Next Article in Journal
Effects of Flavonoids and Phenols from Moringa oleifera Leaf Extracts on Biofilm Processes in Xanthomonas campestris pv. campestris
Previous Article in Journal
Fine Mapping of the Affecting Tillering and Plant Height Gene CHA-1 in Rice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Network Pharmacological Analysis and Experimental Validation of the Effect of Smilacis Glabrae Rhixoma on Gastrointestinal Motility Disorder

1
Division of Longevity and Biofunctional Medicine, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
2
Department of Clinical Korean Medicine, Graduate School of Kyung Hee University, Seoul 02447, Republic of Korea
3
Department of Gastroenterology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
4
College of Oriental Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Plants 2023, 12(7), 1509; https://doi.org/10.3390/plants12071509
Submission received: 3 March 2023 / Revised: 21 March 2023 / Accepted: 29 March 2023 / Published: 30 March 2023
(This article belongs to the Section Phytochemistry)

Abstract

:
Gastrointestinal motility disorder (GMD) is a disease that causes digestive problems due to inhibition of the movement of the gastrointestinal tract and is one of the diseases that reduce the quality of life of modern people. Smilacis Glabrae Rhixoma (SGR) is a traditional herbal medicine for many diseases and is sometimes prescribed to improve digestion. As a network pharmacological approach, we searched the TCMSP database for SGR, reviewed its constituents and target genes, and analyzed its relevance to gastrointestinal motility disorder. The effects of the SGR extract on the pacemaker activity in interstitial cells of Cajal (ICC) and gastric emptying were investigated. In addition, using the GMD mouse model through acetic acid (AA), we investigated the locomotor effect of SGR on the intestinal transit rate (ITR). As a result of network pharmacology analysis, 56 compounds out of 74 candidate compounds of SGR have targets, the number of targets is 390 targets, and there are 904 combinations. Seventeen compounds of SGR were related to GMD, and as a result of comparing the related genes with the GMD-related genes, 17 genes (active only) corresponded to both. When looking at the relationship network between GMD and SGR, it was confirmed that quercetin, resveratrol, SCN5A, TNF, and FOS were most closely related to GMD. In addition, the SGR extract regulated the pacemaker activity in ICC and recovered the delayed gastric emptying. As a result of feeding the SGR extract to AA-induced GMD mice, it was confirmed that the ITR decreased by AA was restored by the SGR extract. Through network pharmacology, it was confirmed that quercetin, resveratrol, SCN5A, TNF, and FOS were related to GMD in SGR, and these were closely related to intestinal motility. Based on these results, it is suggested that SGR in GMD restores digestion through the recovery of intestinal motility.

1. Introduction

Gastrointestinal motility disorder (GMD) is a disease in which digestive function is impaired due to impaired motility of the gastrointestinal tract and can occur throughout the intestine, including the esophagus and colon. These gastrointestinal motility disorders are accompanied by a variety of chronic symptoms, including vomiting and nausea [1]. Many people experience discomfort in their stomach, intestines, or bowel movements. These disorders degrade a person’s quality of life and result in significant increases in health care costs [2]. The gastrointestinal tract is an essential system for digesting food, absorbing nutrients, and excreting waste products. In this action, the movement of content in sequence is key, and this movement is supported by active and passive peristalsis (coordinated slow waves of muscle contraction and relaxation) [3,4].
Smilacis Glabrae Rhixoma (SGR) is an herbaceous vine widely distributed in Burma, India, Vietnam, Thailand, and China [5]. SGR is described in the Chinese Pharmacopoeia regarding the treatment of syphilis, furunculosis, dermatitis, eczema, cystitis, nephritis, and mercury poisoning [6]. Modern scientific research has demonstrated that SGR possesses several pharmacological and biological effects, including anti-inflammatory [7], antioxidant [8], antipsoriatic [9], anticancer [10,11], and antihyperuricemic [12] effects. It is known to the public that SGR is effective in helping digestion, but the specific mechanism is not yet known.
For the scientific analysis of SGR regarding gastrointestinal motility disorder, we took a network pharmacological approach integrating bioinformatics and systems biology. Traditional drugs such as SGR induce therapeutic effects based on the complexity of their ingredients and their effects on various biological systems, so the Western pharmacological approach of the existing ‘single target single drug’ is difficult to explain. However, network pharmacology is specialized in the analysis of ‘multi-target, multi-component’, suggesting a new paradigm for the scientific analysis of traditional drugs [13]. We intend to predict the components and mechanisms that SGR can treat for GMD through network pharmacological analysis of SGR, and to prove the therapeutic effect of SGR on GMD through mouse experiments. Network pharmacology analysis was conducted according to the scheme in Figure 1.

2. Results

2.1. Information of 390 Targets Derived through Correlation Investigation between Compounds and Targets

We identified 74 potential active compounds of SGR using the traditional Chinese medicine systems pharmacology database and analysis platform (TCMSP) database (Supplementary Materials Table S1). Among them, 56 compounds had target information (Supplementary Materials Table S2), and it was found that 56 compounds and 390 targets interact with each other by the combination of 904 compounds (Figure 2). As shown in Figure 2, quercetin was linked to the most targets (154 genes), followed by resveratrol (151 genes), succinic acid (60 genes), oleic acid (48 genes), beta-sitosterol (38 genes), naringenin (37 genes), (-)-epicatechin (32 genes), Stigmasterol (31 genes), (L)-alpha-Terpineol (22 genes), HMF (20 genes), and (R)-linalool (20 genes).

2.2. Seventeen Active Compounds Achieved the Criteria for Absorption, Distribution, Metabolism, and Excretion Parameters Applied

Seventeen compounds were included in the active compound screening criteria (Table 1), including (-)-taxifolin, (2R,3R)-2-(3,5-dihydroxyphenyl)-3,5,7-trihydroxychroman-4-one, 2H-3,9a-Methano-1-benzoxepin-9-methanol, octahydro-2,2,5a-trimethyl-, (3R-(3alpha,5aalpha,9alpha,9aalpha))-, 4,7-Dihydroxy-5-methoxyl-6-methyl-8-formyl-flavan, beta-sitosterol, cis-Dihydroquercetin, Dihydroresveratrol, diosgenin, EIC, Enhydrin, Methyllinolenate, naringenin, oleic acid, quercetin, sitosterol, Stigmasterol, and taxifolin.

2.3. Thirty-Nine Compounds Related to Gastrointestinal (GI) Disease Were Identified in SGR

We also investigated the compound–target–disease relationship using the TCMSP database. We noted that 39 compounds are associated with GI disease (Table 2). In particular, (-)-taxifolin, (2R,3R)-2-(3,5-dihydroxyphenyl)-3,5,7-trihydroxychroman-4-one, 4,7-Dihydroxy-5-methoxyl-6-methyl-8-formyl-flavan, beta-sitosterol, cis-Dihydroquercetin, Dihydroresveratrol, diosgenin, EIC, Methyllinolenate, naringenin, oleic acid, quercetin, Stigmasterol, and taxifolin were found to be active compounds associated with gastrointestinal disease, and other compounds, (-)-epicatechin, (2S,3R)-3,5,7-trihydroxy-2-(4-hydroxyphenyl)chroman-4-one, (L)-alpha-Terpineol, (R)-linalool, 3-O-caffeoylshikimic acid, Aromadedrin, astilbin, delta-amorphene, engeletin, FER, HMF, isoengelitin, Istidina, L-Bornyl acetate, myristic acid, n-butyl-β-D-fructopyronoside, n-butyl-β-D-fructoufranoside, NCA, nonane, palmitic acid, resveratrol, Sitogluside, SKM, stearic acid, and succinic acid, associated with gastrointestinal diseases, were classified as not active compounds (Figure 3).

2.4. Seventeen GI-Disease-Related Compounds in SGR Were Associated with GMD

To confirm the network relationship between SGR and GMD, genetic information related to GMD was checked using the cytoscape app. GMD-associated genes were found with a cutoff of 0.40 with a confidence (score) of 0.40 by limiting to a maximum of 100 proteins (Supplementary Materials Table S3). Based on the derived data, a network of GMD-related genes and SGR target genes was created (Figure 4). There are 19 genes in both gene sets, and among the GMD-related genes, the genes targeted by SGR are ACHE, CCK, CXCL8, DPP4, FOS, GCG, IL10, IL1B, IL6, INS, MPO, OPRM1, PYY, SCN5A, SLC6A4, TNF, NOS1, HTR3A and TRPV1.

2.5. The Network of GMD-Related Genes and Compounds for Identification of Interesting Molecules

Figure 5 shows a network of relationships between SGR compounds and GMD-related target genes. As a result of the network analysis, it was found that quercetin, resveratrol, and SCN5A were the most related to GMD. In short, (-)-epicatechin, (L)-alpha-Terpineol, (R)-linalool, 4,7-Dihydroxy-5-methoxyl-6-methyl-8-formyl-flavan, EIC, HMF, Sitogluside, Stigmasterol, astilbin, beta-sitosterol, n-butyl-β-D-fructopyronoside, nonane, oleic acid, palmitic acid, quercetin, resveratrol, and succinic acid are active compounds targeting GMD-related genes, which means these compounds can be potential medicinal candidates.

2.6. Effects of SGR Extract on Pacemaker Activity on Interstitial Cells of Cajal (ICC) in Murine Small Intestine

The SGR extract depolarized pacemaker potentials and decreased the frequency (Figure 6A–C). The degrees of depolarization were 6.2 ± 0.9 mV (p < 0.0001) at 1 mg/mL, 10.8 ± 1.3 mV (p < 0.0001) at 3 mg/mL, and 25.4 ± 1.7 mV (p < 0.0001) at 5 mg/mL (Figure 6D). The degrees of frequency were 10.9 ± 0.8 mV at 1 mg/mL, 7.1 ± 0.8 mV (p < 0.0001) at 3 mg/mL, and 1.3 ± 0.4 mV (p < 0.0001) at 5 mg/mL (Figure 6E). These results indicated that SGR regulates the pacemaker potential of ICC.

2.7. Effects of SGR Extract on Delayed Gastric Emptying

Macroscopic observations showed that loperamide injecting reduced the movement of foods in the stomach, while pretreatment with the SGR extract decreased these phenomena (Figure 7B). The gastric weight of the SGR-extract-treated group was reduced compared to that of the loperamide group (* p < 0.05, Figure 7C). In addition, the amount of phenol red retention in the SGR-extract-pretreated group was reduced compared to that of the loperamide group (* p < 0.05, ** p < 0.01, Figure 7D). The Mosapride-pretreated group had similar results to the SGR-extract-pretreated group.

2.8. Effects of SGR Extract on Intestinal Transit Rate (ITR) in Normal Mice

The ITR was 46.9 ± 1.5% in normal mice (Figure 8A). The ITR values of SGR mice were 47.9 ± 2.8% at 0.01 g/kg, 51.9 ± 2.6% (p < 0.05), and 60.8 ± 1.5% (p < 0.0001) (Figure 8A).

2.9. Effects of SGR Extract on ITR in Mice Regarding Gastrointestinal Motility Dysfunction

We used GMD models (acetic acid (AA) models). The AA model decreased ITR (34.9 ± 3.2% vs. 47.9 ± 1.9% in normal Controls; p < 0.0001) (Figure 8B). However, the SGR extract at 0.01, 0.1, and 1 g/kg restored this response to 50.8 ± 1.5% (p < 0.0001), 53.1 ± 1.9% (p < 0.0001), and 56.5 ± 2.8% (p < 0.0001), respectively (Figure 8B). These results suggest that SGR restores the ITR in GMD mice.

3. Discussion

As a result of the analysis through network pharmacology based on these previous studies, 74 compounds including 17 active compounds were identified in SGR (Supplementary Materials Table S2). Of the 74 compounds found, 56 compounds had target information, and a total of 390 target genes were collected (Supplementary Materials Table S3). Seventeen of these compounds were associated with GMD (Figure 5), and GMD-related genes belonging to the target genes of the active compounds of SGR were ACHE, CCK, CXCL8, DPP4, FOS, GCG, IL10, IL1B, IL6, INS, MPO, OPRM1, PYY, SCN5A, SLC6A4, TNF, NOS1, HTR3A and TRPV1 (Figure 4). These results are consistent with previous studies. Specifically, SCN5A was the target of many compounds, including quercetin (Table 2, Figure 5). Nav1.5 encoded by the SCN5A gene is mediated by the rapid introduction of Na+ ions (INa). Voltage-gated Na+ channels are important for excitation and propagation of electrical impulses in excitable cells, for example nerve or cardiomyocytes [14]. SCN5A is known to contribute to improving gastrointestinal motility [15]. In addition, TNF, a target gene of several compounds including quercetin, is also known to be closely related to intestinal motility [16]. In addition, FOS, a target gene for several compounds including quercetin, is considered a metabolic marker of nerve activation and is also related to gastrointestinal motility [17]. As shown in Figure 5, quercetin (11), resveratrol (8), oleic acid (5), and beta-sitosterol (4) were identified as compounds with many target genes among compounds related to GMD. Several studies have reported the relationship between key compounds and GMD. It is known that quercetin, which has the most target genes, blocks Ca2+ channels and inhibits intestinal contraction [18]. In addition, resveratrol was reported to have a function of inducing the relaxation of gastric smooth muscle through high-conductivity calcium-activated potassium channels [19]. Oleic acid is reported to slow gastrointestinal transit and reduce diarrhea by activating a nutrient-induced inhibitory feedback mechanism [20]. In this mouse study, we studied the effect of the SGR extract on pacemaker activity in ICC and gastric emptying. In addition, we examined GI function by the ITR in normal or experimentally induced GMD mice. The SGR extract depolarized the pacemaker activity in ICC (Figure 6) and restored the delayed gastric emptying (Figure 7). In addition, oral administration of the SGR extract increased the ITR and restored ITR delay (Figure 8). AA induction showed a significant decrease in the ITR. This reduction was largely blocked by the SGR extract. As a result of the network analysis of this study, SGR is strongly related to SCN5A, TNF, and FOS, which are genes highly related to intestinal motility [14,15,16,17], and oleic acid, quercetin, and resveratrol, which are major compounds of SGR also known to have an effect on intestinal motility [18,19,20]. In general, traditional herbal medicines are known to be contaminated with heavy metals, microorganisms, pesticides, etc., and can cause serious side effects [21]. In addition, if herbal medicines change in a similar shape, serious complications occur [21]. Side effects caused by SGR are not well known and are known to cause hepatotoxicity, a common side effect of herbal medicines [22]. In addition, the Chinese classic book shows that the LD50 in mice is 161 g/kg and is 100 g/kg in rats; 45 consecutive days of administration at these concentrations resulted in significant experimental animal death (p < 0.01), and rats’ BUN exceeded the normal range [23]. In addition, when administered at 50 g/kg to rats for 60 days, abnormalities in BUN occurred partially, but recovered after discontinuation of administration, and it was reported that degenerative necrosis appeared in hepatocytes and renal cells [23]. Therefore, in this paper, when administered at a maximum SGR of 5 g/kg for 14 days, there was no special change in important organs, so it is considered that there was no toxic reaction.
ICC are cells that control the pacemaker of the gastrointestinal tract and play an important role in regulating gastrointestinal motility [24]. Therefore, it is known that problems with ICC cause various gastrointestinal motility diseases, and ICC are an important research tool in the study of functional gastrointestinal motility regulation. In this study, it is thought that the SGR extract causes depolarization of ICC (Figure 6), and as a result of designing an experiment and confirming the gastric emptying and ITR, it was confirmed that decreased gastric emptying and intestinal mobility was restored through the SRG extract (Figure 7 and Figure 8).
It takes about 60 to 100 min for solid food and about 10 to 45 min for food containing liquid to move to the duodenum from the stomach. After about 180 min, about 70% of the food must be digested. An abnormal gastric emptying is called Gastroparesis. Gastroparesis is a syndrome that is expressed as symptoms such as nausea, vomiting, and abdominal pain due to various causes [25]. In the United States, it is observed in about 3% of the total population, and the prevalence is known to be somewhat higher in women [26]. According to recent research results, there are also reports that the prevalence rate has increased compared to the past [27]. It is reported that 30–50% of patients with type 1 diabetes and 15–30% of patients with type 2 diabetes have Gastroparesis [28], and it is on the rise recently. Intestinal motility is movement caused by the digestion of the intestines. It refers to sending food digested in the stomach to the spring intestines and large intestine. Impairment of bowel motility occurs everywhere in the gastrointestinal tract. In particular, irritable bowel syndrome (IBS) is a general abnormal condition of the intestine that shows symptoms such as abdominal pain, abdominal cramps, abdominal distension, bowel pain, and nausea [29]. The etiology of IBS is largely unknown or incomprehensible, and therefore there is no complete cure [29]. Despite the importance of gastrointestinal motility, it is difficult to choose suitable treatment because the pathophysiological stage varies due to delayed gastric emptying, hypersensitivity to gastric expansion, adverse reactions to duodenal fat or acid, and psychoneurotic disorders [30]. According to this trend, various traditional medical treatment methods and approaches to treatment of gastrointestinal motility abnormalities are being made [31]. Research on gastrointestinal motility abnormalities in traditional medicine has been actively conducted [31]. It was found that the possibility of controlling gastrointestinal movement and reduced gastrointestinal movement increased due to various traditional medicines, such as Pyungwi-san [32], Lizhong Tang [33], Poncirus trifoliate fruit [34], So-Cheong-Ryong-Tang [35], San-Huang-Xie-Xin-tang [36], Schisandra chinensis [37], Ge-Gen-Tang [38], Liriope Platyphylla Wang Et tang [39], Hwangryunhaedok-tang [40], Gamisoyo-San [41], Banhasasim-Tang [42], and Pinellia ternata Breitenbach [43]. In order for the oriental medical approach to become an efficient approach in controlling gastrointestinal motility, large-scale clinical studies are needed to prove the reproducibility and validity of the effects.

4. Materials and Methods

4.1. Network Pharmacology Analysis by SGR

4.1.1. Classification of the Compound of SGR

In order to classify the potential active compounds of SGR, TCMSP databases and analysis platforms were used to investigate. We entered ‘Smilacis Glabrae Rhixoma’ as the query to search with the herb name.

4.1.2. Analysis of Target

Target information of compounds was found using search through TCMSP [44]. We used the UniProtKB database to link the target protein to the official genetic name (https://www.uniprot.org/uniprot; accessed on 4 July 2022) [45].

4.1.3. Analysis of Network

Cytoscape 3.9.2 (https://cytoscape.org; accessed on 4 July 2022) was used to build the compound–target network [46]. GMD-related genes were collected by Cytoscape App. using ‘Gastrointestinal mobility disorder’ as a query, which integrates evidence and updates weekly [47].

4.1.4. Active Compound Screening

On the basis of absorption, distribution, metabolism, and excretion (ADME) parameters such as molecular weight (MW), oral bioavailability (OB), Caco-2 permeability (Caco-2), and drug likeness (DL), physiologically active compounds of SGR were screened using the following criteria: OB ≥ 30%, DL ≥ 0.10, and Caco-2 ≥ −0.4. The compounds that meet the applied criteria were recognized as active compounds.

4.2. Animal Validation

4.2.1. SGR

SGR was purchased from Korea Plant Extract Bank (Cheongju, Republic of Korea). SGR was dissolved in distilled water and mice were administered doses in the quantities of 0.01, 0.1, and 1 g/kg. To check the toxicity of SGR extract in mice, it was administered intragastrically through an orogastric tube at different doses (0, 0.5, 1, 2, or 5 g/kg of SGR extract delivered at 10 mL/kg). Seven mice of each gender were tested, and thus, a total of 70 mice were used. Each group was carefully observed for overt clinical signs and mortality at hourly intervals for 6 h after administration, and then on a daily basis for 14 days. Individual body weights were measured before dosing and on days 1, 3, 7, and 14 after administration. At 14 days, the last day of observation, animals were necropsied, and vital organs and tissues appeared normal by gross inspection. Accordingly, the SGR extract appeared to be safe and did not elicit acute toxicity for a single oral dose of at least 5 g/kg or below. In this paper, SGR 0.01 g/kg, 0.1 g/kg, and 1 g/kg were used in the animal experiment, and this concentration has been widely used in the mouse gastrointestinal animal experiments [33,37,40,41,42,48,49,50,51]. When converted to human concentration, 1 g/kg for a mouse is 4.9 g for adults (60 kg) [52], which is less than the typical human dose of 15–60 g [23]. In addition, in rats, SGR was used at a similar concentration to this paper [53].

4.2.2. Animal Experiments

A total of 48 mice (Control (n = 12, male 6: female 6), SGR 1 mg (n = 12, 6:6), SGR 3 mg (n = 12, 6:6), and SGR 5 mg (n = 12, 6:6); 4–8 days old; weighing 2.0–2.3 g) of the Institute of Cancer Research (ICR) mice from Samtako Bio Korea Co., Ltd. (Osan, Republic of Korea) were used for the ICC experiments. A total of 86 male mice (Naïve (n = 15), Control (n = 15), SGR 0.01 mg (n = 14), SGR 0.1 mg (n = 15), SGR 1 mg (n = 14), Mosapride (n = 13)); 7 weeks old; weighing 23–26 g) for the gastric emptying experiments. Also, 59 male mice (Naïve (n = 15), SGR 0.01 mg (n = 15), SGR 0.1 mg (n = 14), SGR 1 mg (n = 15)); 7 weeks old; weighing 23–26 g) for the normal ITR experiments and 74 male mice (Naïve (n = 15), Control (n = 15); SGR 0.01 mg (n = 15), SGR 0.1 mg (n = 15), SGR 1 mg (n = 14); 7 weeks old; weighing 23–26 g) were used for the ITR experiments with GMD. All mice were housed in a specific pathogen-free laboratory environment under controlled temperature (20 ± 2 °C) and humidity (49 ± 5%) with day and night cycles (light on at 7:00 a.m. and light off at 7:00 p.m.) and ad libitum access to normal diet and autoclaved water. During the study, indicators of the general condition of the mice were observed daily, such as fur brightness, food and water intake, defecation, and behavior. In the ICC experiments, ICC were placed under a microscope after making the cells, and SGR was administered to the cells to confirm the change in pacemaker potentials. In the gastric emptying and ITR experiments, SGR was administered directly to the mouse mouth.

4.2.3. Preparation of ICC and Electrophysiological Experiments

After taking out the small intestine, the contents of the small intestine were removed with a Kreb Ringer solution. The tissues were pinned to the sylgard dish, and the mucous membrane was removed with scissors. The small intestine tissue was finely cut and then immersed in Hank solution (KCl 5.36 mM, NaCl 125 mM, NaOH 0.34 mM, Na2HCO3 0.44 mM, Glucose 10 mM, Sucrose 2.9 mM, and Hepes 11 mM) for 30 min. The cells were then separated in a solution containing various enzymes. The cells were sprayed on collagen-coded cover glass and maintained in a 37 °C environment in media containing smooth muscle growth medium (Clonetics, San Diego, CA, USA) and stem cell factor (Sigma-Aldrich, St. Louis, MO, USA). The whole-cell patch clamp method was used to measure the change in the membrane potential in the cultured ICC. Axopatch 200B (Axon Instruments, Foster, CA, USA) was used as an amplifier, and the results were summarized using pCLAMP and Origin (version 2018; MicroCal, Northampton, MA, USA). All experiments were conducted between 30 °C and 33 °C.

4.2.4. Gastric Weight and Gastric Emptying by Phenol Red

Gastric emptying was assessed by administering a 0.05% (w/v) phenol red solution (0.5 mL/mouse) 30 min after administering SGR. The mice were euthanized 30 min after treatment with phenol red, their stomachs were removed immediately, and the weights were measured. Next, the stomach was treated with 5 mL of 0.1 N sodium hydroxide solution to check the optical density of the phenol red remaining in the stomach. The homogenate was centrifuged at 3000 rpm for 20 min. Then, 1 mL of the supernatant was added to 4 mL of 0.5N sodium hydroxide solution, and the optical density of this pink liquid was measured using a spectrophotometer (560 nm). For experiments, mice were fasted for 19 h with a free supply of tap water. The selection of the phenol red solution volume (500 µL) and the 50% delayed gastric emptying time point (30 min after intraperitoneal injection of 10 mg/kg of loperamide) was conducted according to earlier study protocols [54,55]. The above emission values were obtained according to the following formula: Gastric emptying (%) = (1 − X/Y) × 100. X: Optical density of the phenol red remaining on it. Y: Optical density of the phenol red mixture with sodium hydroxide under test tube conditions.

4.2.5. Intestinal Transit Rate Evaluation

Evans Blue was orally administered after SGR administration, and animals were sacrificed 30 min later. The ITR was measured by checking the length that Evans Blue passed in the intestine. The ITR was measured as a percentage of the total length passed by Evans Blue. A peritoneal irritation by the acetic acid (PIA) model, one of the experimental GMD models, was used. Peritoneal irritation was induced using acetic acid (AA) in mice 30 min after the intragastric administration of SGR. PIA was induced by an intraperitoneal (i.p.) injection of acetic acid (0.6%, w/v, in saline) at a dose of 10 mL/kg, as previously described [37,39]. After injecting acetic acid, mice were placed in individual cages and allowed to recover for 30 min.

4.2.6. Statistical Analysis

The results were represented as mean ± standard error of the mean. We first checked the normality and homogeneity and then analyzed the results with ANOVA with Bonferroni post hoc tests for multiple comparisons and Prism 6.0 (La Jolla, CA, USA) programs; p < 0.05 was considered statistically important.

5. Conclusions

Alternative medicine is still not recognized as general medicine. Nevertheless, around 51% of people with gastrointestinal disorders are receiving help from alternative medicine, and 10% of all alternative medicines are for digestive problems. In addition, herbal medicines are a source of alternative medicines and are known to have fewer side effects. An association of SGR with gastrointestinal motility was found through network pharmacological analysis, and recovery of gastric emptying and intestinal motility were confirmed through mouse experiments. Through this, the great potential of SGR as a powerful traditional medicine treatment for GMD was discovered.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/plants12071509/s1, Table S1: Potential active compounds of Smilacis Glabrae Rhizoma. Table S2: Target genes of Smilacis Glabrae Rhizoma. Table S3: One hundred gastrointestinal-motility-disorder-related genes.

Author Contributions

Conceptualization, N.-R.C., K.L., J.-W.P. and B.-J.K.; methodology, N.-R.C., K.L., M.S. and B.-J.K.; software, N.-R.C., K.L. and M.S.; validation, N.-R.C., K.L. and M.S.; formal analysis, N.-R.C., K.L., M.S., J.K., J.-W.P. and B.-J.K.; investigation, N.-R.C., K.L., M.S., S.-J.K. and W.-G.C.; resources, N.-R.C., K.L., M.S., S.-J.K. and S.-C.K.; data curation, N.-R.C., K.L., M.S., S.-J.K., W.-G.C., J.K., J.-W.P. and B.-J.K.; writing—original draft preparation, N.-R.C., K.L., M.S., J.-W.P. and B.-J.K.; writing—review and editing, S.-C.K., J.K., J.-W.P. and B.-J.K.; visualization, N.-R.C. and B.-J.K.; supervision, J.-W.P. and B.-J.K.; project administration, J.-W.P. and B.-J.K.; funding acquisition, J.-W.P. and B.-J.K. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2021R1I1A3042479) to B.J.K. and by the Korea Health Industry Development Institute (grant number HI20C0145) to J.-W.P.

Institutional Review Board Statement

The animal protocol used in this study has been reviewed by the Pusan National University Institutional Animal Care and Use Committee (PNU-IACUC) on their ethical procedures and scientific care, and it has been approved (Approval Number PNU-2022-0120).

Informed Consent Statement

Not applicable.

Data Availability Statement

The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lacy, B.E.; Weiser, K. Gastrointestinal Motility Disorders: An Update. Dig. Dis. 2006, 24, 228–242. [Google Scholar] [CrossRef]
  2. On behalf of the International Working Group for Disorders of Gastrointestinal Motility and Function; Keller, J.; Bassotti, G.; Clarke, J.; Dinning, P.; Fox, M.; Grover, M.; Hellström, P.M.; Ke, M.; Layer, P.; et al. Advances in the Diagnosis and Classification of Gastric and Intestinal Motility Disorders. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 291–308. [Google Scholar] [CrossRef] [Green Version]
  3. Huizinga, J.D.; Lammers, W.J.E.P. Gut Peristalsis Is Governed by a Multitude of Cooperating Mechanisms. Am. J. Physiol. Gastrointest. Liver Physiol. 2009, 296, G1–G8. [Google Scholar] [CrossRef] [PubMed]
  4. Huizinga, J.D. Gastrointestinal Peristalsis: Joint Action of Enteric Nerves, Smooth Muscle, and Interstitial Cells of Cajal. Microsc. Res. Tech. 1999, 47, 239–247. [Google Scholar] [CrossRef]
  5. Lee, J.; Li, C.; Surayot, U.; Yelithao, K.; Lee, S.; Park, W.; Tabarsa, M.; You, S. Molecular Structures, Chemical Properties and Biological Activities of Polysaccharide from Smilax Glabra Rhizome. Int. J. Biol. Macromol. 2018, 120, 1726–1733. [Google Scholar] [CrossRef] [PubMed]
  6. Chinese Pharmacopoeia Commission. Smilacis Glabrae Rhizoma. Pharmacopoeia of the People’s Republic of China; Medical Science Press: Beijing, China, 2020; Volume I, p. 19. [Google Scholar]
  7. Shi, Y.; Tian, C.; Yu, X.; Fang, Y.; Zhao, X.; Zhang, X.; Xia, D. Protective Effects of Smilax Glabra Roxb. Against Lead-Induced Renal Oxidative Stress, Inflammation and Apoptosis in Weaning Rats and HEK-293 Cells. Front. Pharmacol. 2020, 11, 556248. [Google Scholar] [CrossRef] [PubMed]
  8. Park, G.; Kim, T.; Kim, J.H.; Oh, M.S. Antioxidant Effects of the Sarsaparilla via Scavenging of Reactive Oxygen Species and Induction of Antioxidant Enzymes in Human Dermal Fibroblasts. Environ. Toxicol. Pharmacol. 2014, 38, 305–315. [Google Scholar] [CrossRef] [PubMed]
  9. Zhang, C.; Xu, Q.; Tan, X.; Meng, L.; Wei, G.; Liu, Y.; Zhang, C. Astilbin Decreases Proliferation and Improves Differentiation in HaCaT Keratinocytes. Biomed. Pharmacother. 2017, 93, 713–720. [Google Scholar] [CrossRef]
  10. Kwon, O.Y.; Ryu, S.; Choi, J.K.; Lee, S.H. Smilax Glabra Roxb. Inhibits Collagen Induced Adhesion and Migration of PC3 and LNCaP Prostate Cancer Cells through the Inhibition of Beta 1 Integrin Expression. Molecules 2020, 25, 3006. [Google Scholar] [CrossRef]
  11. Sa, F.; Gao, J.-L.; Fung, K.-P.; Zheng, Y.; Lee, S.M.-Y.; Wang, Y.-T. Anti-Proliferative and pro-Apoptotic Effect of Smilax Glabra Roxb. Extract on Hepatoma Cell Lines. Chem. Biol. Interact. 2008, 171, 1–14. [Google Scholar] [CrossRef]
  12. Wang, S.; Fang, Y.; Yu, X.; Guo, L.; Zhang, X.; Xia, D. The Flavonoid-Rich Fraction from Rhizomes of Smilax Glabra Roxb. Ameliorates Renal Oxidative Stress and Inflammation in Uric Acid Nephropathy Rats through Promoting Uric Acid Excretion. Biomed. Pharmacother. 2019, 111, 162–168. [Google Scholar] [CrossRef] [PubMed]
  13. Zhang, R.; Zhu, X.; Bai, H.; Ning, K. Network Pharmacology Databases for Traditional Chinese Medicine: Review and Assessment. Front. Pharmacol. 2019, 10, 123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Gellens, M.E.; George, A.L.; Chen, L.Q.; Chahine, M.; Horn, R.; Barchi, R.L.; Kallen, R.G. Primary Structure and Functional Expression of the Human Cardiac Tetrodotoxin-Insensitive Voltage-Dependent Sodium Channel. Proc. Natl. Acad. Sci. USA 1992, 89, 554–558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Verstraelen, T.E.; ter Bekke, R.M.A.; Volders, P.G.A.; Masclee, A.A.M.; Kruimel, J.W. The Role of the SCN5A-Encoded Channelopathy in Irritable Bowel Syndrome and Other Gastrointestinal Disorders. Neurogastroenterol. Motil. 2015, 27, 906–913. [Google Scholar] [CrossRef] [PubMed]
  16. Kinoshita, K.; Hori, M.; Fujisawa, M.; Sato, K.; Ohama, T.; Momotani, E.; Ozaki, H. Role of TNF-Alpha in Muscularis Inflammation and Motility Disorder in a TNBS-Induced Colitis Model: Clues from TNF-Alpha-Deficient Mice. Neurogastroenterol. Motil. 2006, 18, 578–588. [Google Scholar] [CrossRef]
  17. Zhang, S.; Liu, Y.; Li, S.; Ye, F.; Foreman, R.D.; Chen, J.D.Z. Effects of Electroacupuncture on Stress-Induced Gastric Dysrhythmia and Mechanisms Involving Autonomic and Central Nervous Systems in Functional Dyspepsia. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2020, 319, R106–R113. [Google Scholar] [CrossRef]
  18. Nigusse, T.; Zhang, L.; Wang, R.; Wang, X.; Li, J.; Liu, C. Flavonoids in a Crude Extract of Catha Edulis Inhibit Rat Intestinal Contraction via Blocking Ca2+ Channels. Neurogastroenterol. Motil. 2019, 31, e13602. [Google Scholar] [CrossRef]
  19. Modzelewska, B.; Drygalski, K.; Hady, H.R.; Kiełczewska, A.; Chomentowski, A.; Koryciński, K.; Głuszyńska, P.; Kleszczewski, T. Resveratrol Relaxes Human Gastric Smooth Muscles Through High Conductance Calcium-Activated Potassium Channel in a Nitric Oxide-Independent Manner. Front. Pharmacol. 2022, 13, 823887. [Google Scholar] [CrossRef]
  20. Lin, H.C.; van Citters, G.W.; Heimer, F.; Bonorris, G. Slowing of Gastrointestinal Transit by Oleic Acid: A Preliminary Report of a Novel, Nutrient-Based Treatment in Humans. Dig. Dis. Sci. 2001, 46, 223–229. [Google Scholar] [CrossRef]
  21. Xu, B.; Cheng, Q.; So, W.K.W. Review of the Effects and Safety of Traditional Chinese Medicine in the Treatment of Cancer Cachexia. Asia Pac. J. Oncol. Nurs. 2021, 8, 471–486. [Google Scholar] [CrossRef]
  22. Cheng, S.; Sun, H.; Li, X.; Yan, J.; Peng, Z.; You, Y.; Zhang, L.; Chen, J. Effects of Alismatis Rhizoma and Rhizoma Smilacis Glabrae Decoction on Hyperuricemia in Rats. Evid. Based Complement. Alternat. Med. 2019, 2019, 4541609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Zhang, M.Z.; Dong, Z.H.; She, J. Modern Study of Traditional Chinese Medicine; Xueyuan Press: Beijing, China, 1997; Volume 1, p. 316. [Google Scholar]
  24. Kim, B.J.; Lim, H.H.; Yang, D.K.; Jun, J.Y.; Chang, I.Y.; Park, C.S.; So, I.; Stanfield, P.R.; Kim, K.W. Melastatin-type transient receptor potential channel 7 is required for intestinal pacemaking activity. Gastroenterology 2005, 129, 1504–1517. [Google Scholar] [CrossRef] [PubMed]
  25. Camilleri, M.; Chedid, V.; Ford, A.C.; Haruma, K.; Horowitz, M.; Jones, K.L.; Low, P.A.; Park, S.Y.; Parman, H.P.; Stanghellini, V. Gastroparesis. Nat. Rev. Dis. Primers 2018, 4, 41. [Google Scholar] [CrossRef] [PubMed]
  26. Jung, H.K.; Choung, R.S.; Locke, G.R., 3rd; Schleck, C.D.; Zinsmeister, A.R.; Szarka, L.A.; Mullan, B.; Talley, N.J. The incidence, prevalence, and outcomes of patients with gastroparesis in Olmsted County, Minnesota, from 1996 to 2006. Gastroenterology 2009, 136, 1225–1233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Moshiree, B.; Potter, M.; Talley, N.J. Epidemiology and pathophysiology of gastroparesis. Gastrointest. Endosc. Clin. 2019, 29, 1–14. [Google Scholar] [CrossRef] [PubMed]
  28. Choung, R.S.; Locke, G.R., 3rd; Schleck, C.D.; Zinsmeister, A.R.; Melton, L.J., 3rd; Talley, N.J. Risk of gastroparesis in subjects with type 1 and 2 diabetes in the general population. Am. J. Gastroenterol. 2012, 107, 82–88. [Google Scholar] [CrossRef]
  29. McHarg, A.S.; Leach, S. The role of the gut microbiome in paediatric irritable bowel syndrome. AIMS Microbiol. 2022, 8, 454–469. [Google Scholar] [CrossRef]
  30. Singh, R.; Zogg, H.; Ghoshal, U.C.; Ro, S. Current Treatment Options and Therapeutic Insights for Gastrointestinal Dysmotility and Functional Gastrointestinal Disorders. Front. Pharmacol. 2022, 13, 808195. [Google Scholar] [CrossRef]
  31. Sankararaman, S.; Velayuthan, S.; Chen, Y.; Robertson, J.; Sferra, T.J. Role of Traditional Chinese Herbal Medicines in Functional Gastrointestinal and Motility Disorders. Curr. Gastroenterol. Rep. 2022, 24, 43–51. [Google Scholar] [CrossRef]
  32. Kim, J.N.; Song, H.J.; Lim, B.; Kwon, Y.K.; Kim, B.J. Modulation of pacemaker potentials by pyungwi-san in interstitial cells of cajal from murine small intestine: Pyungwi-san and interstitial cells of Cajal. J. Pharmacopunct. 2013, 16, 43–49. [Google Scholar] [CrossRef]
  33. Lee, M.C.; Ha, W.; Park, J.; Kim, J.; Jung, Y.; Kim, B.J. Effects of Lizhong Tang on gastrointestinal motility in mice. World J. Gastroenterol. 2016, 22, 7778–7786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Kim, B.J.; Kim, H.W.; Lee, G.S.; Choi, S.; Jun, J.Y.; So, I.; Kim, S.J. Poncirus trifoliate fruit modulates pacemaker activity in interstitial cells of Cajal from the murine small intestine. J. Ethnopharmacol. 2013, 149, 668–675. [Google Scholar] [CrossRef] [PubMed]
  35. Hwang, M.W.; Lee, H.J.; Song, H.J.; Kim, B.J. Involvement of MAPKs and PLC pathways in modulation of pacemaking activity by So-Cheong-Ryong-Tang in interstitial cells of Cajal from murine small intestine. Sci. World J. 2013, 2013, 536350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Kim, B.J.; Kim, H.; Lee, G.S.; So, I.; Kim, S.J. Effects of San-Huang-Xie-Xin-tang, a traditional Chinese prescription for clearing away heat and toxin, on the pacemaker activities of interstitial cells of Cajal from the murine small intestine. J. Ethnopharmacol. 2014, 155, 744–752. [Google Scholar] [CrossRef] [PubMed]
  37. Ahn, T.S.; Kim, D.G.; Hong, N.R.; Park, H.S.; Kim, H.; Ha, K.T.; Jeon, J.H.; So, I.; Kim, B.J. Effects of Schisandra chinensis extract on gastrointestinal motility in mice. J. Ethnopharmacol. 2015, 169, 163–169. [Google Scholar] [CrossRef] [PubMed]
  38. Lee, S.; Gim, H.; Shim, J.H.; Kim, J.H.; Lee, J.R.; Kim, S.C.; Kwon, Y.K.; Ha, K.T.; So, I.; Kim, B.J. The traditional herbal medicine, Ge-Gen-Tang, inhibits pacemaker potentials by nitric oxide/cGMP dependent ATP-sensitive K(+) channels in cultured interstitial cells of Cajal from mouse small intestine. J. Ethnopharmacol. 2015, 170, 201–209. [Google Scholar] [CrossRef]
  39. Kim, H.J.; Park, S.Y.; Kim, D.G.; Park, S.H.; Lee, H.; Hwang, D.Y.; Jung, M.H.; Ha, K.T.; Kim, B.J. Effects of the roots of Liriope Platyphylla Wang Et tang on gastrointestinal motility function. J. Ethnopharmacol. 2016, 184, 144–153. [Google Scholar] [CrossRef]
  40. Kim, H.; Kim, I.; Lee, M.C.; Kim, H.J.; Lee, G.S.; Kim, H.; Kim, B.J. Effects of Hwangryunhaedok-tang on gastrointestinal motility function in mice. World J. Gastroenterol. 2017, 23, 2705–2715. [Google Scholar] [CrossRef]
  41. Shin, S.J.; Kim, D.; Kim, J.S.; Kim, I.; Lee, J.R.; Kim, S.C.; Kim, B.J. Effects of Gamisoyo-San Decoction, a Traditional Chinese Medicine, on Gastrointestinal Motility. Digestion 2018, 98, 231–237. [Google Scholar] [CrossRef]
  42. Kim, J.N.; Nam, J.H.; Lee, J.R.; Kim, S.C.; Kim, B.J. The Traditional Medicine Banhasasim-Tang Depolarizes Pacemaker Potentials of Cultured Interstitial Cells of Cajal through M3 Muscarinic and 5-HT3 Receptors in Murine Small Intestine. Digestion 2020, 101, 536–551. [Google Scholar] [CrossRef]
  43. Choi, N.R.; Park, J.; Ko, S.J.; Kim, J.N.; Choi, W.; Park, J.W.; Kim, B.J. Prediction of the Medicinal Mechanisms of Pinellia ternata Breitenbach, a Traditional Medicine for Gastrointestinal Motility Disorders, through Network Pharmacology. Plants 2022, 11, 1348. [Google Scholar] [CrossRef]
  44. Ru, J.; Li, P.; Wang, J.; Zhou, W.; Li, B.; Huang, C.; Li, P.; Guo, Z.; Tao, W.; Yang, Y.; et al. TCMSP: A Database of Systems Pharmacology for Drug Discovery from Herbal Medicines. J. Cheminform. 2014, 6, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. The UniProt Consortium UniProt: A Worldwide Hub of Protein Knowledge. Nucleic Acids Res. 2019, 47, D506–D515. [CrossRef] [PubMed] [Green Version]
  46. Shannon, P.; Markiel, A.; Ozier, O.; Baliga, N.S.; Wang, J.T.; Ramage, D.; Amin, N.; Schwikowski, B.; Ideker, T. Cytoscape: A Software Environment for Integrated Models of Biomolecular Interaction Networks. Genome Res. 2003, 13, 2498–2504. [Google Scholar] [CrossRef] [PubMed]
  47. Doncheva, N.T.; Morris, J.H.; Gorodkin, J.; Jensen, L.J. Cytoscape StringApp: Network Analysis and Visualization of Proteomics Data. J. Proteome Res. 2019, 18, 623–632. [Google Scholar] [CrossRef]
  48. Lee, H.T.; Seo, E.K.; Chung, S.J.; Shim, C.K. Effect of an aqueous extract of dried immature fruit of Poncirus trifoliate (L.) Raf. on intestinal transit in rodents with experimental gastrointestinal motility dysfunctions. J. Ethnopharmacol. 2005, 102, 302–306. [Google Scholar] [CrossRef] [PubMed]
  49. Kim, H.J.; Han, T.; Kim, Y.T.; So, I.; Kim, B.J. Magnolia Officinalis Bark Extract Induces Depolarization of Pacemaker Potentials Through M2 and M3 Muscarinic Receptors in Cultured Murine Small Intestine Interstitial Cells of Cajal. Cell. Physiol. Biochem. 2017, 43, 1790–1802. [Google Scholar] [CrossRef] [Green Version]
  50. Han, D.; Kim, J.N.; Kwon, M.J.; Han, T.; Kim, Y.T.; Kim, B.J. Salvia miltiorrhiza induces depolarization of pacemaker potentials in murine small intestinal interstitial cells of Cajal via extracellular Ca2+ and Na+ influx. Mol. Med. Rep. 2021, 23, 348. [Google Scholar] [CrossRef]
  51. Moon, S.B.; Choi, N.R.; Kim, J.N.; Kwon, M.J.; Kim, B.S.; Ha, K.T.; Lim, E.Y.; Kim, Y.T.; Kim, B.J. Effects of black garlic on the pacemaker potentials of interstitial cells of Cajal in murine small intestine in vitro and on gastrointestinal motility in vivo. Anim. Cells Syst. 2022, 26, 37–44. [Google Scholar] [CrossRef]
  52. Nair, A.B.; Jacob, S. A simple practice guide for dose conversion between animals and humans. J. Basic Clin. Pharm. 2016, 7, 27–31. [Google Scholar] [CrossRef] [Green Version]
  53. Zou, W.; Zhou, H.; Hu, J.; Zhang, L.; Tang, Q.; Wen, X.; Xiao, Z.; Wang, W. Rhizoma Smilacis Glabrae inhibits pathogen-induced upper genital tract inflammation in rats through suppression of NF-κB pathway. J. Ethnopharmacol. 2017, 202, 103–113. [Google Scholar] [CrossRef] [PubMed]
  54. Asano, T.; Aida, S.; Suemasu, S.; Mizushima, T. Anethole restores delayed gastric emptying and impaired gastric accommodation in rodents. Biochem. Biophys. Res. Commun. 2016, 472, 125–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Scarpignato, C.; Capovilla, T.; Bertaccini, G. Action of caerulein on gastric emptying of the conscious rat. Arch. Int. Pharmacodyn. Ther. 1980, 246, 286–294. [Google Scholar] [PubMed]
Figure 1. Schematic of study.
Figure 1. Schematic of study.
Plants 12 01509 g001
Figure 2. Compound–target network of SGR. The node size depends on the connected edges’ number. The compounds are expressed as orange diamond-shaped nodes, and the targets are expressed as green square-shaped nodes. SGR: Smilacis Glabrae Rhixoma.
Figure 2. Compound–target network of SGR. The node size depends on the connected edges’ number. The compounds are expressed as orange diamond-shaped nodes, and the targets are expressed as green square-shaped nodes. SGR: Smilacis Glabrae Rhixoma.
Plants 12 01509 g002
Figure 3. Active compounds and gastrointestinal (GI)-related compounds of SGR. The Venn diagram between active compounds and GI-disease-related compounds in SGR. SGR: Smilacis Glabrae Rhixoma.
Figure 3. Active compounds and gastrointestinal (GI)-related compounds of SGR. The Venn diagram between active compounds and GI-disease-related compounds in SGR. SGR: Smilacis Glabrae Rhixoma.
Plants 12 01509 g003
Figure 4. Network of GMD-related genes and SGR target genes. The 19 genes included in both “gene related to gastrointestinal motility disorder” and “Smilacis Glabrae Rhixoma target genes” are classified in the center. GMD: gastrointestinal motility disorder. SGR: Smilacis Glabrae Rhixoma.
Figure 4. Network of GMD-related genes and SGR target genes. The 19 genes included in both “gene related to gastrointestinal motility disorder” and “Smilacis Glabrae Rhixoma target genes” are classified in the center. GMD: gastrointestinal motility disorder. SGR: Smilacis Glabrae Rhixoma.
Plants 12 01509 g004
Figure 5. Network of compounds of SGR and GMD related genes. GMD: Gastrointestinal Motility Disorder. SGR: Smilacis Glabrae Rhixoma.
Figure 5. Network of compounds of SGR and GMD related genes. GMD: Gastrointestinal Motility Disorder. SGR: Smilacis Glabrae Rhixoma.
Plants 12 01509 g005
Figure 6. Effects of SGR extract on pacemaker activity of ICC. (AC) SGR depolarized the ICC pacemaker activity; (D) the depolarization changes; (E) the frequency changes. CTRL (n = 12). SGR 1 mg (n = 12). SGR 3 mg (n = 12). SGR 5 mg (n = 12). Bars represent mean ± standard error. **** p < 0.0001. CTRL: Control. SGR: Smilacis Glabrae Rhixoma. ICC: interstitial cells of Cajal.
Figure 6. Effects of SGR extract on pacemaker activity of ICC. (AC) SGR depolarized the ICC pacemaker activity; (D) the depolarization changes; (E) the frequency changes. CTRL (n = 12). SGR 1 mg (n = 12). SGR 3 mg (n = 12). SGR 5 mg (n = 12). Bars represent mean ± standard error. **** p < 0.0001. CTRL: Control. SGR: Smilacis Glabrae Rhixoma. ICC: interstitial cells of Cajal.
Plants 12 01509 g006
Figure 7. Effects of SGR extract on delayed gastric emptying. (A) summary of the experimental schedule; (B) visualization; (C) stomach weight; (D) gastric emptying. Naïve (n = 15). CTRL (n = 15). SGR 0.01 g (n = 14). SGR 0.1 g (n = 15). SGR 1 g (n = 14). Mosapride (n = 13). Bars represent mean ± standard error. * p < 0.05, ** p < 0.01 for the Control group; #### p < 0.0001 for the loperamide group. CTRL: Control. SGR: Smilacis Glabrae Rhixoma.
Figure 7. Effects of SGR extract on delayed gastric emptying. (A) summary of the experimental schedule; (B) visualization; (C) stomach weight; (D) gastric emptying. Naïve (n = 15). CTRL (n = 15). SGR 0.01 g (n = 14). SGR 0.1 g (n = 15). SGR 1 g (n = 14). Mosapride (n = 13). Bars represent mean ± standard error. * p < 0.05, ** p < 0.01 for the Control group; #### p < 0.0001 for the loperamide group. CTRL: Control. SGR: Smilacis Glabrae Rhixoma.
Plants 12 01509 g007
Figure 8. Effect of SGR extract on ITR in normal and AA-induced GMD mice. (A) In normal mice, SGR extract increased ITR. Naïve (n = 15). SGR 0.01 g (n = 15). SGR 0.1 g (n = 14). SGR 1 g (n = 15). (B) In this AA-induced case, the ITR was recovered by SGR extract. Naïve (n = 15). CTRL (n = 15). SGR 0.01 g (n = 15). SGR 0.1 g (n = 15). SGR 1 g (n = 14). Bars represent mean ± standard error. * p < 0.05. *** p < 0.001. **** p < 0.0001. CTRL: Control. SGR: Smilacis Glabrae Rhixoma. ITR: intestinal transit rate. AA: acetic acid. GMD: gastrointestinal motility disorder.
Figure 8. Effect of SGR extract on ITR in normal and AA-induced GMD mice. (A) In normal mice, SGR extract increased ITR. Naïve (n = 15). SGR 0.01 g (n = 15). SGR 0.1 g (n = 14). SGR 1 g (n = 15). (B) In this AA-induced case, the ITR was recovered by SGR extract. Naïve (n = 15). CTRL (n = 15). SGR 0.01 g (n = 15). SGR 0.1 g (n = 15). SGR 1 g (n = 14). Bars represent mean ± standard error. * p < 0.05. *** p < 0.001. **** p < 0.0001. CTRL: Control. SGR: Smilacis Glabrae Rhixoma. ITR: intestinal transit rate. AA: acetic acid. GMD: gastrointestinal motility disorder.
Plants 12 01509 g008
Table 1. Active compounds of Smilacis Glabrae Rhixoma.
Table 1. Active compounds of Smilacis Glabrae Rhixoma.
Molecule NameStructureMW *OB (%) *Caco-2 *DL *
(-)-taxifolinPlants 12 01509 i001304.2760.51−0.240.27
(2R,3R)-2-(3,5-dihydroxyphenyl)-3,5,7-trihydroxychroman-4-onePlants 12 01509 i002304.2763.17−0.340.27
2H-3,9a-Methano-1-benzoxepin-9-methanol, octahydro-2,2,5a-trimethyl-, (3R-(3alpha,5aalpha,9alpha,9aalpha))-Plants 12 01509 i003238.4198.381.010.14
4,7-Dihydroxy-5-methoxyl-6-methyl-8-formyl-flavanPlants 12 01509 i004314.3637.030.480.28
beta-sitosterolPlants 12 01509 i005414.7936.911.320.75
cis-DihydroquercetinPlants 12 01509 i006304.2766.44−0.340.27
DihydroresveratrolPlants 12 01509 i007230.2887.270.810.11
diosgeninPlants 12 01509 i008414.6980.880.820.81
EICPlants 12 01509 i009280.541.91.160.14
EnhydrinPlants 12 01509 i010464.5140.56−0.360.74
MethyllinolenatePlants 12 01509 i011292.5146.151.480.17
naringeninPlants 12 01509 i012272.2759.290.280.21
oleic acidPlants 12 01509 i013282.5233.131.170.14
quercetinPlants 12 01509 i014302.2546.430.050.28
sitosterolPlants 12 01509 i015414.7936.911.320.75
StigmasterolPlants 12 01509 i016412.7743.831.440.76
taxifolinPlants 12 01509 i017304.2757.84−0.230.27
* Molecular weight (MW), oral bioavailability (OB), Caco-2 permeability (Caco-2), and drug likeness (DL).
Table 2. Compounds and targets related to gastrointestinal disease.
Table 2. Compounds and targets related to gastrointestinal disease.
Molecule NameTarget NameDisease Name
(-)-epicatechinHeat shock protein HSP 90Gastrointestinal Stromal Tumors (GIST)
Interleukin-6* Gastrointestinal motility disorders
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
Transcription factor AP-1* Gastrointestinal motility disorders
Tumor necrosis factor* Gastrointestinal motility disorders
Crohns’s Disease, unspecified
(-)-taxifolinHeat shock protein HSP 90Gastrointestinal Stromal Tumors (GIST)
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
(2R,3R)-2-(3,5-dihydroxyphenyl)-3,5,7-trihydroxychroman-4-oneHeat shock protein HSP 90Gastrointestinal Stromal Tumors (GIST)
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
(2S,3R)-3,5,7-trihydroxy-2-(4-hydroxyphenyl)chroman-4-oneHeat shock protein HSP 90Gastrointestinal Stromal Tumors (GIST)
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
(L)-alpha-TerpineolHeat shock protein HSP 90Gastrointestinal Stromal Tumors (GIST)
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
Sodium channel protein type 5 subunit alpha* Gastrointestinal motility disorders
(R)-linaloolHeat shock protein HSP 90Gastrointestinal Stromal Tumors (GIST)
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
Sodium channel protein type 5 subunit alpha* Gastrointestinal motility disorders
3-O-caffeoylshikimic acidProstaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
4,7-Dihydroxy-5-methoxyl-6-methyl-8-formyl-flavanHeat shock protein HSP 90Gastrointestinal Stromal Tumors (GIST)
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz-Jeghers syndrome
Sodium channel protein type 5 subunit alpha* Gastrointestinal motility disorders
AromadedrinHeat shock protein HSP 90Gastrointestinal Stromal Tumors (GIST)
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
astilbinInterleukin-10* Gastrointestinal motility disorders
Tumor necrosis factor* Gastrointestinal motility disorders
Crohns’s Disease, unspecified
beta-sitosterolHeat shock protein HSP 90Gastrointestinal Stromal Tumors (GIST)
Mu-type opioid receptor* Gastrointestinal motility disorders
Diarrhea
Opioid-induced bowel dysfunction
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz-Jeghers syndrome
Sodium channel protein type 5 subunit alpha* Gastrointestinal motility disorders
Sodium-dependent serotonin transporter* Gastrointestinal motility disorders
Transcription factor AP-1* Gastrointestinal motility disorders
cis-DihydroquercetinHeat shock protein HSP 90Gastrointestinal Stromal Tumors (GIST)
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
delta-amorpheneProstaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
DihydroresveratrolHeat shock protein HSP 90Gastrointestinal Stromal Tumors (GIST)
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
diosgeninProstaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
Vascular endothelial growth factor AColorectal Neoplasms
EICProstaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
Transient receptor potential cation channel subfamily V member 1* Gastrointestinal motility disorders
engeletinHeat shock protein HSP 90Gastrointestinal Stromal Tumors (GIST)
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
FERHeat shock protein HSP 90Gastrointestinal Stromal Tumors (GIST)
Leukotriene A-4 hydrolaseEsophageal cancer
Nitric oxide synthase, endothelialColon cancer
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
HMFAcetylcholinesterase* Gastrointestinal motility disorders
Dipeptidyl peptidase IV* Gastrointestinal motility disorders
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
isoengelitinProstaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
IstidinaProstaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
L-Bornyl acetateProstaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
MethyllinolenateProstaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
myristic acidProstaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
naringeninGlutathione S-transferase PColorectal Neoplasms
Gastrointestinal Neoplasms
Rectal Neoplasms
Heat shock protein HSP 90Gastrointestinal Stromal Tumors (GIST)
Peroxisome proliferator-activated receptor gammaCrohns’s Disease, unspecified
Inflammatory Bowel Disease
Ulcerative colitis
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
n-butyl-β-D-fructopyronosideNitric oxide synthase, brain* Gastrointestinal motility disorders
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
n-butyl-β-D-fructoufranosideProstaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
NCAProstaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
nonaneTumor necrosis factor* Gastrointestinal motility disorders
Crohns’s Disease, unspecified
oleic acidCholecystokinin* Gastrointestinal motility disorders
Glucagon* Gastrointestinal motility disorders
Insulin* Gastrointestinal motility disorders
Myeloperoxidase* Gastrointestinal motility disorders
Peptide YY* Gastrointestinal motility disorders
Peroxisome proliferator-activated receptor gammaCrohns’s Disease, unspecified
Inflammatory Bowel Disease
Ulcerative colitis
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
palmitic acidInterleukin-10* Gastrointestinal motility disorders
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
Tumor necrosis factor* Gastrointestinal motility disorders
Crohns’s Disease, unspecified
quercetinAcetylcholinesterase* Gastrointestinal motility disorders
Arachidonate 5-lipoxygenaseGastrointestinal cancers
Inflammatory Bowel Disease
Ulcerative colitis
Cytochrome P450 1A2Colorectal Neoplasms
Dipeptidyl peptidase IV* Gastrointestinal motility disorders
Epidermal growth factor receptorColorectal cancer
Glutathione S-transferase PColorectal Neoplasms
Gastrointestinal Neoplasms
Rectal Neoplasms
Heat shock protein HSP 90Gastrointestinal Stromal Tumors (GIST)
Interleukin-1 beta* Gastrointestinal motility disorders
Interleukin-10* Gastrointestinal motility disorders
Interleukin-6* Gastrointestinal motility disorders
Interleukin-8* Gastrointestinal motility disorders
Myeloperoxidase* Gastrointestinal motility disorders
Nitric oxide synthase, endothelialColon cancer
Ornithine decarboxylaseHereditary Polyposis Syndromes
Peroxisome proliferator-activated receptor gammaCrohns’s Disease, unspecified
Inflammatory Bowel Disease
Ulcerative colitis
Pro-epidermal growth factorColorectal Neoplasms
Rectal Neoplasms
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz-Jeghers syndrome
Proto-oncogene c-Fos* Gastrointestinal motility disorders
Sodium channel protein type 5 subunit alpha* Gastrointestinal motility disorders
ThrombomodulinRadiation enteropathy
Transcription factor AP-1* Gastrointestinal motility disorders
Tumor necrosis factor* Gastrointestinal motility disorders
Crohns’s Disease, unspecified
Vascular endothelial growth factor AColorectal Neoplasms
resveratrolHeat shock protein HSP 90Gastrointestinal Stromal Tumors (GIST)
Interleukin-1 beta* Gastrointestinal motility disorders
Interleukin-10* Gastrointestinal motility disorders
Interleukin-6* Gastrointestinal motility disorders
Interleukin-8* Gastrointestinal motility disorders
Mitogen-activated protein kinase 8Crohns’s Disease, unspecified
Myeloperoxidase* Gastrointestinal motility disorders
Nitric oxide synthase, endothelialColon cancer
Ornithine decarboxylaseHereditary Polyposis Syndromes
Peroxisome proliferator-activated receptor gammaCrohns’s Disease, unspecified
Inflammatory Bowel Disease
Ulcerative colitis
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
Proto-oncogene c-Fos* Gastrointestinal motility disorders
Transcription factor AP-1* Gastrointestinal motility disorders
Tumor necrosis factor* Gastrointestinal motility disorders
Crohns’s Disease, unspecified
Vascular endothelial growth factor AColorectal Neoplasms
Sitogluside5-hydroxytryptamine receptor 3A* Gastrointestinal motility disorders
Diarrhea
Irritable bowel syndrome
Postoperative nausea and vomiting
Heat shock protein HSP 90Gastrointestinal Stromal Tumors (GIST)
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
Sodium channel protein type 5 subunit alpha* Gastrointestinal motility disorders
SKMProstaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
stearic acidProstaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
StigmasterolLeukotriene A-4 hydrolaseEsophageal cancer
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
Sodium channel protein type 5 subunit alpha* Gastrointestinal motility disorders
succinic acidNitric oxide synthase, brain* Gastrointestinal motility disorders
taxifolinHeat shock protein HSP 90Gastrointestinal Stromal Tumors (GIST)
Prostaglandin G/H synthase 2Adenomatous polyposis
Colorectal cancer
Oropharyngeal squamous cell carcinoma
Peutz–Jeghers syndrome
* After investigating the relationship between Smilacis Glabrae Rhixoma and GI motility disorder using Cytoscape stringApp, genes related to GI motility disorder were added to this table.
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

Choi, N.-R.; Lee, K.; Seo, M.; Ko, S.-J.; Choi, W.-G.; Kim, S.-C.; Kim, J.; Park, J.-W.; Kim, B.-J. Network Pharmacological Analysis and Experimental Validation of the Effect of Smilacis Glabrae Rhixoma on Gastrointestinal Motility Disorder. Plants 2023, 12, 1509. https://doi.org/10.3390/plants12071509

AMA Style

Choi N-R, Lee K, Seo M, Ko S-J, Choi W-G, Kim S-C, Kim J, Park J-W, Kim B-J. Network Pharmacological Analysis and Experimental Validation of the Effect of Smilacis Glabrae Rhixoma on Gastrointestinal Motility Disorder. Plants. 2023; 12(7):1509. https://doi.org/10.3390/plants12071509

Chicago/Turabian Style

Choi, Na-Ri, Kangwook Lee, Mujin Seo, Seok-Jae Ko, Woo-Gyun Choi, Sang-Chan Kim, Jinsung Kim, Jae-Woo Park, and Byung-Joo Kim. 2023. "Network Pharmacological Analysis and Experimental Validation of the Effect of Smilacis Glabrae Rhixoma on Gastrointestinal Motility Disorder" Plants 12, no. 7: 1509. https://doi.org/10.3390/plants12071509

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