Next Article in Journal
Chemical Analysis and Quality Assessment of Honey Obtained from Different Sources
Previous Article in Journal
Anaerobic Digestion of the Residue (Combination of Wastewater and Solid Waste) from a New Olive-Oil Manufacturing Process Based on an Olive Cold-Pressing System: Kinetic Approach and Process Performance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Hypoglycemic Effect and Experimental Validation of Scutellariae Radix based on Network Pharmacology and Molecular Docking

1
School of Pharmacy, Xinxiang Medical University, Xinxiang 453002, China
2
Sanquan College of Xinxiang Medical University, Xinxiang 453002, China
3
Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, College of Pharmacy, Xinxiang Medical University, Xinxiang 453002, China
*
Author to whom correspondence should be addressed.
Processes 2022, 10(12), 2553; https://doi.org/10.3390/pr10122553
Submission received: 20 October 2022 / Revised: 18 November 2022 / Accepted: 23 November 2022 / Published: 1 December 2022
(This article belongs to the Section Pharmaceutical Processes)

Abstract

:
Scutellariae Radix (SR) is a well-known traditional herb that has good pharmacological effects against diabetes. However, the mechanism of SR against diabetes is not clear. In this study, the ingredient–target–pathway relationship and hypoglycemic effect of SR on diabetes were explored using network pharmacology, molecular docking and an animal experiment. The targets of SR and diabetes were mined. The selected targets were studied using Gene Ontology (GO) enrichment analysis and pathway enrichment analysis. The network of active components, targets and pathways was integrated to analyze the ingredient–target–pathway relationship. Then, the correspondence between the active components and targets was verified using molecular docking. Finally, an animal experiment was used to verify the hypoglycemic effect of SR. There were 52 components and 22 targets for the hypoglycemic effect of SR. We identified 18 biological processes, 9 cellular components, 15 molecular functions and 25 signaling pathways. Molecular docking results indicated that the targets of diabetes bound strongly to the main components. The animal experiments showed that SR could significantly decrease the blood glucose level of diabetic rats (p ≤ 0.05). This study explored the potential targets and signaling pathways of SR in diabetes, and the results may help to illustrate the hypoglycemic mechanism of SR.

Graphical Abstract

1. Introduction

Diabetes mellitus is a metabolic disorder caused by a decrease in insulin secretion and/or lower insulin activity [1]. The global report of the World Health Organization (WHO) shows that from 1980 to 2019, the number of diabetic patients has almost doubled to 422 million, and it is expected to increase to 693 million by 2045 [2]. Although chemotherapy and/or insulin injections could decrease the blood glucose level [3], there is no effective treatment for diabetes.
Nowadays, traditional Chinese medicine has attracted more and more attention due to its remarkable therapeutic effects and low side effects on diabetes [4,5]. As a perennial herb in Labiatae, Scutellariae Radix (SR), well known as Huang-Qin in Chinese, is derived from the dry roots of Scutellaria baicalensis Georgi. SR is a famous traditional herb in East Asia, especially in China. Recent experimental studies found that SR has good pharmacological effects against diabetes [6,7,8,9]. Zhao Lijuan et al. [6] and Xiao Suwei et al. [7] found that SR could ameliorate hyperglycemia by adjusting the gut microbiota and bile acid metabolism in diabetic rats. Sun Min Park et al. [8] revealed that the extracts of SR had an insulinotropic action in cell experiments of pancreatic islets. Fang Penghua et al. [9] studied the major active ingredients of SR (baicalin and its aglycone baicalein) on metabolic disorders and proposed a novel treatment strategy against hyperglycemia, insulin resistance and type 2 diabetes. However, the hypoglycemic components and mechanism of SR in diabetes are not clear.
Network pharmacology is widely used in drug research. It uses bioinformatics, molecular biology and database systems to study the drug–target–pathway–disease relationship [10]. Therefore, as a useful tool, it could help to further study the role of traditional Chinese medicine in the treatment of diseases [10,11]. Molecular docking is a method that is used to simulate the interaction between a ligand (active component) and a receptor (biological macromolecule or target). The binding ability between the active components and the target is studied through molecular docking technology [12]. Qiqiang Zhang et al. revealed the active components, targets and molecular mechanisms of the Gandhi capsule in treating diabetic nephropathy through network pharmacology and molecular docking technology [12]. L. Chen et al. studied the Yangxinshi tablet in the treatment of heart failure using a network pharmacology–molecular docking strategy [11].
In this study, the potential targets and signal pathways of SR in diabetes were identified and the potential underlying mechanisms were uncovered by constructing the ingredient–target–pathway network. The binding ability of ingredients and targets were analyzed via molecular docking. Furthermore, an animal experiment was used to confirm the hypoglycemic effect of SR.

2. Materials and Methods

2.1. Reagents

Streptozotocin (STZ, no. WXB8D5718V) was obtained from Sigma Aldrich Company Co. Ltd. (Saint Louis, MO, USA). Sodium citrate (no. GB/T 16493-1996), citric acid (no. GB/T 9855-2008) and ethanol (no. 64-17-5) were supplied by Tianjin Hengxing Chemical Reagent Manufacturing Co. Ltd. (Tianjin, China). Metformin hydrochloride tablets (no. H11021518) were purchased from Bei-Jing-Jing-Feng Pharmaceutical Co., Ltd. (Beijing, China). Normal saline (no. H13023201) was supplied by Shijiazhuang-Si-Yao Co., Ltd. (Shijiazhuang, China). The blood glucose meter was the product of Jiangsu Yuwell Medical Equipment Co., Ltd. (Jiangsu, China).
SR was obtained from Inner Mongolia Province, China. The voucher specimen was deposited at Xinxiang Medical University (Xinxiang, China). About 5.0 g of SR was first decocted with 100 mL water for 1.0 h, and then 50 mL water for 0.5 h. Then, the above extracts were mixed and concentrated to 48 mL.

2.2. Chemical Components and Targets

TCMSP (https://www.tcmsp-e.com/, accessed on 1 July 2020) is an open database that can provide information about the chemical composition, chemical structure and clinical application of traditional Chinese medicines. The chemical components of SR were obtained from the TCMSP database. The chemical structures were drawn with ChemOffice software (Version 8.0, CambridgeSoft Corporation, Cambridge, MA, USA) [13]. The Batman-tcm database (http://bionet.ncpsb.org.cn/batman-tcm/, accessed on 1 July 2020) is a good bioinformatics analysis tool for studying the molecular mechanisms of traditional Chinese medicine [14]. PharmMapper (http://www.lilab-ecust.cn/pharmmapper/, accessed on 1 July 2020) is a useful pharmacophore-matching database for target identification [15]. The targets of components were obtained from the TCMSP database, Batman-tcm database and Pharmmapper database.

2.3. Selection of Targets for SR in Diabetes

Drugbank (https://go.drugbank.com/, accessed on 1 July 2020) is an online database containing drug and target information. The related targets of diabetes were screened in the DrugBank database. The potential targets of SR were compared with the targets of diabetes to confirm the targets of SR in diabetes.

2.4. Network of Targets

The String database (https://cn.string-db.org/, accessed on 1 July 2020) can analyze protein interaction information and calculate the confidence scores of all protein interactions. A score, ranging from 0 to 1, indicates confidence in the interaction between targets, and medium confidence (score ≥ 0.4) was used in this study [16]. The targets of SR in diabetes were analyzed in the Homo sapiens species mode. At last, the network of interrelationships between targets was analyzed.

2.5. Gene Ontology (GO) and KEGG Pathway Enrichment Analysis

The Metascape database (https://metascape.org/gp/index.html, accessed on 1 July 2020) was used for the target annotation and analysis. First, the targets of SR in diabetes were input into the Metascape database. Then, the parameters were set as follows: Homo sapiens for the species option, 1 for the minimum overlap and 0.05 for the p-value. Finally, the GO analysis and KEGG pathway enrichment analysis were carried out.

2.6. Network Analysis

Cytoscape software was used to analyze molecular interactions and integrate targets and biological pathways. In this study, the active ingredients, potential targets and KEGG pathways were integrated to obtain the ingredient–target–pathway relationship network.

2.7. Animal Experiment

Sprague Dawley rats (60 adult males weighing 200 to 300 g) were supplied by the Laboratory Animal Centre at Xinxiang Medical University (Xinxiang, China). The rats were raised with sufficient feed and drinking water at 25 ± 2 °C and exposed to light for 12 h a day. After one week of adaptive feeding, the rats were randomly divided into a normal group, a model group, a positive group and an SR group, as shown in Table 1. Then, the rats were raised with different diets for one week. After 12 h starvation and enough water, the rats were administered STZ through an intraperitoneal injection. The STZ (1% m/v) was dissolved in a citric acid–sodium citrate buffer (pH 4.2–4.5). The normal group was treated with the same volume of buffer solution. If the fasting blood glucose of rats exceeded 16.6 mmol/L, it was judged that the diabetic model was successful. Then, 6 eligible rats were randomly selected from each group and administered each day via gavage for 4 weeks. The rats were monitored weekly for blood glucose, water intake and body weight. Four weeks later, the rats were anesthetized with isoflurane first, and then the necks were pressed down while its tail was sharply pulled to the rear. The pancreata of rats were collected and fixed with 4% paraformaldehyde for HE staining.
This study was approved by the Ethics Committee of Xinxiang Medical University (no. XXLL-2020055) and followed the internationally accepted principles for laboratory animal use and care, such as the European Community guidelines (EEC Directive of 1986; 86/609/EEC) and Guidelines for the Euthanasia of Animals (2020).

2.8. Molecular Docking Analysis

The chembio3D (ChemBio3D, Ultra 14.0) function of ChemOffice software was used to convert the chemical structure of the ingredient into a three-dimensional format. The three-dimensional protein structure of the target was obtained from the PDB database (http://www.rcsb.org/pdb/home/home.do, accessed on 1 July 2020). The target protein was routinely processed (hydrogenation, dehydration, charging, etc.) by using Autodock tools software (version 1.5.6) (La Jolla, CA, USA). Then, the structures of the protein and ingredient were converted into pdqpt format files. Autodock tools software was used to set the molecular docking parameters, and Autodock Vina software (version 1.1.2) was used for molecular docking. Finally, PyMOL software (version 1.7.2.1) (Schrödinger, Inc., New York, NY, USA) was used to analyze the docking results.

2.9. Statistical Analysis

In this study, all data were analyzed in the form of mean ± standard deviation using the IBM SPSS Statistics 26 software (Armonk, NY, USA). In the statistical comparison, p < 0.05 indicated there was statistical significance.

3. Results and Discussion

3.1. Hypoglycemic Ingredients and Targets

A total of 140 active ingredients with 408 related targets were obtained. According to the intersection analysis of the diabetic targets and the SR targets, 22 targets were obtained, as shown in Table 2. There were 52 SR components related to these 22 targets (as listed in Table S1), and 15 components (listed in Table 3) had more than two targets.

3.2. Relationship between Targets

The relationship between these 22 targets was obtained through the transformation using the String database. As shown in Figure 1, the long-chain acyl CoA synthase 4 (ACSL4), cytochrome P450 2B6 (CYP2B6), lipoprotein lipase (LPL), maltase-glucoamylase (MGAM), peroxisome proliferator-activated receptor D (PPARD), peroxisome proliferator-activated receptor G (PPARG) and prostaglandin G/H synthase 1 (PTGS1, cyclooxygenase 1, COX1) were closely connected.
Peroxisome proliferator-activated receptor (PPAR) belongs to the nuclear receptor superfamily of regulatory factors, and its function is mainly related to the regulation of genes on glucose and lipid metabolism [17]. Studies found that PPARA, PPARG and PPAR signal transduction pathways and the adipocytokine signal pathway were upregulated to prevent hyperglycemia, and PPARG receptors regulated lipid uptake and lipogenesis through gene transcriptional activation [18]. On the other hand, PPAR could mediate the induced transcription of the LPL gene [19]. LPL is an important part of the synthesis of triglycerides and it plays a key role in lipoprotein and cholesterol metabolism [20].
MGAM can hydrolyze linear α-1,4-linked oligosaccharide substrates and plays an essential role in the production of human intracavity glucose; therefore, it is an effective drug target for the treatment of diabetes and obesity [21,22].
The targets of methyl palmitelaidate, 1,1,6-trimethyl-1,2-dihydronaphthalene and palmitic acid were PTGS1, CYP2B6 and CYP2C8, respectively. These three targets were related to the cyclooxygenase pathway and arachidonic acid epoxygenase activity, and prostaglandin H synthase (PGHS) was a rate-limiting enzyme. Studies showed that the PGHS-2 inhibitors could prevent streptozotocin (STZ)-induced diabetes [23]. Studies found that the expression of prostaglandin synthase 2 (COX2) in diabetic patients was significantly higher than that in healthy controls, and the COX-2 signal may lead to high inflammatory levels and apoptosis in diabetes [24,25].

3.3. Molecular Docking Results

As shown in Table 2 and Figure 1, four targets (PTGS1, PPARG, MGAM and GAA) were selected for molecular docking with the main active components of SR. It is generally believed that the smaller the binding free energy (affinity, unit: kcal·mol−1) between a molecule and a protein, the more tightly and firmly the two would bind. A binding energy smaller than −5.0 kcal·mol−1 means the active component and protein have good binding activity, and smaller than −7.0 kcal·mol−1 means strong binding activity. The docking results were shown in Figure 2. As shown in Figure 2A, baicalin docked to MGAM through glutamine (GLN) and glycine (GLY) with −8.4 kcal·mol−1 of intermolecular binding energy. Figure 2B indicates the sitoside was bound to GAA targets via glutamate (GLU) and threonine (THR) with −7.8 kcal·mol−1. As shown in Figure 2C, baicalein was bound to the GAA target via aspartic acid (ASP) and GLY with −7.8 kcal·mol−1. Figure 2D shows 4-hydroxycinnamic acid docked to PPARG targets through serine (SER) with −6.2 kcal·mol−1. The molecular docking results illustrated that SR had good pharmacological effects against diabetes.

3.4. GO Analysis, KEGG Analysis and Ingredient–Target–Pathway Network

As shown in Figure 3A and Tables S2–S4, 15 molecular functions were identified (p ≤ 0.01), including monoamine transmembrane transporter activity, nuclear receptor activity, maltose alpha-glucosidase activity and arachidonic acid epoxygenase activity. As shown in Figure 3B and Table S3, nine cellular components were obtained (p ≤ 0.01), including RNA polymerase II transcription regulator complex, presynaptic membrane, ion channel complex and apical plasma membrane. As shown in Figure 3C and Table S2, 18 biological processes were identified (p ≤ 0.01), including a fatty acid metabolic process, maltose metabolic process and positive regulation of lipid localization. As shown in Figure 3D and Table S5, 25 KEGG pathways were obtained (p ≤ 0.05), including the PPAR signaling pathway, arachidonic acid metabolism, galactose metabolism, and starch and sucrose metabolism. Combining the results of the KEGG analysis, molecular docking and relationship between targets, the mechanism of SR in diabetes may be through regulating the metabolism of sugar and resisting the oxidation function.
The active ingredients, potential targets and KEGG pathways were integrated to establish the ingredient–target–pathway relationship diagram. As shown in Figure 4, there were 52 chemical components, 22 targets and 25 pathways for SR in diabetes. Through network pharmacology and molecular docking, it was found that baicalin, sitoside, baicalein and 4-hydroxycinnamic acid of SR could exert hypoglycemic effects through PPAR, MGAM, PTGS1 and so on.

3.5. Animal Experiment

3.5.1. Blood Glucose

As listed in Table 4, the blood glucose of rats, except the rats of the normal group, was more than 16.6 mmol/L; therefore, the diabetic rats were successfully induced. After 4 weeks of treatment, the blood glucose levels of the SR and positive control groups were significantly decreased compared with the rats in the model group (p < 0.05), and no significant difference was found between the SR and positive control groups (p > 0.05). Therefore, the results indicated that SR had a good hypoglycemic effect on diabetes.

3.5.2. Body Weight and Water Drunk

The weights and water drunk of the rats in each group were recorded every week. As shown in Table 4, only the rats of the normal group gained weight, while all the rats of the other three groups lost weight. However, the weight loss level of the SR group and positive control group was significantly smaller than the model group (p < 0.05). The water intake of the diabetic rats was significantly higher than the normal group (p < 0.05). Meanwhile, the amount of water drunk by the SR group was lower than that of the model group (p < 0.05). Therefore, SR had some beneficial effects for diabetic rats in delaying weight loss and reducing water intake.

3.5.3. Effect of SR on the Pancreas in Diabetic Rats

After 4 weeks, the pancreatic tissue was taken out for HE staining. As shown in Figure 5, the islet of normal rats (Figure 5A) was quasi-circular and the cells were closely arranged with a clear nucleus. However, the islet of the diabetic group rats (Figure 5B) changed obviously with an irregular shape; at the same time, the number of islet cells decreased and some islet cells were even completely atrophied with biased nuclei. For the rats of the positive (Figure 5C) and SR groups (Figure 5D), the volume of the pancreas and the number of pancreatic cells were improved, and the cells were arranged neatly with a clear boundary.

4. Conclusions

In this study, 52 SR components, 22 potential targets and 25 KEGG pathways were identified using network pharmacology. The results of molecular docking increased the reliability of SR in the treatment of diabetes. Finally, an animal experiment showed that SR could significantly decrease the blood glucose level of diabetic rats (p ≤ 0.05). In a future study, the hypoglycemic ingredients of an SR extract will be separated and identified. In addition, the targets and signaling pathways will be studied through a cell test and an animal experiment. In summary, further study will be carried out to confirm the ingredient–target–pathway relationship of SR on diabetes. This study explored the potential targets and pathways of SR in diabetes, and the results may help to illustrate the hypoglycemic mechanisms of SR.

Supplementary Materials

The following supporting information can be downloaded from https://www.mdpi.com/article/10.3390/pr10122553/s1. Table S1: The active ingredients and targets for Scutellariae Radix in diabetes. Table S2: The biological process of the therapeutic targets of Scutellariae Radix. Table S3: Cellular component of the therapeutic targets of Scutellariae Radix. Table S4: Molecular function of the therapeutic targets of Scutellariae Radix. Table S5: KEGG analysis results of the therapeutic targets of Scutellariae Radix.

Author Contributions

Study design, J.X., X.L. and Q.C.; sample collection and identification, network pharmacology analysis and animal experiment, X.L., Q.C., X.X., M.L., J.X. and C.L.; writing—original draft preparation, review and editing, X.L. and J.X. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the scientific and technological projects of Henan Province, grant number 222102310566, and the National College Students Innovation and Entrepreneurship Training Program, grant number S201910472030.

Data Availability Statement

The data presented in this study are available in the Supplementary Materials.

Acknowledgments

The authors are extremely grateful for the support of the Cardiovascular Remodeling Intervention and Molecular Targeting Drug Research and Development Key Laboratory and the Vascular Remodelling Intervention and Molecular Targeted Therapy Drug Development Innovation Team.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Padhi, S.; Nayak, A.K.; Behera, A. Type II diabetes mellitus: A review on recent drug based therapeutics. Biomed. Pharmacother. 2020, 131, 110708. [Google Scholar] [CrossRef]
  2. Cho, N.H.; Shaw, J.E.; Karuranga, S.; Huang, Y.; da Rocha Fernandes, J.D.; Ohlrogge, A.W.; Malanda, B. IDF Diabetes Atlas: Global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes Res. Clin. Pract. 2018, 138, 271–281. [Google Scholar] [CrossRef] [PubMed]
  3. Artasensi, A.; Pedretti, A.; Vistoli, G.; Fumagalli, L. Type 2 Diabetes Mellitus: A Review of Multi-Target Drugs. Molecules 2020, 25, 1987. [Google Scholar] [CrossRef]
  4. Sun, R.; Deng, X.; Zhang, D.; Xie, F.; Wang, D.; Wang, J.; Tavallaie, M.S.; Jiang, F.; Fu, L. Anti-diabetic potential of Pueraria lobata root extract through promoting insulin signaling by PTP1B inhibition. Bioorg. Chem. 2019, 87, 12–15. [Google Scholar] [CrossRef] [PubMed]
  5. Gao, H.; Yang, Y.; Deng, J.; Liang, J.; Zhang, W.; Feng, X. A systematic review and meta-analysis on the efficacy and safety of traditional Chinese patent medicine Jinqi Jiangtang Tablet in the treatment of type 2 diabetes. Complement. Ther. Med. 2019, 47, 102021. [Google Scholar] [CrossRef]
  6. Zhao, L.; Ma, P.; Peng, Y.; Wang, M.; Peng, C.; Zhang, Y.; Li, X. Amelioration of hyperglycaemia and hyperlipidaemia by adjusting the interplay between gut microbiota and bile acid metabolism: Radix Scutellariae as a case. Phytomedicine 2021, 83, 153477. [Google Scholar] [CrossRef] [PubMed]
  7. Xiao, S.; Liu, C.; Chen, M.; Zou, J.; Zhang, Z.; Cui, X.; Jiang, S.; Shang, E.; Qian, D.; Duan, J. Scutellariae radix and coptidis rhizoma ameliorate glycolipid metabolism of type 2 diabetic rats by modulating gut microbiota and its metabolites. Appl. Microbiol. Biotechnol. 2020, 104, 303–317. [Google Scholar] [CrossRef]
  8. Park, S.M.; Hong, S.M.; Sung, S.R.; Lee, J.E.; Kwon, D.Y. Extracts of Rehmanniae radix, Ginseng radix and Scutellariae radix improve glucose-stimulated insulin secretion and beta-cell proliferation through IRS2 induction. Genes. Nutr. 2008, 2, 347–351. [Google Scholar] [CrossRef] [Green Version]
  9. Fang, P.; Yu, M.; Shi, M.; Bo, P.; Gu, X.; Zhang, Z. Baicalin and its aglycone: A novel approach for treatment of metabolic disorders. Pharmaco. Rep. 2020, 72, 13–23. [Google Scholar] [CrossRef] [PubMed]
  10. Guo, X.; Ji, J.; Feng, Z.; Hou, X.; Luo, Y.; Mei, Z. A network pharmacology approach to explore the potential targets underlying the effect of sinomenine on rheumatoid arthritis. Int. Immunopharmacol. 2020, 80, 106201. [Google Scholar] [CrossRef] [PubMed]
  11. Chen, L.; Cao, Y.; Zhang, H.; Lv, D.; Zhao, Y.; Liu, Y.; Ye, G.; Chai, Y. Network pharmacology-based strategy for predicting active ingredients and potential targets of Yangxinshi tablet for treating heart failure. J. Ethnopharmacol. 2018, 219, 359–368. [Google Scholar] [CrossRef] [PubMed]
  12. Zhang, Q.; Ye, Q.; Huang, X.; Xu, A.; Liu, Y.; Qi, J.; Zhang, H.; Zhang, J. Revealing active components, action targets and molecular mechanism of Gandi capsule for treating diabetic nephropathy based on network pharmacology strategy. BMC Complement. Med. Ther. 2020, 20, 362. [Google Scholar] [CrossRef]
  13. Jinlong, R.; Peng, L.; Jinan, W.; Wei, Z.; Li, B.; Chao, H.; Pidong, L.; Zihu, G.; Weiyang, T.; Yinfeng, Y.; et al. TCMSP: A database of systems pharmacology for drug discovery from herbalmedicines. J. Cheminform. 2014, 6, 13. [Google Scholar] [CrossRef] [Green Version]
  14. Liu, Z.; Guo, F.; Wang, Y.; Li, C.; Zhang, X.; Li, H.; Diao, L.; Gu, J.; Wang, W.; Li, D.; et al. BATMAN-TCM: A Bioinformatics Analysis Tool for Molecular Mechanism of Traditional Chinese Medicine. Sci. Rep. 2016, 6, 21146. [Google Scholar] [CrossRef] [Green Version]
  15. Wang, X.; Shen, Y.; Wang, S.; Li, S.; Zhang, W.; Liu, X.; Lai, L.; Pei, J.; Li, H. PharmMapper 2017 update: A web server for potential drug target identification with a comprehensive target pharmacophore database. Nucleic Acids Res. 2017, 45, W356–W360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Szklarczyk, D.; Morris, J.H.; Cook, H.; Kuhn, M.; Wyder, S.; Simonovic, M.; Santos, A.; Doncheva, N.T.; Roth, A.; Bork, P.; et al. The STRING database in 2017: Quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 2017, 45, D362–D368. [Google Scholar] [CrossRef]
  17. Ortuño Sahagún, D.; Márquez-Aguirre, A.L.; Quintero-Fabián, S.; López-Roa, R.I.; Rojas-Mayorquín, A.E. Modulation of PPAR-γ by Nutraceutics as Complementary Treatment for Obesity-Related Disorders and Inflammatory Diseases. PPAR Res. 2012, 2012, 318613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Zhou, J.; Zhou, S. Berberine regulates peroxisome proliferator-activated receptors and positive transcription elongation factor b expression in diabetic adipocytes. Eur. J. Pharmacol. 2010, 649, 390–397. [Google Scholar] [CrossRef] [PubMed]
  19. Auwerx, J.; Schoonjans, K.; Fruchart, J.C.; Staels, B. Transcriptional control of triglyceride metabolism: Fibrates and fatty acids change the expression of the LPL and apo C-III genes by activating the nuclear receptor PPAR. Atherosclerosis 1996, 124, S29–S37. [Google Scholar] [CrossRef] [PubMed]
  20. Auwerx, J.; Leroy, P.; Schoonjans, K. Lipoprotein lipase: Recent contributions from molecular biology. Crit. Rev. Clin. Lab. Sci. 1992, 29, 243–268. [Google Scholar] [CrossRef] [PubMed]
  21. Ren, L.; Qin, X.; Cao, X.; Wang, L.; Bai, F.; Bai, G.; Shen, Y. Structural insight into substrate specificity of human intestinal maltase-glucoamylase. Protein. Cell. 2011, 2, 827–836. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Conte, F.; van Buuringen, N.; Voermans, N.C.; Lefeber, D.J. Galactose in human metabolism, glycosylation and congenital metabolic diseases: Time for a closer look. Biochim. Biophys. Acta Gen. Subj. 2021, 1865, 129898. [Google Scholar] [CrossRef]
  23. Tabatabaie, T.; Waldon, A.M.; Jacob, J.M.; Floyd, R.A.; Kotake, Y. COX-2 inhibition prevents insulin-dependent diabetes in low-dose streptozotocin-treated mice. Biochem. Biophys. Res. Commun. 2000, 273, 699–704. [Google Scholar] [CrossRef] [PubMed]
  24. Litherland, S.A.; She, J.X.; Schatz, D.; Fuller, K.; Hutson, A.D.; Peng, R.H.; Li, Y.; Grebe, K.M.; Whittaker, D.S.; Bahjat, K.; et al. Aberrant monocyte prostaglandin synthase 2 (PGS2) expression in type 1 diabetes before and after disease onset. Pediatr. Diabetes 2003, 4, 10–18. [Google Scholar] [CrossRef] [PubMed]
  25. Siewiera, K.; Kassassir, H.; Talar, M.; Wieteska, L.; Watala, C. Long-term untreated streptozotocin-diabetes leads to increased expression and elevated activity of prostaglandin H2 synthase in blood platelets. Platelets 2016, 27, 203–211. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The interaction network relationships between targets (each node represents a target and each edge represents the proteins that jointly contribute to a shared function).
Figure 1. The interaction network relationships between targets (each node represents a target and each edge represents the proteins that jointly contribute to a shared function).
Processes 10 02553 g001
Figure 2. Molecular docking results of baicalin with GMAM (A), sitoside with GAA (B), baicalein with GAA (C) and 4-hydroxycinnamic acid with PPARG (D). The structures of proteins are shown using a cartoon model, while the active component is shown using a stick model and amino acid residues are shown using a line model (the number shows the site in the protein). The hydrogen bonds between the active component and amino acid residue are represented in yellow dashed lines, with the number showing the distance (Å).
Figure 2. Molecular docking results of baicalin with GMAM (A), sitoside with GAA (B), baicalein with GAA (C) and 4-hydroxycinnamic acid with PPARG (D). The structures of proteins are shown using a cartoon model, while the active component is shown using a stick model and amino acid residues are shown using a line model (the number shows the site in the protein). The hydrogen bonds between the active component and amino acid residue are represented in yellow dashed lines, with the number showing the distance (Å).
Processes 10 02553 g002
Figure 3. Molecular function pathway (A), cellular component pathway (B), biological process pathway (C) and KEGG pathway (D) analysis of the therapeutic targets of Scutellariae Radix in treating diabetes.
Figure 3. Molecular function pathway (A), cellular component pathway (B), biological process pathway (C) and KEGG pathway (D) analysis of the therapeutic targets of Scutellariae Radix in treating diabetes.
Processes 10 02553 g003
Figure 4. The interaction network of ingredient, target and pathway. The nodes represent the ingredients (orange arrows), the targets (green octagons) and the pathways (purple diamonds), and the edges represent the interactive relationships between two nodes.
Figure 4. The interaction network of ingredient, target and pathway. The nodes represent the ingredients (orange arrows), the targets (green octagons) and the pathways (purple diamonds), and the edges represent the interactive relationships between two nodes.
Processes 10 02553 g004
Figure 5. Histological analysis of pancreata from the normal group (A), diabetic group (B), positive group (C) and Scutellariae Radix group (D) after four weeks of treatment.
Figure 5. Histological analysis of pancreata from the normal group (A), diabetic group (B), positive group (C) and Scutellariae Radix group (D) after four weeks of treatment.
Processes 10 02553 g005
Table 1. The protocol of the animal experiment.
Table 1. The protocol of the animal experiment.
Normal GroupModel GroupPositive GroupScutellariae Radix Group
N6666
Diet before modelingStandard dietHigh-fat and high-sugar dietHigh-fat and high-sugar dietHigh-fat and high-sugar diet
Streptozotocin/50 mg/kg50 mg/kg50 mg/kg
Dosage (g/kg)Normal salineNormal salineMetformin, 0.16 g/kgScutellariae Radix extract, 1.05 g/kg *
* The dosage “1.05 g/kg” was for raw Scutellariae Radix. Before administration, raw Scutellariae Radix was processed according to the method in Section 2.1: Reagents.
Table 2. The potential hypoglycemic targets of Scutellariae Radix.
Table 2. The potential hypoglycemic targets of Scutellariae Radix.
Target GeneTargetsUniprot IDNumber of Ingredients
HDAC2Histone deacetylase 2Q9276924
PTGS1Prostaglandin G/H synthase 1P2321917
PPARGPeroxisome proliferator-activated receptor gammaP3723112
PPARDPeroxisome proliferator-activated receptor deltaQ0318110
PRKAB15′-AMP-activated protein kinase subunit beta-1Q9Y4789
ACSL4Long-chain-fatty-acid–CoA ligase 4O604885
GAALysosomal alpha-glucosidaseP102533
MGAMMaltase-glucoamylase, intestinalO434513
CFTRCystic fibrosis transmembrane conductance regulatorP135693
SLC6A3Sodium-dependent dopamine transporterQ019592
AGTR1Type-1 angiotensin II receptorP305561
CYP2B6Cytochrome P450 2B6P208131
CYP2C8Cytochrome P450 2C8P106321
KCNJ1ATP-sensitive inward rectifier potassium channel 11Q146541
KCNJ8ATP-sensitive inward rectifier potassium channel 8Q158421
LPLLipoprotein lipaseP068581
RXRARetinoic acid receptor RXR-alphaP197931
RXRBRetinoic acid receptor RXR-betaP287021
RXRGRetinoic acid receptor RXR-gammaP484431
SLC22A1Solute carrier family 22 member 1O152451
SLC6A2Sodium-dependent noradrenaline transporterP239751
SLC6A4Sodium-dependent serotonin transporterP316451
Table 3. The chemical information of active ingredients for Scutellariae Radix.
Table 3. The chemical information of active ingredients for Scutellariae Radix.
No.IngredientMolecular FormulaCAS No.Number of Targets
1EICC18H32O260-33-37
21,1,6-trimethyl-2H-naphthaleneC13H1630364-38-65
3BaicalinC21H18O1121967-41-94
4BaicaleinC15H10O5491-67-84
5Methyl linolelaidateC19H34O22566-97-44
6Methyl palmitelaidateC17H32O23913-63-14
7Methyl (Z)-cinnamateC10H10O2/4
8Methyl icos-11-enoateC21H40O22390-09-24
9TyrosolC8H10O2501-94-03
10Palmitic acidC16H32O257-10-32
11SitosideC35H60O6474-58-82
125-(2-hydroxyethyl)-2-methoxyphenolC9H12O350602-41-02
13P-coumaric acidC9H8O3501-98-42
14HyacinthinC8H8O122-78-12
15WLNC7H6O100-52-72
Table 4. The levels of blood glucose, body weight and drinking water before and after modeling (mean ± SD).
Table 4. The levels of blood glucose, body weight and drinking water before and after modeling (mean ± SD).
GroupNormal GroupModel GroupPositive GroupScutellariae Radix Group
N6666
Blood glucose
(mmol/L)
Before modeling5.2 ± 0.34.7 ± 0.9 5.1 ± 0.55.1 ± 0.4
After modeling4.9 ± 1.023.1 ± 2.4 #20.6 ± 3.0 #22.7 ± 4.4 #
After 4 weeks of
treatment
4.7 ± 0.9 *23.4 ± 6.26.2 ± 0.8 *6.2 ± 0.7 *
Body weight change (g)After 4 weeks of
treatment
37.5 ± 7.7 * ↑66.3 ± 15.9 ↓54.5 ± 12.5 *↓45.3 ± 8.8 * ↓
Drinking water (mL)Before modeling19.9 ± 2.020.1 ± 2.020.4 ± 1.419.9 ± 1.3
After 4 weeks of
treatment
54.7 ± 3.1 *195.5 ± 30.2198.5 ± 14.0144.3 ± 24.3 *
For the blood glucose levels before and after modeling, a significant difference between them is shown as # (p < 0.05); after four weeks of treatment, a significant difference compared with the model group is marked as * (p < 0.05); ↑: weight gain; ↓: weight loss.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Liu, X.; Li, C.; Chen, Q.; Xiao, X.; Li, M.; Xue, J. Hypoglycemic Effect and Experimental Validation of Scutellariae Radix based on Network Pharmacology and Molecular Docking. Processes 2022, 10, 2553. https://doi.org/10.3390/pr10122553

AMA Style

Liu X, Li C, Chen Q, Xiao X, Li M, Xue J. Hypoglycemic Effect and Experimental Validation of Scutellariae Radix based on Network Pharmacology and Molecular Docking. Processes. 2022; 10(12):2553. https://doi.org/10.3390/pr10122553

Chicago/Turabian Style

Liu, Xiaolong, Chunyan Li, Qijian Chen, Xian Xiao, Manman Li, and Jintao Xue. 2022. "Hypoglycemic Effect and Experimental Validation of Scutellariae Radix based on Network Pharmacology and Molecular Docking" Processes 10, no. 12: 2553. https://doi.org/10.3390/pr10122553

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