Skip to main content
ACS Omega logoLink to ACS Omega
. 2021 Jan 11;6(3):2354–2366. doi: 10.1021/acsomega.0c05680

Exploring the Active Components of Simotang Oral Liquid and Their Potential Mechanism of Action on Gastrointestinal Disorders by Integrating Ultrahigh-Pressure Liquid Chromatography Coupled with Linear Ion Trap-Orbitrap Analysis and Network Pharmacology

Zhiqiang Luo , Guohua Yu , Xing Han , Yang Liu §,*, Guopeng Wang , Xueyan Li §, Haiyang Yang §, Wenyan Sun §,*
PMCID: PMC7841926  PMID: 33521474

Abstract

graphic file with name ao0c05680_0006.jpg

Simotang oral liquid (SMT), a well-known traditional Chinese medicine formula composed of four medicinal and edible plants, has been extensively used for treating gastrointestinal disorders (GIDs) since ancient times. However, the major active constituents and the underlying molecular mechanism of SMT on GIDs are still partially understood. Herein, the preliminary chemical profile of SMT was first identified by ultrahigh-pressure liquid chromatography coupled with linear ion trap-Orbitrap tandem mass spectrometry (UHPLC-LTQ-Orbitrap). In total, 70 components were identified. Then, a network pharmacology approach integrating target prediction, pathway enrichment analysis, and network construction was adopted to explore the therapeutic mechanism of SMT. As a result, 170 main targets were screened out and considered as effective players in ameliorating GIDs. More importantly, the major hubs were found to be highly enriched in a calcium signaling pathway. Furthermore, 26 core SMT-related genes were identified, which may play key roles in ameliorating gastrointestinal motility. In conclusion, this work would provide valuable information for further development and clinical application of SMT.

1. Introduction

Gastrointestinal disorders (GIDs) are those that affect any part of the gastrointestinal tract, which directly impact on the intake, digestion, and absorption of nutrients.1 GIDs mainly include functional gastrointestinal disorder, irritable bowel syndrome, inflammatory bowel disease, colorectal cancer, and malabsorptive syndromes. Among them, functional gastrointestinal disorders (FGIDs) are one of the most common health-related conditions in gastroenterology practice.2 GIDs are highly prevalent and account for considerable health care utilization and spending in the world.3 It has been reported that the total expenditures for GIDs were about $135.9 billion annually in the United States.4 The pathogenesis of GIDs is quite complicated, with the current morbidity rate more than 41% worldwide.5 GIDs could be induced by several factors including infection, genetic factor, smoking, and eating misbehavior.6 Although many different types of drugs, such as dopamine receptor antagonists (metoclopramide and domperidone), 5-hydroxytryptamine receptor agonist (cisapride and mosapride), and motilin receptor agonist (erythromycin), have been used to alleviate some major symptoms of GIDs, patient outcomes still remain unsatisfying due to the unavoidable adverse effects after long-term use of these western medicines.7 Therefore, novel and safety therapeutic strategies for GIDs are urgently required.

As an important part of the complementary and alternative medical system, TCM is universally acknowledged by its adoption of “multi-components” to take “multi-effects” on “multi-targets”.8,9 It has been widely used in treating GIDs for thousands of years, especially the classical prescription called SMT. SMT was first recorded in YAN’s Jishengfang and approved by the Chinese Food and Drug Administration in the1980s.10 It contains four Chinese herbs including Citrus aurantium L. (CL), Aucklandia costus Falc (AF), Areca catechu L (AL), and Lindera aggregata (Sims) Kosterm.(LK) at the weight ratio of 3:2:3:3. Modern pharmacological and clinical studies have revealed that SMT exerts significant therapeutic effects on functional dyspepsia,11 the contraction of antral circular smooth muscle,12 and constipation-predominant irritable bowel syndrome.13 However, the underlying complex mechanisms of SMT acting on GIDs remain unclear.

With the rapid development of bioinformatics, network pharmacology has emerged as a systemic and promising tool for the mechanistic study of TCM.14,15 It shifts the “one target, one drug” paradigm to the “network target, multi-component” strategy, which is in line with the holistic view of the TCM theory.16 Until now, the network pharmacology approach has been successfully used to uncover the synergistic mechanisms of multiple components in TCM for treating specific diseases, such as the therapeutic mechanism of Carthamus tinctorius L. in the treatment of cardiovascular disease,17 the action mechanism of Pulsatilla decoction (PD) on acute ulcerative colitis,18 and so on.

In this work, an integrated strategy based on phytochemical analysis and network pharmacology was used to illuminate the complicated mechanism of action of SMT on GIDs. The flowchart of the experimental procedures of the current study is shown in Figure 1.

Figure 1.

Figure 1

Whole framework of this study based on network pharmacology for deciphering pharmacological mechanisms of SMT acting on GIDs.

2. Results and Discussion

2.1. Characterization and Identification of Chemical Constituents Contained in SMT

By using the established UHPLC-LTQ-Orbitrap method, the chemical constituents of SMT in both positive and negative modes were well separated and detected (see Figure S1). In this study, for the compounds with available standards, the compounds were identified by comparing the retention time and high-resolution accurate mass with the reference compounds. Moreover, the MS fragmentation behaviors of the reference substances were summarized, which were really helpful for structural elucidation of their derivatives with the same basic skeleton. For those unavailable standard compounds, the structures were tentatively identified by comparing with previous reports, according to accurate mass, chromatographic behavior, MS/MS data, and fragmentation rules.19 The mass errors for all the precursor ions of all identified constituents were within ±5 ppm. Overall, 70 components, including 26 flavonoids, 14 alkaloids, 4 coumarins, 5 amino acids, 6 organic acids, 13 terpenoids, and 2 other compounds in SMT were unambiguously or tentatively identified. Detailed information on these compounds is summarized in Tables 1 and 2 and Table S1.

Table 1. Identification of Compounds in SMT by UHPLC-LTQ-Orbitrap in Positive Ion Mode.

peak no. tR (min) molecular formula calculated mass (m/z) measured mass [M + H]+ error (ppm) MS/MS fragments identification compound source
1 1.27 C4H8N2O3 132.0529 133.0608 0.071 116.0343[M + H–NH3]+, 87.0553[M + H–CO–H2O]+, 70.0286[M + H–CO–H2O–NH3]+ asparagine AF
2 1.41 C5H9NO2 115.0627 116.0704 –0.185 70.0650[M + H–H2O–CO]+ proline AL
3 1.62 C6H9NO2 127.0627 128.0704 –0.175 110.0601[M + H–H2O]+, 109.0284[M + H–NH5]+, 99.0440[M + H–CH3N]+, 82.0650[M + H–H2O–CO]+, 81.0333[M + H–CH5NO] guvacine AL
4 1.69 C7H11NO2 141.0784 142.0862 –0.085 124.0758[M + H–H2O]+, 96.0807[M + H–H2O–CO]+, 81.0333[M + H–CH3OH–CH3N]+ arecaidine AL
5 1.87 C5H11NO2 117.0784 118.0861 –0.145 72.0807[M + H–H2O–CO]+ valine AL
6 2.09 C6H6O3 126.0311 127.0389 –0.101 109.0284[M + H–H2O]+, 99.0440[M + H–CO]+, 81.0333[M + H–CO–H2O]+ maltol AL
7 2.35 C9H13NO2 167.0940 168.1012 –4.314 150.0916[M + H–H2O]+ synephrine CL
8 2.85 C9H11NO3 181.0733 182.0811 –0.040 165.0551[M + H–NH3]+, 136.0762[M + H–H2O–CO]+ tyrosine AL
9 3.16 C7H11NO2 141.0784 142.0862 –0.025 110.0602[M + H–CH4O]+, 113.0598[M + H–CH3N]+, 96.0807[M + H–CH2O2]+, 81.0334[M + H–C2H7NO]+ guvacoline AL
10 3.47 C8H13NO2 155.0940 156.1018 –0.085 138.0553[M + H–CO]+, 124.0758[M + H–CH3OH]+, 113.0598[M + H–CH3–CH2N]+, 81.0334[M + H–CH3–CH3OH–CH2N]+ arecoline AL
11 4.87 C6H6O3 126.0311 127.0389 –0.031 109.0286[M + H–H2O]+, 81.0335[M + H–H2O–CO]+ triacetate lactone AL
12 4.93 C9H13NO 151.0991 152.1069 –0.071 121.0651[M + H–CH5N]+, 93.0698[M + H–CH5N–CO]+ N-methyltyramine CL
13 6.32 C9H11NO2 165.0784 166.0862 –0.105 149.0599[M + H–NH3]+, 131.0493[M + H–NH3–H2O]+, 120.0810[M + H–H2O–CO]+, 80.0493[M + H–C4H6O2]+ phenylalanine AL
17 11.09 C9H6O4 178.0260 179.0330 –4.721 161.0234[M + H–H2O]+, 151.0390[M + H–CO]+, 147.0442[M + H–O2]+, 137.0235[M + H–C2H2O]+, 123.0441[M + H–2CO]+, 111.0441[M + H–C3O2]+, 95.0487[M + H–C3O3]+ 5,7-dihydroxycoumarin CL
18 13.08 C27H30O15 594.1579 595.1655 –0.277 577.1582[M + H–H2O]+, 559.1478[M + H–2H2O]+, 529.1372[M + H–2H2O–HCHO]+, 511.1264[M + H–3H2O–HCHO]+ lonicerin CL
19 13.12 C18H19NO4 313.1308 314.1387 0.045 300.1193[M + H-CH2]+, 299.1159[M + H–CH3]+, 285.1006[M + H–C2H5]+, 268.1340[M + H–CO–H2O]+, 151.0758[M + H–C9H9NO2]+ laurolitsine LK
20 13.62 C21H30O8 410.1935 411.1987 –2.554 393.1896[M + H–H2O]+, 321.1677[M + H–C3H6O3]+, 291.1573[M + H–4CH2O]+, 249.1465[M + H–C6H10O5]+, 231.1359[M + H–C7H16O5]+, 203.0531[M + H–C13H20O2]+, 185.0424[M + H–C13H22O3]+ picriside B AF
21 13.77 C20H29NO5 363.2040 364.2118 –0.089 318.2074[M + H–CO–H2O]+, 128.0710[M + H–C14H20O3]+ saussureanine D AF
22 13.91 C9H6O3 162.0311 163.0389 –0.081 145.0286[M + H–H2O]+, 131.0494[M + H–O2]+, 119.0494[M + H–CO2]+, 107.0493[M + H–2CO]+, 91.0542[M + H–C2O3] 7-hydroxycoumarin CL
23 14.22 C18H19NO4 313.1308 314.1387 0.045 297.1133[M + H–NH3]+, 265.0870[M + H–CH3OH–NH3]+ norisoboldine LK
25 14.69 C19H21NO4 327.1465 328.1543 –0.005 313.1315[M + H–CH3]+, 297.1133[M + H–CH5N]+, 285.1134[M + H–COCH3]+, 265.0872[M + H–C2H7N–H2O]+, 253.0872[M + H–CH2O–C2H7N]+ boldine LK
27 14.99 C19H21NO3 311.1515 312.1595 0.030 269.1181[M + H–C2H5N]+, 283.1336[M + H–CH3N]+, 206.1181[M + H–C7H6O]+ pronuciferine LK
28 15.07 C15H16O4 260.1043 261.1120 –0.166 243.1013[M + H–H2O]+, 217.1203[M + H–CO2]+, 201.0914[M + H–2CH2O]+, 189.0551[M + H–C4H8O]+, 127.0004[M + H–C10H14]+ isomerancin CL
30 15.35 C15H12O5 272.0679 273.0755 –0.250 153.0186[M + H–C8H8O]+, 147.0446[M + H–H2O–C6H4O2]+, 179.0343[M + H–C6H6O]+ naringenin CL
32 15.39 C6H8O2 112.0518 113.0595 –0.186 95.0492[M + H–H2O]+, 85.0648[M + H–CO]+, 67.0541[M + H–CO–H2O]+ sorbic acid CL
34 15.43 C19H23NO4 329.1621 330.1697 –0.305 192.1024[M + H–C8H10O2]+, 299.1286[M + H–CH5N]+ reticuline LK
35 15.57 C15H12O5 272.0679 273.0756 –0.190 153.0186[M + H–C8H8O]+, 147.0445[M + H–H2O–C6H4O2]+, 179.0343[M + H–C6H6O]+ garbanzol CL
36 15.60 C21H22O9 418.1258 419.1330 –0.689 404.1112[M + H–CH3]+, 389.0879[M + H–2CH3]+ natsudaidain CL
38 15.81 C19H21NO4 327.1465 328.1543 –0.005 297.1130[M + H–CH3N]+, 265.0869[M + H–C2H9NO]+ laurotetanine LK
40 15.93 C16H14O6 302.0784 303.0849 –4.569 179.0343[M + H–C7H8O2]+, 177.0553[M + H–C6H4O2–H2O]+, 153.0187[M + H–C9H10O2]+, 285.0767[M + H–H2O]+ hesperetin CL
41 15.93 C22H24O10 448.1363 449.1442 –0.513 431.1353[M + H–H2O]+, 413.1249[M + H–2H2O]+, 369.0984[M + H–C2H8O3]+, 345.0984[M + H–C4H8O3]+, 303.0876[M + H–C6H10O4]+, 263.0561[M + H–C9H14O4]+ isosakuranin CL
43 16.11 C17H16O4 284.1043 285.1097 –2.426 267.1000[M + H–H2O]+, 257.0815[M + H-C2H4]+, 241.1205[M + H–CO2]+, 211.0735[M + H–CH2O2]+, 201.0551[M + H–C5H8O]+ ethyllucidone LK
44 16.38 C19H21NO4 327.1465 328.1540 –0.315 297.1127[M + H–CH3N]+, 265.0866[M + H–C2H9NO]+, 311.1284[M + H–NH3]+ morphine acetate LK
45 16.49 C15H16O4 260.1043 261.1120 –0.136 243.1024[M + H-H2O]+, 189.0553[M + H–CH3–C3H5O]+, 177.0553[M + H–C5H8O]+, 217.0865[M + H–CO2]+ linderane LK
46 16.52 C17H16O5 300.0992 301.1044 –2.650 283.0949[M + H–H2O]+, 243.1011[M + H–C2H2O2]+ pashanone LK
47 17.13 C19H21NO4 327.1465 328.1542 –0.125 311.1290[M + H–NH3]+ naloxone LK
49 17.32 C16H14O5 286.0835 287.0914 –0.030 245.0813[M + H–C2H2O]+, 179.0343[M + H–C7H8O]+, 161.0601[M + H–C6H6O3]+, 153.0186[M + H–C9H10O]+ isosakuranetin CL
50 17.63 C19H18O6 342.1097 343.1154 –2.225 325.1050[M + H–H2O]+, 311.1284[M + H-O2]+, 283.0947[M + H–CO–CH3OH]+ 4′,5,7,8-tetramethoxyflavone CL
51 17.80 C10H16O 152.1195 153.1273 –0.072 135.1172[M + H–H2O]+, 109.1013[M + H–C2H4O]+, 107.0857[M + H–C2H6O]+, 95.0856[M + H–C3H6O]+, 93.0700[M + H–C3H8O]+, 81.0698[M + H–C4H8O]+ 2-methyl-5-propan-5-ylcyclohex-2-en-1-one AF
52 18.10 C20H27NO4 345.1934 346.2012 –0.085 328.3216[M + H–H2O]+, 300.1967[M + H–HCOOH]+, 128.0710[M + H–C14H18O2]+, 100.0758[M + H–C15H18O3]+ saussureanine B AF
53 18.46 C15H20O2 232.1457 233.1536 0.034 215.1435[M + H–H2O]+, 205.1591[M + H–CO]+, 197.1329[M + H–2H2O]+, 187.1486[M + H–H2O–CO]+ costunolide AF
54 18.77 C20H29NO4 347.2091 348.2170 0.045 330.2073[M + H–H2O]+,128.0710[M + H–C14H18O]+ saussureanine A AF
55 18.81 C20H20O7 372.1203 373.1282 –0.009 358.1057[M + H–CH3]+, 343.0824[M + H–2CH3]+ tangeretin CL
56 19.04 C20H20O8 388.1152 389.1232 –0.136 374.1009[M + H–CH3]+, 359.0774[M + H–2CH3]+, 328.3221[M + H–C2H5O2]+ demethylnobiletin CL
57 19.12 C15H18O3 246.1250 247.1329 0.029 229.1229[M + H–H2O]+, 211.1123[M + H–2H2O]+, 201.1279[M + H–H2O–CO]+, 183.1173[M + H–CO–2H2O]+, 173.1329[M + H–H2O–2CO]+, 135.0808[M + H–C6H8O2]+, 107.0857[M + H–C7H7O3]+ isozaluzanin C AF
58 19.37 C15H20O2 232.1457 233.1536 0.004 215.1436[M + H–H2O]+, 205.1593[M + H–CO]+, 197.1130[M + H–2H2O]+, 187.1487[M + H–H2O–CO]+ isoalantolactone AF
59 19.53 C20H20O7 372.1203 373.1268 –3.617 358.1057[M + H–CH3]+, 343.0824[M + H–2CH3], 329.1031[M + H–C2H5O]+, 312.1002[M + H–2CH3–CH3O]+ sinensetin CL
60 19.68 C15H16O4 260.1043 261.1124 0.234 243.1025[M + H–H20]+, 189.0554[M + H–C4H8O]+ meranzin CL
61 19.94 C15H20O2 232.1457 233.1536 0.034 215.1436[M + H–H2O]+, 205.1593[M + H–CO]+, 197.1330[M + H–2H2O]+, 187.1487[M + H–H2O–CO]+ helenine AF
62 20.17 C15H18O3 246.1250 247.1327 –0.181 229.1228[M + H–H2O]+, 201.1279[M + H–H2O–CO]+ zaluzanin C AF
63 20.32 C21H22O8 402.1309 403.1383 –0.404 388.1164[M + H–CH3]+, 373.0928[M + H–2CH3]+, 355.0822[M + H–2CH3–H2O]+, 342.1107[M + H–C2H5O2]+ nobiletin CL
64 20.47 C19H18O6 342.1097 343.1177 0.095 328.0952[M + H–CH3]+, 310.0847[M + H–CH3–H2O]+, 299.0924[M + H–C2H4O]+, 282.0897[M + H–CO–H2O–CH3]+ 3′,4′7,8-tetramethoxyflavone CL
65 20.83 C15H18O3 246.1250 247.1328 –0.041 229.1228[M + H–H2O]+, 201.1279[M + H–H2O–CO]+ irofulven AF
66 20.86 C22H24O9 432.1414 433.1493 –0.049 418.1273[M + H–CH3]+, 403.1039[M + H–2CH3]+, 385.0933[M + H–2CH3–H2O]+ 3,3′,4′,5,6,7,8-heptamethoxyflavone CL
67 21.26 C20H20O7 372.1203 373.1268 –3.617 358.1060[M + H–CH3]+, 343.0826[M + H–2CH3]+ auranetin CL
68 21.33 C10H16 136.1246 137.1326 0.093 109.1013[M + H–C2H4]+, 95.0856[M + H–C3H6]+, 81.0699[M + H–C4H8]+ α-terpinene AF
69 21.85 C20H20O8 388.1152 389.1230 –0.074 356.0905[M + H–CH5O]+, 341.0667[M + H–C2H8O]+, 374.1006[M + H–CH3]+, 359.0772[M + H–2CH3]+, 328.3221[M + H–C2H5O2]+ uumuhengerin CL
70 23.28 C15H18O2 230.1301 231.1378 –0.136 213.1280[M + H–H2O]+, 195.1174[M + H–2H2O]+, 185.1331[M + H–H2O–CO]+, 145.1016[M + H–C4H6O2]+, 131.0860[M + H–C5H8O2]+ dehydrocostus lactone AF
a

Note: tR: retention time; AF: Aucklandia costus Falc; LK: Lindera aggregata (Sims) Kosterm.; CL: Citrus aurantium L.; AL: Areca catechu L.

Table 2. Identification of Compounds in SMT by UHPLC-LTQ-Orbitrap in Negative Ion Modea.

peak no. tR (min) molecularformula calculated mass (m/z) measured mass [M – H] error(ppm) MS/MS fragments identification compound source
14 7.95 C16H18O9 354.0945 353.0865 –0.189 191.0560[M – H–C9H6O3], 179.0352[M–H–C7H10O5], 173.0455[M – H–C9H6O3–H2O], 135.0453[M + H–C8H10O7] chlorogenic acid AL
15 9.34 C16H18O9 354.0945 353.0863 –0.379 191.0559[M – H–C9H6O3] neochlorogenic acid AL
16 9.87 C16H18O9 354.0945 353.0868 0.051 179.0560[M – H–C7H10O5], 173.0452[M – H–C9H6O3–H2O], 191.0560[M – H–C9H6O3], 135.0453[M + H–C8H10O7] 4-dicaffeoylquinic acid AL
24 14.55 C27H32O15 596.1735 595.1632 –2.537 475.1077[M – H–C8H8O], 459.1149[M – H–C8H8O2], 287.0563[M – H–C12H20O9], 271.0617[M – H–C12H20O10], 235.0251[M – H–C16H24O9] neoeriocitrin CL
26 14.81 C27H32O15 596.1735 595.1652 –0.587 459.1149[M – H–C8H8O2], 287.0563[M – H–C12H20O9], 235.0251[M – H–C16H24O9] eriocitrin CL
29 15.26 C27H32O14 580.1786 579.1705 –0.302 271.0608[M – H–C12H20O9], 459.1131[M – H–C8H8O] narirutin CL
31 15.37 C27H30O14 578.1630 577.1536 –1.552 269.0455[M – H–C12H20O9], 413.0869[M – H–C6H12O5], 431.0979[M – H–C6H10O4] rhoifolin CL
33 15.41 C27H32O14 580.1786 579.1704 –0.482 459.1141[M – H–C8H8O], 353.0822[M – H–C12H14O4], 339.0718[M – H–C12H16O5], 313.0715[M – H–C10H18O8], 271.0608[M – H–C12H20O9], 235.0250[M – H–C16H24O8] naringin CL
37 15.64 C28H34O15 610.1892 609.1798 –1.587 489.1440[M – H–C4H8O4], 447.1294[M – H–C6H10O5], 403.1031[M – H–C8H14O6], 343.0826[M – H–C10H18O8], 325.0717[M – H–C10H20O9], 301.0720[M – H–C12H20O9], 286.0484[M – H–C13H23O9], 242.0587[M – H–C14H23O11] hesperidin CL
39 15.86 C28H34O15 610.1892 609.1802 –1.157 489.1440[M – H–C4H8O4], 447.1294[M – H–C6H10O5], 403.1031[M – H–C8H14O6], 343.0826[M – H–C10H18O8], 325.0717[M – H–C10H20O9], 301.0720[M – H–C12H20O9], 286.0484[M – H–C13H23O9], 242.0587[M – H–C14H23O11] neohesperidin CL
42 16.05 C22H24O11 464.1313 463.1233 –0.168 301.0718[M – H–C6H10O5] hesperetin 5-O-glucoside CL
48 17.26 C28H34O14 594.1943 593.1868 0.348 473.1454[M + H–C4H10O4], 327.0875[M + H–C10H20O8], 285.0768[M + H–C12H22O9] neoponcirin CL
a

Note: tR: retention time; AF:Aucklandia costus Falc; LK:Lindera aggregata (Sims) Kosterm.; CL:Citrus aurantium L.; AL:Areca catechu L.

2.1.1. Flavones and Their Glycosides

Flavonoids have a diphenylpropane skeleton bearing two benzene rings (A and B) connected by a pyran ring attached to the A ring, and are further classified into several subclasses (flavones, flavonols, flavanones, flavanonol, and so on).20 In the present study, a total of 26 flavonoids were identified from SMT in the positive and negative mode in which the main source of flavonoids was CL. Among them, five standards, including naringin (peak 33), narirutin (peak 29), neohesperidin (peak 39), hesperetin (peak 40), and nobiletin (peak 63) were first identified and used to explore the fragmentation rules of flavonoids. Briefly, the MS/MS behaviors of aglycones were described by the retro-Diels–Alder (RDA) fragmentation pathway. Successive loss of CO from the ketone group, C-fragmentation and loss of radicals, such as CH3 and CHO, had been described.21 For flavonoid O-glycosides, the neutral loss of 146, 162, and 308 Da were the characteristic fragment ions.22 Taking compound 33 as an example, it gave the [M – H] ion at m/z 579.1704 (C27H31O14). In the MS2 spectra, the main fragment ions were observed at m/z values of 459.1141[M – H–C8H8O], 339.0718[M – H–C12H16O5], 313.0715[M – H–C10H18O8], 271.0608[M – H–C12H20O9], and 235.0250[M – H–C16H24O8]. Thus, compound 33 was unambiguously identified as naringin. Its mass spectrum and proposed fragmentation pathways in negative mode are shown in Figure S2.

Compound 37 and neohesperidin (compound 39) showed the same molecular formula and similar [M – H] ions at m/z values of 609.1798(C28H35O15) and 609.1802(C28H35O15). Moreover, they exhibited the similar fragment ions of m/z values of 489.1440[M – H–C4H8O4], 447.1294[M – H–C6H10O5], 403.1031[M – H–C8H14O6], 343.0826[M – H–C10H18O8], 325.0717[M – H–C10H20O9], 301.0720[M – H–C12H20O9], 286.0484[M – H–C13H23O9], and 242.0587[M – H–C14H23O11], so it was tentatively identified as hesperidin.

Similarly, based on the chromatographic behavior and the similar fragmentation pathways, seven flavone glycosides (peaks 18, 24, 26, 31, 41, 42, and 48) were tentatively identified as lonicerin, neoeriocitrin, eriocitrin, rhoifolin, isosakuranin, hesperetin 7-O-β-d-glucuronide, and neoponcirin, respectively.2325 The remaining compounds (peaks 30, 35, 36, 46, 48, 49, 50, 55, 56, 59, 66, 67, and 69) were tentatively identified as flavonoids via comparing their exact molecular masses and MS/MS spectra with the literature data.25 Detailed information on these compounds is summarized in Tables 1 and 2.

2.1.2. Alkaloids

Alkaloids are a group of naturally occurring chemical compounds that mostly contain basic nitrogen atoms.26 In the present study, the main source of alkaloids was AL and LK. Fourteen alkaloids were tentatively identified, including four piperidines (guvacoline, arecaidine, guvacine, and arecoline), eight isoquinolines (pronuciferine, laurolitsine, norisoboldine, boldine, laurotetanine morphine acetate, naloxone, and reticuline), and two organic amine alkaloids (N-methyltyramine and synephrine). Among them, two reference compounds, guvacoline (peak 9) and arecoline (peak 10) were first identified and used to characterize the fragmentation behaviors. Their fragment ions are summarized in Tables 1 and 2, and the losses of methanol, the methyl group, methylamine (CH3NH), H2O, and CO were their main pathways.27,28 Taking compound 9 as an example, it exhibited a [M + H]+ ion at an m/z of 142.0862(C7H12NO2) and in the positive ion spectrum, the main fragment ions at m/z values of 110.0602[M + H–CH4O]+, 113.0598[M + H–CH3N]+, 96.0807[M + H–CH2O2]+, and 81.0334[M + H–C2H7NO]+ were detected. Thus, compound 9 was inferred as guvacoline. The proposed fragmentation pathway of guvacoline was shown in Figure S3. Similarly, 13 alkaloids (peaks 3, 4, 7, 10, 12, 19, 23, 25, 27, 34, 38, 44, and 47) were identified as guvacine, arecaidine, synephrine, arecoline, N-methyltyramine, laurolitsine, boldine, norisoboldine, pronuciferine, reticuline, laurotetanine, morphine acetate, and naloxone, respectively, based on the MS information as well as the literature reports.2933

2.1.3. Coumarins

In the present study, the main source of coumarins was CL. Four coumarins (7-hydroxycoumarin, 5,7-dihydroxycoumarin, isomerancin, and meranzin) were identified from SMT, and the structures could be postulated based on high-accuracy quasi-molecular ion and MSn mass spectrometry. Among them, one standard compound (7-hydroxycoumarin) was first used to characterize the fragmentation behaviors and explore the fragmentation rules. For most of coumarins, the primary fragmentation mechanism was first a neutral loss of side chain then a loss of CO and CO2 in MS/MS mode.34 Compound 22 gave a [M + H]+ ion at an m/z of 163.0389(C9H7O3) and yielded fragment ions at m/z values of 145.0286[M + H–H2O]+, 119.0494[M + H–CO2]+, and 107.0493[M + H–2CO]+, which suggested that it was 7-hydroxycoumarin. The specific cleavage process is shown in Figure S4. Similarly, compounds 17, 28, and 60 were identified as 5,7-dihydroxycoumarin, isomerancin, and meranzin, respectively.35

2.1.4. Amino Acids

Amino acids are organic compounds containing amine (−NH2) and carboxyl (−COOH) functional groups, along with a side chain (R group) specific to each amino acid.36 In the present study, the main source of amino acids was AL. Four amino acids and one derivative of aspartic acid were identified. Among them, two standard compounds, phenylalanine (peak 13) and valine (peak 5), were first identified and used to characterize the fragmentation behaviors and explore the fragmentation rules for amino acids. Their fragmentation mechanisms have been extensively studied in positive ion mode. Because of the presence of amino and acid groups in amino acid structure, fragment ions were usually generated by the loss of NH3 (17 Da), H2O (18 Da), CO2 (44 Da), and HCOOH (46 Da).28 Taking compound 13 as an example, the precise molecular weight was 166.0862 (C9H12NO2), and in the positive ion spectrum, the main fragment ions were observed at m/z values of 149.0599[M + H–NH3]+, 131.0493[M + H–NH3–H2O]+, 120.0810[M + H–H2O–CO]+, and 80.0493[M + H–C4H6O2]+. Therefore, compound 13 was unambiguously identified as phenylalanine. The detailed fragmentation pathways of compound 13 are shown in Figure S5. Peak 1 exhibited a [M + H]+ ion at an m/z of 133.0608 and gave the fragment ions at m/z values of 116.0343[M + H–NH3]+, 87.0553[M + H–CO–H2O]+, 70.0286[M + H–CO–H2O–NH3]+. Thus, it was identified as the derivative of aspartic acid, asparagine. Similarly, peaks 2, 5, and 8 were identified or tentatively characterized as proline, valine, and tyrosine, respectively, from SMT (Tables 1 and 2) based on the MS information and reported data.3739

2.1.5. Sesquiterpenoid and Derivatives

In the present study, the main source of sesquiterpenoids was AF and LK. A total of 13 sesquiterpenoids were found in SMT. Among them, four standard compounds, linderane (peak 45), costunolide (peak 61), isoalantolactone (peak 58), and helenine (peak 53) were first identified and used to characterize the fragmentation behaviors. The details of the identified components are summarized in Tables 1 and 2. Their MS/MS behaviors can be described as the loss of radicals, such as H2O, CH3, and CO.40 For example, compound 45 showed a [M + H]+ ion at an m/z of 261.1120 with an elemental composition of C15H17O4. The MS/MS spectrum of [M + H]+ exhibited fragment ions at m/z values of 243[M + H–H2O]+, 189[M + H–CH3–C3H5O]+, 177[M + H–C5H8O]+, and 217[M + H–CO2]+. Thus, compound 52 was identified as linderane. The detailed fragmentation pathways of compound 45 are shown in Figure S6. Based on these similar fragmentation patterns, nine sesquiterpenoids (peaks 20, 21, 43, 52, 54, 57, 62, 65, and 70) were identified as picriside B, saussureanine D, ethyllucidone, saussureanine B, saussureanine A, isozaluzanin C, zaluzanin C, garbanzol, and dehydrocostus lactone, respectively.4044

2.1.6. Organic Acids

In the present study, the main source of organic acids was AL and CL, and six organic acids were identified from SMT. Peak 32 gave a [M + H]+ ion at an m/z of 113.0595(C6H8O2). The fragment ions at m/z values of 95.0492[M + H–H2O]+, 85.0648[M + H–CO]+, 67.0541[M + H–CO–H2O]+ suggested the structure of carboxyl. Based on the fragmentation information, peak 32 was tentatively identified as sorbic acid. Peaks 6 and 11 were isomers and yielded the fragment ions at m/z 127.0389. They also exhibited similar fragmentation ions. According to their MS behavior and reported data, peaks 6 and 11 were tentatively identified as maltol and triacetate lactone, respectively.45,46 Peaks 14, 15, and 16 were isomers of chlorogenic acid, which gave [M – H] ions at an m/z of 353.0865 (C16H19O9). The characteristic fragment ions at m/z values of 191.0560[M – H–C9H6O3], 179.0352[M – H–C7H10O5], 173.0455[M – H–C9H6O3–H2O], and 135.0453[M + H–C8H10O7] suggest that these compounds may be the ester of caffeic acid and quinic acid.47

2.1.7. Miscellaneous

Another two obvious peaks in the extracted ion chromatogram of SMT were identified. Among them, compound 51 was tentatively assigned as 2-methyl-5-propan-5-ylcyclohex-2-en-1-one. Compound 68, with [M + H]+ at an m/z of 137.1326(C10H17), yielded ions at m/z values of 109.1013[M + H–C2H4]+, 95.0856[M + H–C3H6]+, and 81.0699[M + H–C4H8]+. It was plausibly identified as α-terpinene.48

2.2. Putative Targets of SMT

A total of 461 targets of SMT were predicted by MedChem Studio, as given in Table S2. The results showed that the candidate compounds could act on multiple targets, and one target could also be linked to multiple components.

2.3. Known Therapeutic Targets Acting on GIDs

We collected 315 known therapeutic targets of GIDs from three databases after eliminating redundancy, of which six were from Therapeutic Target Database (TTD), 19 were from DrugBank and 290 were from Online Mendelian Inheritance in Man (OMIM). Detailed information about putative targets of GIDs is provided in Table S3.

2.4. Network and Pathway Analysis

To illustrate the potential relationships between SMT and GIDs, a protein–protein interaction (PPI) network of interactions between SMT-related targets and GID-related targets was established. The network contains 515 nodes and 5957 edges. Detailed information about the PPI data is provided in Table S4. After calculating the values of the three topological features of all nodes in the network, 170 nodes were identified as major hubs because they satisfied the screening criteria (degree centrality (DC) > 20, betweenness centrality (BC) > 0.0015, closeness centrality (CC) > 0.3757). Among them, 152 hubs were putative targets of SMT and 16 hubs were known therapeutic targets of GIDs and 3 hubs were both putative targets of SMT and known therapeutic targets of GIDs. The details of these major hubs are shown in Table S5.

To facilitate scientific interpretation of the action mechanisms of SMT on GIDs, GO enrichment analysis was conducted on these major hubs. As depicted in Figure 2a–c, the top 10 significant GO entries of the biological process (BP), cellular components (CC), and molecular function (MF) were obtained after filtering by a parameter P-value cut-off of ≤0.05. The major biological processes included drug response, chemical synaptic transmission, response to hypoxia, phospholipase C (PLC)-activating G protein-coupled receptor (GPCR) signaling pathway, signal transduction, transport, calcium ion transmembrane transport, cellular amino acid biosynthetic process, cytosolic calcium ion concentration, cellular calcium ion homeostasis (Figure 2a). The majority of the protein responses were situated in a variety of cell components like postsynaptic membrane, plasma membrane, dendrite, neuronal cell body, cell junction, synapse, voltage-gated calcium channel complex, cytosol, cell surface, and mitochondrion (Figure 2b). Moreover, the associated molecular functions included ligand-gated ion channel activity, enzyme binding, drug binding, amino acid binding, dopamine binding, dopamine neurotransmitter receptor activity, heme binding, oxygen binding, voltage-gated calcium channel activity, and receptor binding (Figure 2c).

Figure 2.

Figure 2

GO term performance and pathway enrichment analysis of major hubs. (a) GO term performance by a biological process (BP); (b) GO term performance by a molecular function (MF); (c) GO term performance by a cellular component (CC); (d) Pathway enrichment analysis by KEGG. The ordinate stands for GO terms or main pathways, the primary abscissa stands for minus log10(P).

To further explore the functional effects and systemic association of the major hubs, KEGG pathway enrichment analysis was carried out. As depicted in Figure 2d, the top 10 pathways were obtained, which could be classified into three major functional modules according to the functional annotation (Figure 3). Maximum module was associated with neuro-immune regulation (neuroactive ligand–receptor interaction, cholinergic synapse, inflammatory regulation of TRP channels, HIF-1 signaling pathway, and biosynthesis of amino acids) and the second module was sorted as signal transduction (calcium signaling pathway, cAMP signaling pathway, and MAPK signaling pathway), while the minimum module was concentrated in cardiomyocyte contraction (vascular smooth muscle contraction and adrenergic signaling in cardiomyocytes). The network diagram composed of the interactions among the active components of SMT, major SMT-related targets, and major pathways was finally generated, as shown in Figure 3. It was worthy to note that the calcium signaling pathway was highly enriched in KEGG pathway analysis, which played an important role in smooth muscle contraction49 and thus may mainly contribute to therapeutic effects of SMT on the improvement of gastrointestinal motility.50

Figure 3.

Figure 3

SMT herbs–major hubs–main pathways network. Green diamonds represent each herb in SMT; round purple nodes represent putative targets of chemicals in SMT; round yellow nodes represent known therapeutic targets for GIDs; round red nodes represent both putative targets of components in SMT and known therapeutic targets for GIDs; orange rectangles represent top 10 pathways from enrichment analysis of major targets; edges represent interactions among SMT herbs, putative targets, known therapeutic targets for the treatment of GIDs, and main pathways.

2.5. SMT Attenuates the Gastrointestinal Dysmotility via Regulating the Calcium Signaling Pathway

Gastrointestinal dysmotility (GD), affecting the neuromuscular functions needed for propulsion of intraluminal contents51 is closely associated with the pathophysiology of functional GIDs.52 According to the network analysis (Table S6), the SMT putative targets associated with the calcium signaling pathway include cholinergic receptor muscarinic 1(CHRM1), CHRM2, dopamine receptor D1 (DRD1), DRD5, adrenoceptor alpha 1A (ADRA1A), ADRA1B, ADRA1D, adrenoceptor beta 2 (ADRB2), 5-hydroxytryptamine receptor 2A (HTR2A), HTR2B, HTR2C, protein kinase cAMP-activated catalytic subunit alpha (PRKACA), calcium voltage-gated channel subunit alpha1 A (CACNA1A), CACNA1B, CACNA1C, CACNA1D, CACNA1H, CACNA1S, cholinergic receptor nicotinic alpha 7 subunit (CHRNA7), glutamate ionotropic receptor NMDA type subunit 1(GRIN1), GRIN2A, GRIN2C, protein tyrosine kinase 2 beta (PTK2B), protein kinase C alpha (PRKCA), PRKCB, and nitric oxide synthase 1 (NOS1). Figure 4 shows the calcium signaling pathway affected by major putative targets of SMT, and the details are discussed below.

Figure 4.

Figure 4

Illustration of the calcium signaling pathway induced by major putative targets of SMT.

Ca2+ ions play key roles in the process known as excitation–contraction coupling.53 Upon an appropriate autacoid or depolarizing stimulation, extracellular Ca2+ ions enter smooth muscle cells primarily through voltage-operated channels (VOCs) and receptor-operated channels (ROCs) of the plasma membrane, which in turn induces the release of Ca2+ ions from sarcoplasmic reticulum (SR), leading to a transient rise in intracellular Ca2+. Then, the contraction of gastrointestinal smooth muscles was initiated via the activation of Ca2+/calmodulin (CaLM)-dependent myosin light chain kinase (MLCK).5456 According to our predicted results, several alkaloid components of SMT were involved in Ca2+ influx and Ca2+ release from SR, such as boldine57 and arecoline.58 Therefore, SMT may modify the GD status by increasing intracellular Ca2+.

The PTK/PLC pathway has been recognized as an important upstream signaling leading to calcium release.59 In response to growth factors, PTK activates phospholipase C (PLC) by phosphorylating the gamma 1 isoform of PLC.60 PLC is a critical enzyme in the regulation of phosphatidylinositol (PIP2) metabolism, which catalyzes PIP2 to produce diacylglycerol (DAG) and inositol 1,4,5-triphosphate (IP3). IP3 acts on its receptors to release Ca2+ from intracellular stores. Also, DAG could trigger the activation of protein kinase C, which plays an important role in cell growth.60,61 Accumulated evidences indicate that plant flavonoids exert significant inhibition effects on PKC, which may partially contribute to the anti-GID function of SMT.62,63

GPCR signaling also involves in the regulation of luminal SR Ca2+ channel. In gastrointestinal smooth muscle cells, neurotransmitters or autacoids trigger the release of the α subunit of a guanine nucleotide-binding protein Gs. Then, Gs binds to adenylyl cyclase (ADCY), which catalyzes the production of cyclic adenosine monophosphate (cAMP). Subsequently, cAMP induces the activation of PKA and then leads to the phosphorylation of phospholamban (PLN), which in turn alleviates the suppression effects of PLN on Ca2+-transport ATPases (SERCA), thereby improving SR Ca2+ uptake and contributing to relaxation.56 In addition, the Gq-PLC-IP3-Ca2+ pathway also plays an important role in maintaining calcium homeostasis.64 It has been evident that many components from SMT could ameliorate GD via regulating the GPCR pathway, such as N-methyltyramin65 and naringenin.66

Other downstream targets such as NOS, PTK2B, and PKC could also be regulated by Ca2+/(CaMKII) and thus involved in the pathological process of GD. Inducible NOS is responsible for the increased NO production, leading to smooth muscle relaxation. PTK2B, a cytoplasmic tyrosine kinase, could initiate multiple signaling pathways associated with several cellular functions such as cell migration and proliferation.67,68 Thus, NOS, and PTK2B may also be the potential targets of SMT on GD.

Recently, the pharmacological activity of a single herb of SMT on gastrointestinal disorders has also been reported. AL could promote gastrointestinal motility by promoting the secretion of glucagon like peptide-1, bile, and cholesterol.69 The flavonoids from CL also possess prominent gastrointestinal motility promoting efficacy. However, AF produced actually a spasmolytic effect on gastrointestinal motility via the suppression of muscarinic receptors, 5-hydroxytryptamine receptors, and Ca2+ influx.70 In addition, LK has been reported to show a protective effect on the intestinal barrier, ameliorating gut microflora dysbiosis and attenuating inflammation.71 According to the TCM theory, the combination use of these four herbs would achieve higher efficacy and fewer side effects compared with a single herb. Taken together, the various types of constituents contained in SMT exert their synergistic effects on GIDs through acting on multiple target genes/proteins on multiple pathways. However, this work has some inevitable limitations. First, and particularly, some identified constituents may not be absorbed into the blood circulatory system, and in many situations, it is the metabolites, not the parent compounds, which have therapeutic effects. Second, it is really hard to confirm the inhibitory or activated effects of the predicted targets. Lastly and importantly, the in silicon work may be influenced by possible biases to frequently studied biological processes.17

3. Conclusions

In the present study, UHPLC-LTQ-Orbitrap combined with a network pharmacology method was used to unveil the chemical basis and investigate the action mechanism of SMT for treating GIDs. The results showed that the pharmacological mechanism of SMT in the treatment of GIDs may be mainly associated with the calcium signaling pathway. Moreover, 26 core genes were identified as key active targets involved in this pathway, and they were mapped to 51 key active components. However, to enhance the reliability of the results, further experiments were demanded to validate these hypotheses.

4. Materials and Methods

4.1. Reagents and Chemicals

SMT was purchased from Hunan Hansen Pharmaceutical Co., Ltd. (Yiyang, Hunan Province, China), which was prepared according to the preparation process of Chinese Pharmacopoeia (2015 edition). HPLC grade acetonitrile, methanol, and formic acid used in this study were purchased from Fisher Scientific. High pure water was prepared by using a Millipore Milli Q plus purification system. All other reagents were analytical grade and commercially available. Reference standards of valine, guvacoline, arecoline, phenylalanine, 7-hydroxycoumarin, narirutin, naringin, neohesperetin, hesperetin, linderane, costunolide, isoalantolactone, helenine, and nobiletin were purchased from the National Institute for the Control of Pharmaceutical and Biological Products (Beijing, China). All standards were of at least 98% purity and were suitable for UHPLC-LTQ-Orbitrap analysis.

4.2. Sample Preparation

A volume of 1 mL SMT was filtered through a 0.22 μm nylon membrane filter before LC/MS analysis. Individual stock solutions of 14 compounds (500 μg/mL) were prepared by dissolving accurately weighted amount of reference compound in HPLC grade methanol, respectively. Then, all the 14 standards were mixed and serially diluted to 50 μg/mL. The solution was filtered through a filter (0.22 μm) and analyzed directly by LC/MS.

4.3. LC–MS/MS System

Sample analyses were performed on an ultimate 3000 LC system coupled to an LTQ Orbitrap mass spectrometer via an ESI interface. The chromatography system was composed of an autosampler, a column compartment, and two pumps. Xcalibur, Metworks, and Mass Frontier 7.0 software packages were used for data collection and data analysis. Chromatographic separations were performed on a Thermo Scientific BOS Hypersil C18 column (2.1 mm × 150 mm, 2.4 μm). The mobile phase was composed of water containing 0.1% formic acid (A) and acetonitrile (B). The LC gradient program [time (in min)/% mobile phase B] was described as follows: 0.01/2, 5/2, 5.1/10, 10/10, 25/80, 26/2, and 31/2. The chromatographic runs were performed at a flow rate of 0.300 mL/min, and the injection volume was 3 μL. The column was maintained at 35 °C.

The ESI source parameters were set as follows: the capillary temperature, 250 °C; source voltage and spray voltage, 5 kV; sheath gas (N2) flow, 35 psi. The ESI source was operated in both positive and negative ionization modes. In the Fourier transform (FT) cell, full MS scans were acquired in the range of m/z 50–2000. The MS/MS experiments were set as data-dependent scans.

4.4. Prediction of Putative Targets of SMT

MedChem Studio (MedChem Studio, 3.0; Simulations Plus, Inc., Lancaster, CA, USA, 2012) was used to predict the targets associated with the identified SMT components based on structure similarity. The similarity threshold was set at 0.7.

4.5. Known Therapeutic Targets of GIDs

Known human therapeutic targets of GIDs were retrieved from three sources with the query “gastrointestinal disorders”. The first one was TTD (http://db.idrblab.net/ttd/), which could offer abundant information about the known therapeutic protein and nucleic acid targets.72 The second one was DrugBank (https://www.drugbank.ca/), which is a unique bioinformatics and cheminformatics resource containing comprehensive molecular information about drugs and their corresponding targets.73 The last one was OMIM (http://www.omim.org/), which is an authoritative database of human genes and genetic disorders.74

4.6. Protein–Protein Interaction Network

The PPI network of SMT-related targets and the targets of GIDs was constructed by using STRING database (https://string-db.org/), with the species limited to Homo sapiens. STRING is a web-enabled database that provides extensive information about known and predicted protein–protein interactions.75 The cutoff value was set at 0.4.

4.7. Network Construction and Analysis

An interaction network of putative SMT target-known therapeutic targets of GIDs was constructed using the PPI data from STRING database, which was visualized by Cytoscape software 3.7.1 (https://cytoscape.org/). Cytoscape is a popular open-source software for biological network visualization and data integration.76 Next, we used three topological properties, including DC, BC, and CC to evaluate the topological importance of each node. The median values of the three parameters were set as the cutoff values.77

4.8. Pathway Enrichment Analysis

To unveil the potential functions of the critical targets involved in the SMT-mediated treatment of GIDs, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and Gene Ontology (GO) enrichment analysis were carried out by the use of the Database for Annotation, Visualization, and Integrated Discovery (DAVID) system (http://david.abcc.ncifcrf.gov/home.jsp/, v6.7).78P values less than 0.05 were considered statistically significant.

Acknowledgments

This work is supported by the special research grant for non-profit public service (nos. 201507004 and BUCMHZ-2015026).

Supporting Information Available

The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsomega.0c05680.

  • Chemical structures of the identified compounds, total ion chromatograms of SMT, and mass spectra and proposed fragmentation pathways of naringin, guvacoline, 7-hydroxycoumarin, phenylalanine, and linderane (PDF)

  • Putative targets for SMT, known therapeutic targets acting on GIDs, PPI network between SMT-related targets and GID-related targets, major hubs in the network, and potential active ingredients involved in the calcium signaling pathway (XLSX)

Author Contributions

Z.L., G.Y., and X.H. contributed equally to this study.

The authors declare no competing financial interest.

Supplementary Material

ao0c05680_si_001.pdf (1.2MB, pdf)
ao0c05680_si_002.xlsx (278KB, xlsx)

References

  1. Carey S.; Ferrie S.; Young J.; Allman-Farinelli M. Long-term nutrition support in gastrointestinal disease-A systematic review of the evidence. Nutrition 2012, 28, 4–8. 10.1016/j.nut.2011.07.004. [DOI] [PubMed] [Google Scholar]
  2. Sanger G. J. Gastrointestinal diseases. Drug Discovery Today: Ther. Strategies 2007, 4, 153–154. 10.1016/j.ddstr.2008.04.001. [DOI] [Google Scholar]
  3. Peery A. F.; Crockett S. D.; Barritt A. S.; Dellon E. S.; Eluri S.; Gangarosa L. M.; Jensen E. T.; Lund J. L.; Pasricha S.; Runge T.; Schmidt M.; Shaheen N. J.; Sandler R. S. Burden of Gastrointestinal, Liver, and Pancreatic Diseases in the United States. Gastroenterology 2015, 149, 1731–1741.e3. 10.1053/j.gastro.2015.08.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Peery A. F.; Crockett S. D.; Murphy C. C.; Lund J. L.; Dellon E. S.; Williams J. L.; Jensen E. T.; Shaheen N. J.; Barritt A. S.; Lieber S. R.; Kochar B.; Barnes E. L.; Fan Y. C.; Pate V.; Galanko J.; Baron T. H.; Sandler R. S. Burden and Cost of Gastrointestinal, Liver, and Pancreatic Diseases in the United States: Update 2018. Gastroenterology 2019, 156, 254–272.e11. 10.1053/j.gastro.2018.08.063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Hatton G. B.; Madla C. M.; Rabbie S. C.; Basit A. W. All disease begins in the gut: Influence of gastrointestinal disorders and surgery on oral drug performance. Int. J. Pharm. 2018, 548, 408–422. 10.1016/j.ijpharm.2018.06.054. [DOI] [PubMed] [Google Scholar]
  6. Neamsuvan O.; Phumchareon T.; Bunphan W.; Kaosaeng W. Plant materials for gastrointestinal diseases used in Chawang District, Nakhon Si Thammarat Province, Thailand. J. Ethnopharmacol. 2016, 194, 179–187. 10.1016/j.jep.2016.09.001. [DOI] [PubMed] [Google Scholar]
  7. Karthik R.; Karthik K. S.; David C.; Ameerunnisa; Keerthi G. Oral adverse effects of gastrointestinal drugs and considerations for dental management in patients with gastrointestinal disorders. J. Pharm. BioAllied Sci. 2012, 4, 239–241. 10.4103/0975-7406.100217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Suo T.; Liu J.; Chen X.; Yu H.; Wang T.; Li C.; Wang Y.; Wang C.; Li Z. Combining Chemical Profiling and Network Analysis to Investigate the Pharmacology of Complex Prescriptions in Traditional Chinese Medicine. Sci. Rep. 2017, 7, 40529. 10.1038/srep40529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Yu G.; Wang W.; Wang X.; Xu M.; Zhang L.; Ding L.; Guo R.; Shi Y. Network pharmacology-based strategy to investigate pharmacological mechanisms of Zuojinwan for treatment of gastritis. BMC Complementary Altern. Med. 2018, 18, 292. 10.1186/s12906-018-2356-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Cai G.-X.; Liu B.-Y.; Yi J.; Chen X.-M.; Liu F.-L. Simotang enhances gastrointestinal motility, motilin and cholecystokinin expression in chronically stressed mice. World J. Gastroenterol. 2011, 17, 1594–1599. 10.3748/wjg.v17.i12.1594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Liang M.; Xu X.. Effect Analysis of Simotang Oral Liquid on Functional Dyspepsia of Incoordination between the Liver and the Spleen and Gastrointestinal Hormone. In Proceedings of the 2016 7th International Conference on Mechatronics, Control and Materials(ICMCM 2016); Atlantis Press: 2016. [Google Scholar]
  12. Dai C.; Liu N.; Chen W.; Qian W.; Hou X. Simo decoction promotes contraction of antral circular smooth muscle mainly via muscarinic M3 receptor. J. Ethnopharmacol. 2012, 144, 270–276. 10.1016/j.jep.2012.09.008. [DOI] [PubMed] [Google Scholar]
  13. Ping W.; Qun X.; Wei M. Clinical Observation of Simo Decoction Oral Liquid for Treatment of Constipation-predominant Irritable Bowel Syndrome. J. Guangzhou Univ. Trad. Chin. Med. 2019, 36, 177–180. [Google Scholar]
  14. Gao L.; Wang K.-x.; Zhou Y.-z.; Fang J.-s.; Qin X.-m.; Du G.-h. Uncovering the anticancer mechanism of Compound Kushen Injection against HCC by integrating quantitative analysis, network analysis and experimental validation. Sci. Rep. 2018, 624. 10.1038/s41598-017-18325-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Feng G.; Sun Y.; Liu S.; Song F.; Pi Z.; Liu Z. Stepwise targeted matching strategy from in vitro to in vivo based on ultra–high performance liquid chromatography tandem mass spectrometry technology to quickly identify and screen pharmacodynamic constituents. Talanta 2019, 194, 619–626. 10.1016/j.talanta.2018.10.074. [DOI] [PubMed] [Google Scholar]
  16. Zhang Y.-q.; Mao X.; Guo Q.-y.; Lin N.; Li S. Network Pharmacology-based Approaches Capture Essence of Chinese Herbal Medicines. Chin. Herb. Med. 2016, 8, 107–116. 10.1016/S1674-6384(16)60018-7. [DOI] [Google Scholar]
  17. Yu G.; Luo Z.; Zhou Y.; Zhang L.; Wu Y.; Ding L.; Shi Y. Uncovering the pharmacological mechanism of Carthamus tinctorius L. on cardiovascular disease by a systems pharmacology approach. Biomed. Pharmacother. 2019, 117, 109094. 10.1016/j.biopha.2019.109094. [DOI] [PubMed] [Google Scholar]
  18. Zhang W.; Chen Y.; Jiang H.; Yang J.; Wang Q.; Du Y.; Xu H. Integrated strategy for accurately screening biomarkers based on metabolomics coupled with network pharmacology. Talanta 2020, 211, 120710. 10.1016/j.talanta.2020.120710. [DOI] [PubMed] [Google Scholar]
  19. Wang F.; Zhang Q.; Lu Z.; Wang Q.; Wang M.; Liu Y.; Fu S.; Gao X.; Tang X. Identification of chemical constituents in traditional Chinese medicine formula using HPLC coupled with linear ion trap-Orbitrap MS from high doses of medicinal materials to equivalent doses of formula: Study on Xiang-Sha-Liu-Jun-Zi-Jia-Jian granules. J. Sep. Sci. 2016, 39, 1619–1627. 10.1002/jssc.201501223. [DOI] [PubMed] [Google Scholar]
  20. Zou D.; Wang J.; Zhang B.; Xie S.; Wang Q.; Xu K.; Lin R. Analysis of chemical constituents in Wuzi-Yanzong-Wan by UPLC-ESI-LTQ-Orbitrap-MS. Molecules 2015, 20, 21373–21404. 10.3390/molecules201219765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Wang S.; Chen L.; Leng J.; Chen P.; Fan X.; Cheng Y. Fragment ion diagnostic strategies for the comprehensive identification of chemical profile of Gui-Zhi-Tang by integrating high-resolution MS, multiple-stage MS and UV information. J. Pharm. Biomed. Anal. 2014, 98, 22. 10.1016/j.jpba.2014.05.013. [DOI] [PubMed] [Google Scholar]
  22. Liu Q.; Jiao Z.; Liu Y.; Li Z.; Shi X.; Wang W.; Wang B.; Zhong M. Chemical profiling of San-Huang decoction by UPLC-ESI-Q-TOF-MS. J. Pharm. Biomed. Anal. 2016, 131, 20–32. 10.1016/j.jpba.2016.07.036. [DOI] [PubMed] [Google Scholar]
  23. Nishiura M.; Kamiya S.; Esaki S.; Ito F. Flavonoids in citrus and related genera: Part II. Isolation and identification of isonaringin and neoeriocitrin from citrus. Agric. Biol. Chem. 1971, 35, 1683–1690. 10.1271/bbb1961.35.1683. [DOI] [Google Scholar]
  24. Chen H.-F.; Zhang W.-G.; Yuan J.-B.; Li Y.-G.; Yang S.-L.; Yang W.-L. Simultaneous quantification of polymethoxylated flavones and coumarins in fructus aurantii and fructus aurantii immaturus using HPLC-ESI-MS/MS. J. Pharm. Biomed. Anal. 2012, 59, 90. 10.1016/j.jpba.2011.10.013. [DOI] [PubMed] [Google Scholar]
  25. Chuang C.-C.; Wen W.-C.; Sheu S.-J. Classification of Aurantii Fructus samples by multivariate analysis. J. Sep. Sci. 2007, 30, 1827–1832. 10.1002/jssc.200700016. [DOI] [PubMed] [Google Scholar]
  26. Maldoni B. Alkaloids: Isolation and purification. J. Chem. Educ. 1991, 68, 700. 10.1021/ed068p700. [DOI] [Google Scholar]
  27. Xu J.-D.; Mao Q.; Shen H.; Zhu L.-Y.; Li S.-L.; Yan R. Ultra-high performance liquid chromatography coupled with photo-diode array and quadrupole/time-of-flight mass spectrometry based chemical profiling approach to evaluate the influence of preparation methods on the holistic quality of Qiong-Yu-Gao, a traditional complex herbal medicine. J. Chromatogr. A 2013, 1304, 154–168. 10.1016/j.chroma.2013.07.023. [DOI] [PubMed] [Google Scholar]
  28. Liu M.-H.; Tong X.; Wang J.-X.; Zou W.; Cao H.; Su W.-W. Rapid separation and identification of multiple constituents in traditional Chinese medicine formula Shenqi Fuzheng injection by ultra-fast liquid chromatography combined with quadrupole-time-of-flight mass spectrometry. J. Pharm. Biomed. Anal. 2013, 74, 141–155. 10.1016/j.jpba.2012.10.024. [DOI] [PubMed] [Google Scholar]
  29. Yuan J.; Yang D.; Liang Y.; Gao W.; Ren Z.; Zeng W.; Wang B.; Han J.; Guo D. Alkaloids from areca (betel) nuts and their effects on human sperm motility in vitro. J. Food Sci. 2012, 77, T70–T78. 10.1111/j.1750-3841.2012.02653.x. [DOI] [PubMed] [Google Scholar]
  30. Srimany A.; George C.; Naik H. R.; Pinto D. G.; Chandrakumar N.; Pradeep T. Developmental patterning and segregation of alkaloids in areca nut (seed of Areca catechu) revealed by magnetic resonance and mass spectrometry imaging. Phytochemistry 2016, 125, 35–42. 10.1016/j.phytochem.2016.02.002. [DOI] [PubMed] [Google Scholar]
  31. Luo Y.; Liu M.; Xia Y.; Dai Y.; Chou G.; Wang Z. Therapeutic effect of norisoboldine, an alkaloid isolated from Radix Linderae, on collagen-induced arthritis in mice. Phytomedicine 2010, 17, 726. 10.1016/j.phymed.2010.01.013. [DOI] [PubMed] [Google Scholar]
  32. Gan L.-S.; Yao W.; Mo J.-X.; Zhou C.-X. Alkaloids from Lindera aggregata. Nat. Prod. Commun. 2009, 4, 1934578X0900400111. 10.1177/1934578X0900400111. [DOI] [PubMed] [Google Scholar]
  33. Kozuka M.; Yoshikawa M.; Sawada T. Alkaloids from Lindera strychnifolia. J. Nat. Prod. 1984, 47, 1063. 10.1021/np50036a043. [DOI] [PubMed] [Google Scholar]
  34. Wang P.; Wang B.; Xu J.; Sun J.; Yan Q.; Ji B.; Zhao Y.; Yu Z. Detection and chemical profiling of Ling-Gui-Zhu-Gan decoction by ultra performance liquid chromatography-hybrid linear ion trap-orbitrap mass spectrometry. J. Chromatogr. Sci. 2015, 53, 263–273. 10.1093/chromsci/bmu051. [DOI] [PubMed] [Google Scholar]
  35. Zhao J.; Hu D.-j.; Lao K.; Yang Z.-m.; Li S.-p. Advance of CE and CEC in phytochemical analysis (2012–2013). Electrophoresis 2014, 35, 205–224. 10.1002/elps.201300321. [DOI] [PubMed] [Google Scholar]
  36. Ohtake N.; Takano A.; Ito S.; Yamazaki A.; Fujikake H.; Sueyoshi K.; Ohyama T. Quantitative and isotopic analysis of amino acids, allantoin, and allantoic acid in soybeans by LC-MS using the atmospheric pressure chemical ionization method. Soil Sci. Plant Nutr. 2004, 50, 241–248. 10.1080/00380768.2004.10408473. [DOI] [Google Scholar]
  37. Sun M.; Yan D.; Yang X.; Xue X.; Zhou S.; Liang S.; Wang S.; Meng J. Quality assessment of crude and processed Arecae semen based on colorimeter and HPLC combined with chemometrics methods. J. Sep. Sci. 2017, 40, 2151–2160. 10.1002/jssc.201700006. [DOI] [PubMed] [Google Scholar]
  38. Amudhan M. S.; Begum V. H.; Hebbar K. B. A review on phytochemical and pharmacological potential of Areca catechu. L seed. Int. J. Pharm. Sci. Res. 2012, 3, 182. [Google Scholar]
  39. Hu M.-B.; Peng W.; Liu Y.-J.; Yan D.; Chen X.; Wu C.-J. Maillard reaction induces changes in saccharides and amino acids during stir-baking of areca nuts. Trop. J. Pharm. Res. 2016, 15, 2107–2112. 10.4314/tjpr.v15i10.8. [DOI] [Google Scholar]
  40. Yan X.; Zhang Q.; Feng F. Chemical profiling approach to evaluate the influence of traditional and simplified decoction methods on the holistic quality of Da-Huang-Xiao-Shi decoction using high-performance liquid chromatography coupled with diode-array detection and time-of-flight mass spectrometry. J. Sep. Sci. 2016, 39, 1442–1453. 10.1002/jssc.201501326. [DOI] [PubMed] [Google Scholar]
  41. Vijayakannan R.; Karan M.; Dutt S.; Jain V.; Vasisht K. A rapid densitometric TLC method for simultaneous analysis of costunolide and dehydrocostus lactone in Saussurea costus. Chromatographia 2006, 63, 277–281. 10.1365/s10337-006-0733-x. [DOI] [Google Scholar]
  42. Vadaparthi P. R. R.; Kumar K.; Sarma V. U. M.; Hussain Q. A.; Babu K. S. Estimation of costunolide and dehydrocostus lactone in Saussurea lappa and its polyherbal formulations followed by their stability studies using HPLC-DAD. Pharmacogn. Mag. 2015, 11, 180–190. 10.4103/0973-1296.149736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Gan L.-S.; Zheng Y.-L.; Mo J.-X.; Liu X.; Li X.-H.; Zhou C.-X. Sesquiterpene lactones from the root tubers of lindera aggregata. J. Nat. Prod. 2009, 72, 1497–1501. 10.1021/np900354q. [DOI] [PubMed] [Google Scholar]
  44. Yoshikawa M.; Hatakeyama S.; Inoue Y.; Yamahara J. Saussureamines A, B, C, D, and E, new anti-ulcer principles from Chinese Saussureae Radix. Chem. Pharm. Bull. 1993, 41, 214–216. 10.1248/cpb.41.214. [DOI] [PubMed] [Google Scholar]
  45. Wang C.-K.; Lee W.-H.; Peng C.-H. Contents of phenolics and alkaloids in Areca catechu linn. During maturation. J. Agric. Food Chem. 1997, 45, 1185–1188. 10.1021/jf960547q. [DOI] [Google Scholar]
  46. Rosin M. P. The influence of PH on the convertogenic activity of plant phenolics. Mutat. Res. 1984, 135, 109–113. 10.1016/0165-1218(84)90163-0. [DOI] [PubMed] [Google Scholar]
  47. Xu L.; Han X.; Qi Y.; Xu Y.; Yin L.; Peng J.; Liu K.; Sun C. Multiple compounds determination and fingerprint analysis of Lidanpaishi tablet and keli by high-performance liquid chromatography. Anal. Chim. Acta 2009, 633, 136–148. 10.1016/j.aca.2008.11.043. [DOI] [PubMed] [Google Scholar]
  48. Dongyan H.; Weihua Z.; Ruihua H. Separation and determination of chemical constituents in the volatile oil of three traditional chinese crude drugs. J. Pharm. Biomed. Anal. 1998, 17, 1423–1426. 10.1016/S0731-7085(98)00031-4. [DOI] [PubMed] [Google Scholar]
  49. Perrino B. A. Calcium Sensitization Mechanisms in Gastrointestinal Smooth Muscles. J. Neurogastroenterol. Motil. 2016, 22, 213–225. 10.5056/jnm15186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Xiao Y.; He Z.; Long Y.; Chen W.; Chen D.; Chi R.; Ye H.; Deng X.; Lv B.; Sun C.; Hu B.; Nie Z.; Gu S.; Chen C. Simo decoction versus domperidone suspension for post-pyloric spiral nasoenteric tube placement: A multicenter, randomized, non-inferiority trial. Clin. Nutr. 2020, 39, 2406–2412. 10.1016/j.clnu.2019.11.009. [DOI] [PubMed] [Google Scholar]
  51. Törnblom H.; Lindberg G. A 21st century look at the spectrum of gastrointestinal motility disorders. What is dysmotility; what is functional?. Clin. Gastroenterol. 2011, 40, 715–723. 10.1016/j.gtc.2011.09.011. [DOI] [PubMed] [Google Scholar]
  52. Kusano M.; Hosaka H.; Kawada A.; Kuribayashi S.; Shimoyama Y.; Zai H.; Kawamura O.; Yamada M. Gastrointestinal motility and functional gastrointestinal diseases. Curr. Pharm. Des. 2014, 20, 2775–2782. 10.2174/13816128113199990572. [DOI] [PubMed] [Google Scholar]
  53. Bolton T. B.; Prestwich S. A.; Zholos A. V.; Gordienko D. V. Excitation-contraction coupling in gastrointestinal and other smooth muscles. Annu. Rev. Physiol. 1999, 61, 85–115. 10.1146/annurev.physiol.61.1.85. [DOI] [PubMed] [Google Scholar]
  54. De Ponti F.; Giaroni C.; Cosentino M.; Lecchini S.; Frigo G. Calcium-channel blockers and gastrointestinal motility: basic and clinical aspects. Pharmacol. Ther. 1993, 60, 121–148. 10.1016/0163-7258(93)90024-8. [DOI] [PubMed] [Google Scholar]
  55. Sanders K. M. Spontaneous Electrical Activity and Rhythmicity in Gastrointestinal Smooth Muscles. Adv. Exp. Med. Biol. 2019, 1124, 3–46. 10.1007/978-981-13-5895-1_1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Perrino B. A. Regulation of gastrointestinal motility by Ca2+/calmodulin-stimulated protein kinase II. Arch. Biochem. Biophys. 2011, 510, 174–181. 10.1016/j.abb.2011.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Kang J.-J.; Cheng Y.-W. Effects of boldine on mouse diaphragm and sarcoplasmic reticulum vesicles isolated from skeletal muscle. Planta Med. 1998, 64, 18–21. 10.1055/s-2006-957358. [DOI] [PubMed] [Google Scholar]
  58. Li C.-B.; Yang X.; Tang W.-B.; Liu C.-Y.; Xie D.-P. Arecoline excites the contraction of distal colonic smooth muscle strips in rats via the M3 receptor-extracellular Ca2+ influx - Ca2+ store release pathway. Can. J. Physiol. Pharmacol. 2010, 88, 439–447. 10.1139/Y10-024. [DOI] [PubMed] [Google Scholar]
  59. Kinsey W. H. Intersecting roles of protein tyrosine kinase and calcium signaling during fertilization. Cell Calcium 2013, 53, 32–40. 10.1016/j.ceca.2012.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Yamashita Y.; Hasegawa-Sasaki H.; Sasaki T. Suppression by staurosporine of Ca2+ -mobilization triggered by ligation of antigen-specific receptors on t and B lymphocytes. An essential role of protein tyrosine kinase in the signal transduction. FEBS Lett. 1991, 288, 46–50. 10.1016/0014-5793(91)81000-X. [DOI] [PubMed] [Google Scholar]
  61. Li W.; Liu X.; Yanoff M. Phosphatidylcholine hydrolysis and DNA synthesis in cultured retinal capillary pericytes. Microvasc. Res. 1995, 49, 350–363. 10.1006/mvre.1995.1030. [DOI] [PubMed] [Google Scholar]
  62. Ferriola P. C.; Cody V.; Middleton E. Jr. Protein kinase C inhibition by plant flavonoids. Kinetic mechanisms and structure-activity relationships. Biochem. Pharmacol. 1989, 38, 1617–1624. 10.1016/0006-2952(89)90309-2. [DOI] [PubMed] [Google Scholar]
  63. Hwang S.-L.; Lin J.-A.; Shih P.-H.; Yeh C.-T.; Yen G.-C. Pro-cellular survival and neuroprotection of citrus flavonoid: the actions of hesperetin in PC12 cells. Food Funct. 2012, 3, 1082–1090. 10.1039/c2fo30100h. [DOI] [PubMed] [Google Scholar]
  64. Zhuo R.-G.; Ma X.-Y.; Zhou P.-L.; Liu X.-Y.; Zhang K.; Wei X.-L.; Yan H.-T.; Xu J.-P.; Zheng J.-Q. Mas-related G protein-coupled receptor D is coupled to endogenous calcium-activated chloride channel in Xenopus oocytes. J. Physiol. Biochem. 2014, 70, 185–191. 10.1007/s13105-013-0292-1. [DOI] [PubMed] [Google Scholar]
  65. Ni J.; Guo Y.; Chang N.; Cheng D.; Yan M.; Jiang M.; Bai G. Effect of N-methyltyramine on the regulation of adrenergic receptors via enzymatic epinephrine synthesis for the treatment of gastrointestinal disorders. Biomed. Pharmacother. 2019, 111, 1393–1398. 10.1016/j.biopha.2018.12.145. [DOI] [PubMed] [Google Scholar]
  66. Yang Z.-H.; Yu H.-J.; Pan A.; Du J.-Y.; Ruan Y.-C.; Ko W.-H.; Chan H.-C.; Zhou W.-L. Cellular mechanisms underlying the laxative effect of flavonol naringenin on rat constipation model. PLoS One 2008, 3, e3348 10.1371/journal.pone.0003348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. de Pins B.; Montalban E.; Vanhoutte P.; Giralt A.; Girault J.-A. The non-receptor tyrosine kinase Pyk2 modulates acute locomotor effects of cocaine in D1 receptor-expressing neurons of the nucleus accumbens. Sci. Rep. 2020, 10, 6619. 10.1038/s41598-020-63426-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Schaller M. D. Cellular functions of FAK kinases: insight into molecular mechanisms and novel functions. J. Cell Sci. 2010, 123, 1007–1013. 10.1242/jcs.045112. [DOI] [PubMed] [Google Scholar]
  69. Zhang F.; Yang P.; He Q.; Dong X.; Zhang S. Is gastrointestinal motility related to alkaloids of Charred Semen Arecae?. J. Ethnopharmacol. 2020, 257, 112825. 10.1016/j.jep.2020.112825. [DOI] [PubMed] [Google Scholar]
  70. Guo H.; Zhang J.; Gao W.; Qu Z.; Liu C. Gastrointestinal effect of methanol extract of Radix Aucklandiae and selected active substances on the transit activity of rat isolated intestinal strips. Pharm. Biol. 2014, 52, 1141–1149. 10.3109/13880209.2013.879601. [DOI] [PubMed] [Google Scholar]
  71. Lou Z.; Wang J.; Chen Y.; Xu C.; Chen X.; Shao T.; Zhang K.; Pan H. Linderae radix ethanol extract attenuates alcoholic liver injury via attenuating inflammation and regulating gut microbiota in rats. Braz. J. Med. Biol. Res. 2019, 52, e7628 10.1590/1414-431x20197628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Wang Y.; Zhang S.; Li F.; Zhou Y.; Zhang Y.; Wang Z.; Zhang R.; Zhu J.; Ren Y.; Tan Y.; Qin C.; Li Y.; Li X.; Chen Y.; Zhu F. Therapeutic target database 2020: enriched resource for facilitating research and early development of targeted therapeutics. Nucleic Acids Res. 2020, 48, D1031–D1041. 10.1093/nar/gkz981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Wishart D. S.; Feunang Y. D.; Guo A. C.; Lo E. J.; Marcu A.; Grant J. R.; Sajed T.; Johnson D.; Li C.; Sayeeda Z.; Assempour N.; Iynkkaran I.; Liu Y.; Maciejewski A.; Gale N.; Wilson A.; Chin L.; Cummings R.; Le D.; Pon A.; Knox C.; Wilson M. DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Res. 2018, 46, D1074–D1082. 10.1093/nar/gkx1037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Hamosh A.; Scott A. F.; Amberger J. S.; Bocchini C. A.; McKusick V. A. Online Mendelian Inheritance in Man (OMIM), a knowledgebase of human genes and genetic disorders. Nucleic Acids Res. 2005, 33, D514–D517. 10.1093/nar/gki033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Jensen L. J.; Kuhn M.; Stark M.; Chaffron S.; Creevey C.; Muller J.; Doerks T.; Julien P.; Roth A.; Simonovic M.; Bork P.; von Mering C. STRING 8--a global view on proteins and their functional interactions in 630 organisms. Nucleic Acids Res. 2009, 37, D412–D416. 10.1093/nar/gkn760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. 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. 10.1101/gr.1239303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Luo Z.; Yu G.; Chen X.; Liu Y.; Zhou Y.; Wang G.; Shi Y. Integrated phytochemical analysis based on UHPLC-LTQ-Orbitrap and network pharmacology approaches to explore the potential mechanism of Lycium ruthenicum Murr. for ameliorating Alzheimer’s disease. Food Funct. 2020, 11, 1362–1372. 10.1039/c9fo02840d. [DOI] [PubMed] [Google Scholar]
  78. Dennis G. Jr.; Sherman B. T.; Hosack D. A.; Yang J.; Gao W.; Lane H. C.; Lempicki R. A. DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol. 2003, 4, P3. 10.1186/gb-2003-4-5-p3. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

ao0c05680_si_001.pdf (1.2MB, pdf)
ao0c05680_si_002.xlsx (278KB, xlsx)

Articles from ACS Omega are provided here courtesy of American Chemical Society

RESOURCES