossRef]
  55. Liu, K.Y.; Nakatsu, C.H.; Jones-Hall, Y.; Kozik, A.; Jiang, Q. Vitamin E alpha- and gamma-tocopherol mitigate colitis, protect intestinal barrier function and modulate the gut microbiota in mice. Free Radic. Biol. Med. 2021, 163, 180–189. [Google Scholar] [CrossRef] [PubMed]
  56. Zhang, Y.; Jin, X.; Xia, H.; Wu, X.; Chen, W.; Zhuang, M.; Tang, S. β-sitosterol ameliorates ulcerative colitis through modulation of the AMPK/MLCK anti-inflammatory pathway. J. Biochem. Mol. Toxicol. 2025, 39, e70287. [Google Scholar] [CrossRef]
  57. Thakur, R.S.; Ahirwar, B. A steroidal derivative from Trigonella foenum graecum L. that induces apoptosis In Vitro and In Vivo. J. Food Drug Anal. 2019, 27, 231–239. [Google Scholar] [CrossRef]
  58. Elezabeth, V.D.; Arumugam, S. MS analysis of ethanol extract of Cyperus rotundus leaves. Int. J. Curr. Biotechnol. 2014, 2, 19–23. [Google Scholar]
  59. Sun, J.; Xue, P.; Liu, J.; Huang, L.; Lin, G.; Ran, K.; Yang, J.; Lu, C.; Zhao, Y.Z.; Xu, H.L. Self-cross-linked hydrogel of cysteamine-grafted γ-polyglutamic acid stabilized tripeptide KPV for alleviating TNBS-induced ulcerative colitis in rats. ACS Biomater. Sci. Eng. 2021, 7, 4859–4869. [Google Scholar] [CrossRef]
  60. Msegued Ayam, I.; Zinedine, A.; Lyagoubi, A.; Rocha, J.M.; Raoui, S.M.; Errachidi, F. Chemical composition, antioxidant, and antifungal activity of essential oil extracted from cumin seeds (Cuminum cyminum L.) planted in Morocco. Chem. Biodivers. 2025, e02687. [Google Scholar] [CrossRef]
  61. van Breemen, R.B.; Pajkovic, N. Multitargeted therapy of cancer by lycopene. Cancer Lett. 2008, 269, 339–351. [Google Scholar] [CrossRef] [PubMed]
  62. Costa-Perez, A.; Sanchez-Bravo, P.; Medina, S.; Dominguez-Perles, R.; Garcia-Viguera, C. Bioaccessible organosulfur compounds in broccoli stalks modulate the inflammatory mediators involved in inflammatory bowel disease. Int. J. Mol. Sci. 2024, 25, 800. [Google Scholar] [CrossRef]
  63. Fudman, D.I.; McConnell, R.A.; Ha, C.; Singh, S. Modern advanced therapies for inflammatory bowel diseases: Practical considerations and positioning. Clin. Gastroenterol. Hepatol. 2025, 23, 454–468. [Google Scholar] [CrossRef]
  64. Fansiwala, K.; Sauk, J.S. Small molecules, big results: How JAK inhibitors have transformed the treatment of patients with IBD. Dig. Dis. Sci. 2025, 70, 469–477. [Google Scholar] [CrossRef] [PubMed]
  65. Nagaraj, T.; Shinn, J.; De Felice, K. A practical guide to selecting and using new ulcerative colitis therapies. Curr. Opin. Gastroenterol. 2024, 40, 235–242. [Google Scholar] [CrossRef] [PubMed]
  66. Rahman, S.; Patel, R.K.; Boden, E.; Tsikitis, V.L. Medical management of inflammatory bowel disease. Surg. Clin. N. Am. 2024, 104, 657–671. [Google Scholar] [CrossRef]
  67. Mosaad, Y.O.; Ateyya, H.; Hussein, M.A.; Moro, A.M.; Abdel-Wahab, E.A.; El-Ella, A.A.; Nassar, Z.N. BAO-Ag-NPs as promising suppressor of ET-1/ICAM-1/VCAM-1 signaling pathway in ISO-induced AMI in rats. Curr. Pharm. Biotechnol. 2024, 25, 772–786. [Google Scholar] [CrossRef]
  68. Elwekeel, A.; Hassan, M.H.; Almutairi, E.; AlHammad, M.; Alwhbi, F.; Abdel-Bakky, M.S.; Amin, E.; Mohamed, E.I. Anti-inflammatory, anti-oxidant, GC-MS profiling and molecular docking analyses of non-polar extracts from five salsola species. Separations 2023, 10, 72. [Google Scholar] [CrossRef]
  69. Farooqi, S.S.; Naveed, S.; Qamar, F.; Sana, A.; Farooqi, S.H.; Sabir, N.; Mansoor, A.; Sadia, H. Phytochemical analysis, GC-MS characterization and antioxidant activity of Hordeum vulgare seed extracts. Heliyon 2024, 10, e27297. [Google Scholar] [CrossRef]
  70. Mazumder, K.; Nabila, A.; Aktar, A.; Farahnaky, A. Bioactive variability and in vitro and in vivo antioxidant activity of unprocessed and processed flour of nine cultivars of Australian lupin species: A comprehensive substantiation. Antioxidants 2020, 9, 282. [Google Scholar] [CrossRef]
  71. Smyth, M.; Lunken, G.; Jacobson, K. Insights into inflammatory bowel disease and effects of dietary fatty acid intake with a focus on polyunsaturated fatty acids using preclinical models. J. Can. Assoc. Gastroenterol. 2024, 7, 104–114. [Google Scholar] [CrossRef] [PubMed]
  72. Magisetty, J.; Gadiraju, B.; Kondreddy, V. Genomic analysis in the colon tissues of omega-3 fatty acid-treated rats identifies novel gene signatures implicated in ulcerative colitis. Int. J. Biol. Macromol. 2024, 258, 128867. [Google Scholar] [CrossRef] [PubMed]
  73. Calder, P.C. n-3 PUFA and inflammation: From membrane to nucleus and from bench to bedside. Proc. Nutr. Soc. 2020, 79, 404–416. [Google Scholar] [CrossRef]
  74. Zhao, H.; Li, T.; Li, C.; Xiong, Z.; Rong, W.; Cao, L.; Chen, G.; Liu, Q.; Liu, Y.; Wang, X.; et al. Vitamin C alleviates intestinal damage induced by 17α-methyltestosterone in Carassius auratus. Aquat. Toxicol. 2025, 280, 107266. [Google Scholar] [CrossRef]
  75. Zhu, R.; Liu, Z.; Lu, M.; Wu, X.; Zhao, X.; Wang, H.H.; Quan, Y.N.; Wu, L.F. The protective role of vitamin C on intestinal damage induced by high-dose glycinin in juvenile Rhynchocypris lagowskii Dybowski. Fish Shellfish Immunol. 2023, 134, 108589. [Google Scholar] [CrossRef]
  76. Ekeanyanwu, C.R.; Uzoma, T.C. Chemical characterization and anti-inflammatory activity of flavonoid fraction of garlic skin extract: A potential source of natural anti-inflammatory agents. J. Res. Complement. Med. 2025, 2, 43–58. [Google Scholar] [CrossRef]
  77. Kurumi, H.; Yokoyama, Y.; Hirano, T.; Akita, K.; Hayashi, Y.; Kazama, T.; Isomoto, H.; Nakase, H. Cytokine profile in predicting the effectiveness of advanced therapy for ulcerative colitis: A narrative review. Biomedicines 2024, 12, 952. [Google Scholar] [CrossRef]
  78. Vebr, M.; Pomahacova, R.; Sykora, J.; Schwarz, J. A narrative review of cytokine networks: Pathophysiological and therapeutic implications for inflammatory bowel disease pathogenesis. Biomedicines 2023, 11, 3229. [Google Scholar] [CrossRef] [PubMed]
  79. Lu, P.D.; Zhao, Y.H. Targeting NF-κB pathway for treating ulcerative colitis: Comprehensive regulatory characteristics of Chinese medicines. Chin. Med. 2020, 15, 15. [Google Scholar] [CrossRef]
  80. Yu, X.; Li, C.; Tao, Y.; Xia, T.; Jia, Z. Lacidipine inhibits NF-κB and Notch pathways and mitigates DSS-induced colitis. Dig. Dis. Sci. 2024, 69, 3753–3759. [Google Scholar] [CrossRef]
  81. Laurindo, L.F.; Santos, A.; Carvalho, A.C.A.; Bechara, M.D.; Guiguer, E.L.; Goulart, R.A.; Vargas Sinatora, R.; Araujo, A.C.; Barbalho, S.M. Phytochemicals and regulation of NF-kB in inflammatory bowel diseases: An overview of In Vitro and In Vivo effects. Metabolites 2023, 13, 96. [Google Scholar] [CrossRef] [PubMed]
  82. Mostafa, R.E.; Abdel-Rahman, R.F. Ezetimibe alleviates acetic acid-induced ulcerative colitis in rats: Targeting the Akt/NF-κB/STAT3/CXCL10 signaling axis. J. Pharm. Pharmacol. 2023, 75, 533–543. [Google Scholar] [CrossRef]
  83. Lokman, M.S.; Kassab, R.B.; Salem, F.A.M.; Elshopakey, G.E.; Hussein, A.; Aldarmahi, A.A.; Theyab, A.; Alzahrani, K.J.; Hassan, K.E.; Alsharif, K.F.; et al. Asiatic acid rescues intestinal tissue by suppressing molecular, biochemical, and histopathological changes associated with the development of ulcerative colitis. Biosci. Rep. 2024, 44, BSR20232004. [Google Scholar] [CrossRef] [PubMed]
  84. Branchett, W.J.; Saraiva, M.; O’Garra, A. Regulation of inflammation by Interleukin-10 in the intestinal and respiratory mucosa. Curr. Opin. Immunol. 2024, 91, 102495. [Google Scholar] [CrossRef]
  85. Nguyen, H.D.; Aljamaei, H.M.; Stadnyk, A.W. The production and function of endogenous interleukin-10 in intestinal epithelial cells and gut homeostasis. Cell Mol. Gastroenterol. Hepatol. 2021, 12, 1343–1352. [Google Scholar] [CrossRef]
  86. Wei, H.X.; Wang, B.; Li, B. IL-10 and IL-22 in mucosal immunity: Driving protection and pathology. Front. Immunol. 2020, 11, 1315. [Google Scholar] [CrossRef]
  87. Muro, P.; Zhang, L.; Li, S.; Zhao, Z.; Jin, T.; Mao, F.; Mao, Z. The emerging role of oxidative stress in inflammatory bowel disease. Front. Endocrinol. 2024, 15, 1390351. [Google Scholar] [CrossRef] [PubMed]
  88. Jawhara, S. How do polyphenol-rich foods prevent oxidative stress and maintain gut health? Microorganisms 2024, 12, 1570. [Google Scholar] [CrossRef]
  89. Wu, J.S.; Liu, H.J.; Han, S.J.; Mao, N.F.; Liu, X.F. Chelerythrine ameliorates acetic acid-induced ulcerative colitis via suppression of inflammation and oxidation. Nat. Prod. Commun. 2022, 17, 1–8. [Google Scholar] [CrossRef]
  90. Zheng, X.Y.; Lv, Y.F.; Li, S.; Li, Q.; Zhang, Q.N.; Zhang, X.T.; Hao, Z.M. Recombinant adeno-associated virus carrying thymosin β4 suppresses experimental colitis in mice. World J. Gastroenterol. 2017, 23, 242–255. [Google Scholar] [CrossRef]
  91. Subramanian, S.; Geng, H.; Tan, X.D. Cell death of intestinal epithelial cells in intestinal diseases. Sheng Li Xue Bao 2020, 72, 308–324. [Google Scholar] [PubMed]
  92. Wang, M.; Wang, Z.; Li, Z.; Qu, Y.; Zhao, J.; Wang, L.; Zhou, X.; Xu, Z.; Zhang, D.; Jiang, P.; et al. Targeting programmed cell death in inflammatory bowel disease through natural products: New insights from molecular mechanisms to targeted therapies. Phytother. Res. 2025, 39, 1776–1807. [Google Scholar] [CrossRef] [PubMed]
  93. Ghazavi, F.; Huysentruyt, J.; De Coninck, J.; Kourula, S.; Martens, S.; Hassannia, B.; Wartewig, T.; Divert, T.; Roelandt, R.; Popper, B.; et al. Executioner caspases 3 and 7 are dispensable for intestinal epithelium turnover and homeostasis at steady state. Proc. Natl. Acad. Sci. USA 2022, 119, e2024508119. [Google Scholar] [CrossRef] [PubMed]
  94. Tripathi, K.; Dong, J.; Mishkin, B.F.; Feuerstein, J.D. Patient preference and adherence to aminosalicylates for the treatment of ulcerative colitis. Clin. Exp. Gastroenterol. 2021, 14, 343–351. [Google Scholar] [CrossRef]
  95. Khan, H.M.; Mehmood, F.; Khan, N. Optimal management of steroid-dependent ulcerative colitis. Clin. Exp. Gastroenterol. 2015, 8, 293–302. [Google Scholar] [CrossRef]
  96. Zhdanava, M.; Zhao, R.; Manceur, A.M.; Ding, Z.; Boudreau, J.; Kachroo, S.; Kerner, C.; Izanec, J.; Pilon, D. Burden of chronic corticosteroid use among patients with ulcerative colitis initiated on targeted treatment or conventional therapy in the United States. J. Manag. Care Spec. Pharm. 2024, 30, 141–152. [Google Scholar] [CrossRef]
  97. Nguyen, G.C.; Harris, M.L.; Dassopoulos, T. Insights in immunomodulatory therapies for ulcerative colitis and Crohn’s disease. Curr. Gastroenterol. Rep. 2006, 8, 499–505. [Google Scholar] [CrossRef]
  98. Chhibba, T.; Ma, C. Is there room for immunomodulators in ulcerative colitis? Expert. Opin. Biol. Ther. 2020, 20, 379–390. [Google Scholar] [CrossRef]
  99. Bhattacharya, A.; Osterman, M.T. Biologic therapy for ulcerative colitis. Gastroenterol. Clin. N. Am. 2020, 49, 717–729. [Google Scholar] [CrossRef]
  100. Dutt, K.; Vasudevan, A. Therapeutic drug monitoring for biologic and small-molecule therapies for inflammatory bowel disease. Medicina 2024, 60, 250. [Google Scholar] [CrossRef]
  101. Harindranath, S. Tofacitinib in ulcerative colitis—Second-line therapy, first-rate results. Dig. Dis. Sci. 2024, 69, 3116–3118. [Google Scholar] [CrossRef] [PubMed]
  102. Caballero-Mateos, A.M.; Canadas-de la Fuente, G.A. Game changer: How Janus kinase inhibitors are reshaping the landscape of ulcerative colitis management. World J. Gastroenterol. 2024, 30, 3942–3953. [Google Scholar] [CrossRef] [PubMed]
  103. Müller, J.; Heindl, A. Drying of medicinal plants. In Medicinal and Aromatic Plants: Agricultural, Commercial, Ecological, Legal, Pharmacological and Social Aspects; Bogers, R.J., Craker, L.E., Lange, D., Eds.; Springer: Wageningen, The Netherlands, 2006; pp. 237–252. [Google Scholar]
  104. Lala, P.K. Lab Manuals of Pharmacognosy, 5th ed.; CSI Publishers and Distributors: Calcutta, India, 1993. [Google Scholar]
  105. Ichihara, K.; Fukubayashi, Y. Preparation of fatty acid methyl esters for gas-liquid chromatography. J. Lipid Res. 2010, 51, 635–640. [Google Scholar] [CrossRef] [PubMed]
  106. Ghasemi-Pirbaluti, M.; Motaghi, E.; Najafi, A.; Hosseini, M.J. The effect of theophylline on acetic acid induced ulcerative colitis in rats. Biomed. Pharmacother. 2017, 90, 153–159. [Google Scholar] [CrossRef]
  107. Yagi, K. Simple assay for the level of total lipid peroxides in serum or plasma. Methods Mol. Biol. 1998, 108, 101–106. [Google Scholar]
Figure 1. GC-MS total ion chromatogram (TIC) of analyzed GPE.
Figure 1. GC-MS total ion chromatogram (TIC) of analyzed GPE.
Pharmaceuticals 18 00969 g001
Figure 2. Effect of GPE on final body weight (A) and colon length (B) in AA-induced UC rats. Data expressed as mean values ± SD (n = 6/group). ** = p < 0.001 and *** = p < 0.0001 vs. the AA group. Representative pictures of colon length (C) in all experimental groups.
Figure 2. Effect of GPE on final body weight (A) and colon length (B) in AA-induced UC rats. Data expressed as mean values ± SD (n = 6/group). ** = p < 0.001 and *** = p < 0.0001 vs. the AA group. Representative pictures of colon length (C) in all experimental groups.
Pharmaceuticals 18 00969 g002
Figure 3. Effect of GPE on pro- and anti-inflammatory mediators (A) IL-1, (B) IL-6, (C) IL-17, (D) TNF-α, (E) IL-10, and (F) the transcription factor NF-κB in AA-induced UC rats. Data expressed as mean values ± SD (n = 6/group). ** = p < 0.001 and *** = p < 0.0001 vs. the AA group.
Figure 3. Effect of GPE on pro- and anti-inflammatory mediators (A) IL-1, (B) IL-6, (C) IL-17, (D) TNF-α, (E) IL-10, and (F) the transcription factor NF-κB in AA-induced UC rats. Data expressed as mean values ± SD (n = 6/group). ** = p < 0.001 and *** = p < 0.0001 vs. the AA group.
Pharmaceuticals 18 00969 g003
Figure 4. Effect of GPE on oxidative stress biomarkers (A) CAT, (B) SOD, (C) MDA, and (D) MPO in AA-induced UC rats. Data expressed as mean values ± SD (n = 6/group). *** = p < 0.0001 vs. the AA group.
Figure 4. Effect of GPE on oxidative stress biomarkers (A) CAT, (B) SOD, (C) MDA, and (D) MPO in AA-induced UC rats. Data expressed as mean values ± SD (n = 6/group). *** = p < 0.0001 vs. the AA group.
Pharmaceuticals 18 00969 g004
Figure 5. Effect of GPE on levels of caspase-3 in AA-induced UC rats. Data expressed as mean values ± SD (n = 6/group). *** = p < 0.0001 vs. the AA group.
Figure 5. Effect of GPE on levels of caspase-3 in AA-induced UC rats. Data expressed as mean values ± SD (n = 6/group). *** = p < 0.0001 vs. the AA group.
Pharmaceuticals 18 00969 g005
Figure 6. Photomicrograph of histologically analyzed H&E-stained colon sections in all experimental groups: (A) control rats (Ctrl group) showing normal mucosal structure with intact epithelial surface (M), muscular layer (Mm), and submucosa (Sm); (B) rats with AA-induced colitis (AA group) showing histopathological alterations evidenced by severe mucosal damage, loss of goblet cells (arrowheads), and necrosis of mucosa (M) associated with marked infiltration of mononuclear cells (arrow); (C) GPE-treated rats (GPE group) showing almost nearly normal mucosal structure (M), muscular layer (Mm), and submucosa (Sm); (D) GPE-treated rats with colitis (GPE + AA group) showing improvement in histological structure with slight loss of some surface columnar epithelial cells (arrowhead) and minimal necrosis of mucosa (M). Original magnification: 100×. (E) Scoring of histology sections. Data expressed as mean ± SD (n = 6/group). *** = p < 0.0001 vs. AA group.
Figure 6. Photomicrograph of histologically analyzed H&E-stained colon sections in all experimental groups: (A) control rats (Ctrl group) showing normal mucosal structure with intact epithelial surface (M), muscular layer (Mm), and submucosa (Sm); (B) rats with AA-induced colitis (AA group) showing histopathological alterations evidenced by severe mucosal damage, loss of goblet cells (arrowheads), and necrosis of mucosa (M) associated with marked infiltration of mononuclear cells (arrow); (C) GPE-treated rats (GPE group) showing almost nearly normal mucosal structure (M), muscular layer (Mm), and submucosa (Sm); (D) GPE-treated rats with colitis (GPE + AA group) showing improvement in histological structure with slight loss of some surface columnar epithelial cells (arrowhead) and minimal necrosis of mucosa (M). Original magnification: 100×. (E) Scoring of histology sections. Data expressed as mean ± SD (n = 6/group). *** = p < 0.0001 vs. AA group.
Pharmaceuticals 18 00969 g006
Table 1. Chemical compounds identified in GPE by GC-MS analysis.
Table 1. Chemical compounds identified in GPE by GC-MS analysis.
Name of Detected CompoundRT (min)MFMWA
(%)
CAS #Biological Role(s)
Fatty acid esters
Heptanoic acid, 6-oxo-,methyl ester6.51C8H14O31581.912046-21-1Not reported
Palmitic acid, methyl ester25.60C17H34O22705.26112-39 -0Anti-inflammatory [35]
α-Linolenic acid, methyl ester, omega-3 28.66C19H32O22929.587361-80-0Antioxidant; anti-inflammatory [36,37]
11-Octadecenoic acid, methyl ester28.79C19H36O229615.8752380-33-3Antimicrobial; anti-inflammatory; antioxidant [38]
10-Octadecenoic acid, methyl ester28.91C19H36O22966.9713481-95-3Antimicrobial; antioxidant [39]
6,9-Octadecadiynoic acid, methyl ester 29.10C19H30O22901.0256847-03-1Anticancer; antimicrobial [40]
Stearic acid, methyl ester29.35C19H38O22987.55112-61-8Antioxidant; antifungal [41]
Nonanoic acid, methyl ester29.58C10H20O21726.181731-84-6Not reported
4,7-Octadecadiynoic acid, methyl ester30.15C19H30O22903.6918202-20-5Contribution to antioxidant and cytotoxic activities [42]
7,10-Octadecadienoic acid, methyl ester30.23C19H34O22943.6256554-24-6Might enhance immune function [43]
Stearic acid, 3-(octadecyloxy) propyl ester35.70C39H78O35940.4517367-40-7Not reported
Cyclopropanedodecanoic acid, 2-octyl-, methyl ester35.98C24H46O23660.3710152-65-5Antioxidant; anti-inflammatory; reduction in lipid peroxidation [44]
Fatty acids
Oleic acid (omega-9 fatty acid)4.04C18H34O22820.62112-80-1Antioxidant [45]
Eicosapentaenoic acid (fish omega-3)29.58C20H30O23026.1810417-94-4Anti-inflammatory; antioxidant [5,46,47,48]
Docosahexaenoic acid (omega 3)30.47C22H32O23283.56NAAnti-inflammatory; antioxidant [46,47,49]
Fatty acid derivatives
6-Dodecanone (derived from lauric acid)6.41C12H18O1783.23NANot reported
2-Monolinolenin (derived from linolenic acid)24.65C27H52O4Si24962.9155521-23-8Antibacterial; antifungal [50]
2-Bromotetradecanoic acid (derived from myristic acid)32.78C14H27BrO23060.6110520-81-7Antifungal [51]
Vitamins
Thiamine hydrochloride (vitamin B1)23.83C12H17ClN4OS3380.32NAAntioxidant [52]
L-Ascorbic acid 2,6-dihexadecanoate26.36C38H68O86524.1828474-90-0Antioxidant [53,54]
Tocopherol (Vitamin E)29.50C29H50O24310.36NAMitigation of colitis [55]
Steroids
β-sitosterol11.20C29H50O4153.18NAAmeliorates colitis [56]
Ethyl iso-allocholate31.78C26H44O54360.2147676-48-2Anti-apoptotic [57]
Natural alcohols
Hexahydrofarnesol (sesquiterpene alcohol)9.92C15H32O2280.336750-34-1Not reported
1-Heptatriacotanol (fatty alcohol)30.01C37H76O5362.39105794-58-9Antimicrobial [58]
Organosulfur compounds
Ethanimidothioic acid (derived from sulfur amino acid)4.64C7H13N3O3S2191.2123135-22-0Not reported
Cysteamine sulphonic acid 11.18C2H7NO3S21571.432937-53-3Supporting mucosal healing in colitis [59]
Fatty aldehydes
11-Octadecenal4.12C18H34O2660.2856554-95-1Not reported
Butanedial4.81C4H6O2860.18638-37 -9Not reported
3,5-Heptadienal, 2-ethylidene-6-methyl11.6C10H14O1502.0599172-18-6Antioxidant; antifungal [60]
Carotenoids
ψ,ψ-Carotene30.99C42H64O26002.4313833-01-7Antioxidant [61]
Sugars
Melezitose9.81C18H32O165040.65597-12-6Not reported
Arabinitol36.51C15H22O103620.3426674-23-7
Glucosinolates
Glucobrassicin20.38C16H20N2O9S24480.84356-52-9Modulation of inflammatory mediators in IBD [62]
RT = retention time; MF = molecular formula; MW = molecular weight; A = area; CAS # = chemical abstract service number.
Table 2. Comparative analysis of GPE and conventional UC therapies.
Table 2. Comparative analysis of GPE and conventional UC therapies.
Therapeutic AgentKey Mechanisms and BenefitsMain LimitationsComparative Advantages of GPEReference(s)
GPEMultimodal: antioxidant, anti-inflammatory (↓TNF-α), anti-apoptotic (↓caspase-3); localized deliveryRequires further clinical validationTargeted action; minimal systemic effects; suitable for chronic useOur study
Aminosalicylates (5-ASA)Topical anti-inflammatory (epithelial COX/LOX inhibition)No antioxidant/anti-apoptotic activityBroader cytoprotection beyond inflammation control[94]
Corticosteroids (CSs)Systemic immunosuppression (↓NF-κB/cytokines)Severe metabolic effects; contraindicated for maintenanceSafer long-term profile without steroidogenic effects[95,96]
Immunomodulators (IMMs)Cytotoxic lymphocyte suppressionInfection/malignancy risksNon-cytotoxic immune regulation via natural pathways[97]
Biologics (Anti-TNF)Neutralize TNF-α, effective for moderate-to-severe UCImmunogenicity; high costNatural TNF-α downregulation with lower immunogenicity[99,100]
JAK InhibitorsBlock intracellular cytokine signaling (JAK/STAT pathway)Thromboembolic risk; pan-cytokine inhibition Selective anti-inflammatory action without broad immunosuppression[101,102]
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

Althumairy, D.A.; Abu-Khudir, R.; Alandanoosi, A.I.; Badr, G.M. Garlic Peel-Derived Phytochemicals Using GC-MS: Antioxidant, Anti-Inflammatory, and Anti-Apoptotic Effects in Ulcerative Colitis Rat Model. Pharmaceuticals 2025, 18, 969. https://doi.org/10.3390/ph18070969

AMA Style

Althumairy DA, Abu-Khudir R, Alandanoosi AI, Badr GM. Garlic Peel-Derived Phytochemicals Using GC-MS: Antioxidant, Anti-Inflammatory, and Anti-Apoptotic Effects in Ulcerative Colitis Rat Model. Pharmaceuticals. 2025; 18(7):969. https://doi.org/10.3390/ph18070969

Chicago/Turabian Style

Althumairy, Duaa A., Rasha Abu-Khudir, Afnan I. Alandanoosi, and Gehan M. Badr. 2025. "Garlic Peel-Derived Phytochemicals Using GC-MS: Antioxidant, Anti-Inflammatory, and Anti-Apoptotic Effects in Ulcerative Colitis Rat Model" Pharmaceuticals 18, no. 7: 969. https://doi.org/10.3390/ph18070969

APA Style

Althumairy, D. A., Abu-Khudir, R., Alandanoosi, A. I., & Badr, G. M. (2025). Garlic Peel-Derived Phytochemicals Using GC-MS: Antioxidant, Anti-Inflammatory, and Anti-Apoptotic Effects in Ulcerative Colitis Rat Model. Pharmaceuticals, 18(7), 969. https://doi.org/10.3390/ph18070969

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