Next Article in Journal
Effects of Medium Additives on the Mycelial Growth and Polysaccharide Biosynthesis in Submerged Culture of Bjerkandera fumosa
Previous Article in Journal
Urchin-like Ce(HCOO)3 Synthesized by a Microwave-Assisted Method and Its Application in an Asymmetric Supercapacitor
Previous Article in Special Issue
Comprehensive Analysis of the Effect of 20(R)-Ginsenoside Rg3 on Stroke Recovery in Rats via the Integrative miRNA–mRNA Regulatory Network
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Trollius chinensis Bunge: A Comprehensive Review of Research on Botany, Materia Medica, Ethnopharmacological Use, Phytochemistry, Pharmacology, and Quality Control

1
College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
2
College of Jiamusi, Heilongjiang University of Chinese Medicine, Jiamusi 154007, China
*
Authors to whom correspondence should be addressed.
These authors have contributed equally to this work.
Molecules 2024, 29(2), 421; https://doi.org/10.3390/molecules29020421
Submission received: 30 November 2023 / Revised: 11 January 2024 / Accepted: 13 January 2024 / Published: 15 January 2024
(This article belongs to the Collection Herbal Medicine Research)

Abstract

:
Trollius chinensis Bunge, a perennial herb belonging to the Ranunculaceae family, has been extensively used in traditional Chinese medicine. Documented in the Supplements to the Compendium of Materia Medica, its medicinal properties encompass a spectrum of applications, including heat clearance, detoxification, alleviation of oral/throat sores, earaches, eye pain, cold-induced fever, and vision improvement. Furthermore, T. chinensis is used in clinical settings to treat upper respiratory infections, pharyngitis, tonsillitis, esoenteritis, canker, bronchitis, etc. It is mainly used to treat inflammation, such as inflammation of the upper respiratory tract and nasal mucosa. This comprehensive review explores the evolving scientific understanding of T. chinensis, covering facets of botany, materia medica, ethnopharmacological use, phytochemistry, pharmacology, and quality control. In particular, the chemical constituents and pharmacological research are reviewed. Polyphenols, mainly flavonoids and phenolic acids, are highly abundant among T. chinensis and are responsible for antiviral, antimicrobial, and antioxidant activities. The flower additionally harbors trace amounts of volatile oil, polysaccharides, and other bioactive compounds. The active ingredients of the flower have fewer side effects, and it is used in children because of its minimal side effects, which has great research potential. These findings validate the traditional uses of T. chinensis and lay the groundwork for further scientific exploration. The sources utilized in this study encompass Web of Science, Pubmed, CNKI site, classic monographs, Chinese Pharmacopoeia, Chinese Medicine Dictionary, and doctoral and master’s theses.

Graphical Abstract

1. Introduction

Trollius chinensis Bunge, a perennial herb of the Ranunculaceae family, falls under the genus Trollius [1]. Widely distributed in Northern China, T. chinensis is recognized for its high ornamental and medicinal value [2]. Its dried flowers, known as Flos Trollii, serve as the medicinal component [3]. There are more than 20 identified species in the genus Trollius [4]. They are distributed mainly in the temperate and arctic regions of Asia, Europe, and North America, of which 16 are in China [5]. It usually grows in peatlands, swamps, wet meadows, and banks of reservoirs, as well as in mountain areas up to the alpine zone [6]. T. chinensis, with its significant ornamental and health-related compounds, is highly esteemed for applications in the food, medicine, and cosmetic industries [7]. Traditionally, the Chinese have employed T. chinensis for medicinal and tea purposes, dating back to the Qing Dynasty and recorded in Supplements to the Compendium of Materia Medica (Qing Dynasty) as “bitter in taste, cold in nature, non-toxic, mainly used for heat-clearing and detoxicating” [1,7]. It holds a prominent place in pharmacies, is frequently referenced in medical literature, and is listed in the Chinese Pharmacopoeia (Edition 2020) with five Chinese patent medicines.
Pharmacological tests have substantiated T. chinensis’s anti-inflammatory, anti-oxidant, anti-bacterial, and anti-viral properties, correlating closely with its chemical composition [8,9]. Over 100 compounds have been isolated from Trollius species, including flavonoids, organic acids, coumarins, alkaloids, terpenoids, and prenyl flavonoids in T. chinensis, boasting diverse biological activities [9,10,11]. To date, more than 100 compounds have been isolated from Trollius species. Phytochemical investigations of this plant have demonstrated the presence of flavonoids, organic acids, coumarins, alkaloids, terpenoids, and prenylflavonoids as main constituents of T. chinensis with diverse biological activities [12,13]. For instance, the flavonoid metabolite(s) Orientin and poncirin found in T. chinensis exhibited significant antiviral activity against parainfluenza type 3 (Para 3) [10]. Additionally, researchers have identified seventeen new labdane diterpenoid glycosides A–Q (1–17) in the dried flowers of T. chinensis, possessing therapeutic, antiviral, and antibacterial properties, establishing T. chinensis as a common anti-inflammatory drug and health tea [14,15]. The flowers have traditional uses in treating respiratory infections, pharyngitis, tonsillitis, and bronchitis in Chinese medicine [11]. The exploration of T. chinensis holds immense potential for novel medication research and therapeutic advancements [16]. This review article aims to provide comprehensive information and highlight the potential values associated with the development of T. chinensis.

2. Materials and Methods

Relevant literature was obtained from scientific databases such as TCMSP (https://old.tcmsp-e.com/tcmsp.php, accessed on 21 April 2023), Pubchem (https://pubchem.ncbi.nlm.nih.gov, accessed on 23 April 2023), Scientific Database of China Plant Species (http://db.kib.ac.cn, accessed on 10 April 2023), Google Scholar (https://xs.scqylaw.com, accessed on 5 April 2023), PubMed (https://pubmed.ncbi.nlm.nih.gov, accessed on 5 April 2023), Baidu Scholar (https://xueshu.baidu.com, accessed on 3 April 2023), Vip site (China Science and Technology Journal Database) (http://www.cqvip.com, accessed on 3 April 2023), and CNKI site (Chinese National Knowledge Infrastructure) (https://www.cnki.net, accessed on 3 April 2023). The most extensive collection of publicly available chemical data in the world is found on PubChem. Chemicals can be found using their names, structures, molecular formulas, and other identifiers. Discover information about biological activity, safety and toxicity, chemical and physical qualities, patents, literature citations, et al. The PubChem Compound, Substance, and Bioassay sub-databases are the three sub-databases that make up the PubChem database. TCMSP, which includes 499 Chinese herbal medicines, a total of 29,384 ingredients, 3311 targets, and 837 related diseases. TCMSP is a unique systematic pharmacology platform for Chinese herbal medicines, where we can find the relationship between drugs, targets, and diseases. This database platform provides information that includes identifying active ingredients, compounds, drug target networks, et al. [17]. The Database of China Plant Species is jointly constructed by the Kunming Institute of Botany, Chinese Academy of Sciences (KIB), the Institute of Botany, Chinese Academy of Sciences (IBS), the Wuhan Botanical Garden, Chinese Academy of Sciences (WBG), and the South China Botanical Garden, Chinese Academy of Sciences (SCBG). There are more than 31,000 species of higher plants in more than 3400 genera in more than 300 families, and the data content mainly includes standard names of plant species, basic information, systematic taxonomic information, ecological information, physiological and biochemical characteristic description information, habitat and distribution information, literature information, and other information.
TCMSP, Pubchem, and Web of Huayuan were used to find the chemical composition of T. chinensis. Most of the active components were obtained by searching for T. chinensis in TCMSP. Then, PubChem and Web of Huayuan were used to obtain and validate information related to the chemical structure of organic small molecules contained in the herb and their biological activities. The Web of China Plant Species Information Database is the primary source for the botanical collection of the genus Trollius. All the sites listed above are public databases and have access to the public database. The article is summarized using other websites that gather literature about the development of T. chinensis research. Diverse studies have been published in recent years. Therefore, a comprehensive review is necessary. This paper reviewed the research progress of T. chinensis from six aspects, including botany, materia medica, ethnopharmacological use, phytochemistry, pharmacology, and quality control, with the keywords of chemical constituents such as flavonoids, phenolic acids, anti-inflammatory effects, and antimicrobial effects, as well as related words such as pharmacological effects. We reviewed 350 related papers. This paper draws on over 120 articles on T. chinensis and documents some of the literature on chemical composition and pharmacological studies conducted from 1991 to 2023.

3. Botany

Based on the search results from the Chinese herbal medicine series of the Chinese herbal medicine resource dictionary [18], Flora of China (https://www.plantplus.cn/foc, accessed on 10 April 2023), Scientific Database of China Plant Species (DCP) (http://db.kib.ac.cn, accessed on 10 April 2023), and other websites, and complemented by an extensive array of references, the genus Trollius comprises 26 species, as detailed in Table 1.
T. chinensis, a perennial herb of medicinal significance, features dried flowers as its medicinal components [3,19].
The geographical distribution of T. chinensis mainly spans Asia, Europe, the temperate zones of North America, and the Arctic region. In China, it is located in Tibet, Yunnan, Sichuan, Qinghai, Xinjiang, Gansu, Shaanxi, Shanxi, Henan, Hebei, Liaoning, Jilin, Heilongjiang, Inner Mongolia, and Taiwan [4]. Additionally, it is prevalent in Russia (Far East, Siberia, and Central Asia), North Korea, Inner Mongolia, Sakhalin Island (Sakhalin Island), Nepal, and Northern Europe [20]. Thriving in light and moist conditions, T. chinensis flourishes best in deep, preferably heavy, and consistently moist soil, exhibiting resilience in full sun or partial shade. Typically growing at elevations between 1000 and 2000 m, it is frequently observed at approximately 1400 m in habitats with ample water and optimal light conditions, such as peatlands, marshes, wet meadows, reservoir banks, mountainous areas, and alpine areas (Figure 1) [21].
T. chinensis plants are glabrous, boasting columns reaching up to 70 cm in height (Figure 2). The stems, numbering 1–3, range from 3.5–100 cm tall, either unbranched or branched above the middle, with occasional basal or distal branching and sparse foliage featuring 2–4 leaves. Basal leaves, numbering 1–4, measure 16–36 cm in length and are characterized by long stalks, occasionally accompanied by 1–3 rosette leaves. The leaf blade is pentagonal, with dimensions of 3.8–12.5 cm, exhibiting a cordate, trilobated base; the petiole, measuring 12–30 cm, has a narrowly sheathed base. Cauline leaves mirror basal leaves, with lower leaves possessing long stalks and upper leaves being smaller, short-stalked, or sessile. The pedicel, mostly grey-green, extends 5–9 cm in length. Flowers appear solitarily terminal or in 2–3 cymes, with a diameter ranging from 3.8–5.5 cm. Sepals, numbering 6–19, measure 1.6–2.8 cm and exhibit varying colors among species, including pale purple, pale blue, white, golden yellow, yellow, or orange-yellow. The leaf blade is not green when dried and is isobovate or elliptic-obovate in shape. Petals, numbering 18–21, are narrowly linear, slightly longer than sepals or subequal to sepals apically attenuate, measuring 1.8–2.2 cm in length and 1.2–1.5 mm in width. Stamens, numerous and spirally arranged, range from 0.5–1.1 cm in length. Carpels, numbering 20–30, are sessile, and follicles are 1–1.2 cm in length and approximately 3 mm in width. Seeds are subobovoid, around 1–1.5 mm in length, black, and glossy. Flowering June–July, fruiting August–September [3,22,23].

4. Research on Materia Medica

T. chinensis has various nicknames. T. chinensis was recorded in the Annals of Shan Xi Traditional Chinese Medicine as Golden Pimple. It has been recorded in Wild Plants of Shan Xi under Asian T. chinensis. Tropaeolum majus, T. chinensis was recorded as a Supplement to the Compendium of Materia Medica (Thirty Years of Qianlong, 1765) by Shanxi Tong Zhi. Liao’s History is also recorded in the Annals of Wu Tai Mountain and the Sea of Humanity under Nasturtium. In Liao’s History Ying Wei Zhi, T. chinensis is recorded as T. chinensis, and The Book of Pictorial Guide of Chinese Plants calls it a globeflower [21,24]. T. chinensis was initially recognized as an ornamental plant. It was not until the Qing Dynasty that the medicinal value of T. chinensis was widely developed. The Record of Ennin’s Diary: The Record of a Pilgrimage to China in Search of the Law mentions that T. chinensis blooms in June and July [24]. After that, in the Yuan Dynasty, the poet Zhou Boqi used T. chinensis as the title of the Book of the Squire of Shangdu Poems, left heroic verses with the objects, and recorded the characteristics of the flowers of T. chinensis in the notes of the Book of the Squire of Shangdu Poems. In the Qing Dynasty, the origin of T. chinensis was recorded in the Shanxi Tong Zhi. In the Annals of Mount Wu Tai, under the name of nasturtium, T. chinensis was associated with miracles to record articles. The Widely Manual of Aromatic Plants describes the golden yellow color of the flower, seven petals, and two layers; the heart of the flower is also yellow; there are several flowers on one stem; and so on, describing in detail the flowering period, flowering characteristics, and other botanical characteristics of T. chinensis [19]. It appeared as a companion botanical drug to licorice in the description of licorice in the Bencao ZhengYao (Ming Dynasty, AD 1368–1644) but was not included in the book in its entirety [19]. T. chinensis’s medicinal functions were first recorded in Supplements to the Compendium of Materia Medica (Thirty Years of Qianlong, 1765) [22]. Modern character descriptions and fluorescence identification of T. chinensis have been included in the Chinese Pharmacopeia (1977 edition). T. chinensis is a traditional Mongolian medicine and not a widely used medicinal herb. Initially, its sources of medicinal herbs were mainly wild, and due to the lack of commercial supply, fewer applications, regional herbs, and relatively limited clinical applications and research, as well as the cold nature of T. chinensis, some potential safety and efficacy issues, and other factors, it has not been included in the Pharmacopoeia of China since 1985 [25]. The Chinese Pharmacopoeia (2020 edition) includes only five proprietary Chinese medicines: Jinlianhua Tablets, Jinlianhua Runhou Tablets, Jinlianhua Mixture, Jinlianhua Capsules, and Jinlianhua Granules [26].

5. Ethnopharmacological Use

5.1. Traditional Uses

T. chinensis serves as both a traditional Chinese medicine and a frequently used ethnomedicine. The herb can improve heat clearance, detoxification, alleviation of oral/throat soreness, earache, eye pain, cold-induced fever, and vision improvement [27]. Furthermore, it can effectively treat boils, poisons, and winds. The Shanhai Caozhuan briefly mentions T. chinensis as a remedy for boils, poisons, and all kinds of winds. Flowers are used in the Hebei Handbook of Traditional Chinese Medicine (1970) for chronic tonsillitis. T. chinensis is combined with Juhua and Guanaco, doubled in acute cases, or added with Yazhicao in equal parts. To treat acute otitis media, acute conjunctivitis, and other inflammatory diseases of the upper focus, T. chinensis and Juhua are each taken with three qian, and raw Gancao with one qian. Zhaobing Nan Fang records combining Nanshashen and Beishashen with 12 g of T. chinensis to promote yin and diminish fire, reducing spleen and kidney yin deficiency and inflammation caused by fire inadequacy. It is noted in the Manual of Chinese Herbal Medicine Commonly Used in Guangxi Folklore: Book I that T. chinensis has been utilized for alleviating eye inflammation and pain. Furthermore, T. chinensis, along with Wushuige and Mufurong, are recommended for treating malignant sores via compressing and pounding the affected site [19,28].

5.2. Current Use

In 2003, the Administration of Traditional Chinese Medicine of China announced a prescription for preventing atypical pneumonia. The prescription, T. chinensis Tang, combined six botanical drugs, including T. chinensis, to clear away heat, detoxify toxins, disperse wind, and penetrate evil spirits. This prescription had a significant effect on atypical pneumonia and is now commonly used to prevent and treat “plague”, such as the new coronavirus [19,29]. Its principal effects and clinical use for acute and chronic tonsillitis and other inflammatory conditions are recorded in the National Compendium of Chinese Herbal Medicine (1975). The pharmacological effects of T. chinensis are summarized in the Dictionary of Traditional Chinese Medicine (2006). To cope with the contemporary and rapidly changing lifestyle, the utilization of T. chinensis medicinal decoctions has diminished compared with previous times. Instead, they are now commonly consumed as patented medications—for example, Jinlianhua soft capsules and health products [30]. Moreover, the petals and stamens of T. chinensis are widely employed as a flavoring agent in culinary contexts, imparting a distinctive taste to salads, desserts, and beverages. Moreover, it can be used as a coloring agent, food additive, and dyeing agent [31]. It is also valued as an antioxidant component in cosmetics, including T. chinensis Pure Lotion and T. chinensis Spray. The ethnopharmacological uses of T. chinensis are shown in Table 2.

6. Phytochemistry

According to the search results of TCMSP (old.tcmsp-e.com/tcmsp.php, accessed on 21 April 2023), the Huayuan website (www.chemsrc.com, accessed on 23 April 2023), PubChem (https://pubchem.ncbi.nlm.nih.gov, accessed on 23 April 2023), and other websites combined with much of the literature review, the main components of T. chinensis include flavonoids, fatty acids, alkaloids, sterols, coumarins, tannins, and polysaccharides.

6.1. Flavonoids

Flavonoids stand out as the predominant bioactive metabolites within Trollius chinensis flowers. Numerous studies have substantiated the manifold advantageous biological properties of flavonoids, encompassing anti-oxidation, anti-inflammatory, anti-viral, and anti-tumor characteristics [37]. The flavonoids in T. chinensis consist primarily of flavone C-glycoside, flavone O-glycoside, dihydroflavone, and flavonols. Notably, flavone C-glycosides, predominantly hexose glycosides, exhibit unique stability due to a direct connection between the sugar group and the flavonoid parent nucleus via a c-c bond [4], forming a remarkably stable glycoside structure. The majority of flavone C-glycosides are situated at the flavone C-glycoside C-6 or C-8 positions, with a few occurring at the a-ring C-3 or C-4 positions. In T. chinensis, the flavone C-glycoside is positioned at the flavonoid A-ring C-8 positions [9]. Polyphenols, mainly flavonoids, including Orientin, Vitexin, and isoflavin, are highly abundant among T. chinensis and are responsible for antiviral, antimicrobial, and antioxidant activities. The flavone C-glycoside includes Orientin, Vitexin, and isodoxanthin. Notably, Orientin, Vitexin, and Orientin -2″-O-β-l-galactoside emerge as the most abundant flavonoids in T. chinensis. Vitexin and Orientin glycosyl exhibit robust inhibitory effects against influenza virus, Staphylococcus aureus, and epidermis [38]. In addition to flavone C-glycosides, flavone O-glycosides, such as Quercetin and Isoquercetin, are also discernible in T. chinensis. Noteworthy is the enhanced stability and reduced hydrolysis susceptibility of flavonoid carbosides like Orientin [39]. The therapeutic potential of these constituents extends to the treatment of age-related macular degeneration, cancer, cardiovascular disease, and skin repair following UV damage. Refer to Table 3 and Figure 3 for further details.

6.2. Organic Acids

The concentration of phenolic acids in T. chinensis surpasses only that of flavonoids. Specifically, Veratric acid stands out with a notably high concentration of 0.86–0.91 mg.g−1 [51]. Intriguingly, a distinct study revealed that the bioavailability of phenolic acid constituents in T. chinensis surpassed that of its flavonoid counterparts [52]. Organic acids in T. chinensis encompass both phenolic and fatty acids. Phenolic acids predominantly constitute derivatives of benzoic acid, further classified into two categories. The first category lacks a free hydroxyl group, including Veratric acid, benzonic acid, methyl veratrate, globeflower acid, etc. The second category possesses free hydroxyl groups, including vanillic acid, methyl-p-hydroxybenzoate, p-hydroxybenzonic acid, etc. [31]. T. chinensis houses a repertoire of 21 fatty acids, with saturated fatty acids as the primary components, and a total of 21 elements, constituting 57.95% of the detected substances. Palmitic acid and tetradecanoic acid exhibit relatively substantial content within saturated fatty acids. Additionally, nine types of unsaturated fatty acids comprise 30.35% of the total, featuring oleic acid, linoleic acid, palmitoleic acid, 3-(4-hydroxy-3-methoxybenzene) -2-acrylic acid, 3-(4-hydroxy-benzene) -2-acrylic acid, 4-phenyl-2-butenic acid, 3-phenyl-2-acrylic acid, (E) -11-eicosanoic acid, and (Z, Z, Z) -9, 12, 15-octadecanotrioleic acid [53].
Of significant note, three crucial phenolic acids—proglobeflowery acid (PA), globeflowery acid (GA), and trolloside (TS)—have been isolated from the flowers of T. chinensis. Pharmacological investigations have underscored their diverse biological activities, strongly correlated with the flower’s efficacy in treating respiratory infections, tonsillitis, bronchitis, and pharyngitis [54]. Refer to Table 4 and Figure 4 for detailed insights.

6.3. Alkaloids

Alkaloids, a prominent category of nitrogenous phytochemicals widely distributed in medicinal plants, stand out as crucial constituents in T. chinensis. The exploration of T. chinensis alkaloids remains limited, with only five of these compounds identified thus far. The principal pyrrolidine alkaloids include Senecionine and Integerrimine, the isoquinoline Trolline and Indole (R)-nitrile-methyl-3-hydroxy-oxyindole), and adenine [8,55,56,57,58]. Notably, Trolline emerges as the most abundant among these five ingredients [59]. Investigations indicate that T. chinensis flowers possess the highest total alkaloid content, while roots and branches exhibit the lowest concentrations. Among them, Trolline, an isoquinoline first discovered in T. chinensis, demonstrates significant antiviral and antibacterial activities. Refer to Table 5 and Figure 5 for detailed data.

6.4. Other Chemical Components

In addition to the aforementioned three primary active components, the flowers contain trace amounts of sterols, coumarins, tannins, and polysaccharides. Although these components exist in relatively low concentrations, their pharmacological effects are manifold, holding substantial potential for development. T. chinensis polysaccharides consist of neutral and acidic monosaccharides, predominantly comprising mannose (Man), rhamnose (Rha), galacturonic acid (GalA), glucose (Glu), galactose (Gal), arabinose (Ara), and fucose (Fuc) [60]. T. chinensis also harbors compounds like xantho-phyll-Epoxyde (C40H56O3) and trollixanthin (C40H56O3). The yellow pigment in T. chinensis, characterized as a fat-soluble pigment, exhibits remarkable stability under neutral and acidic conditions [61]. An undescribed phenolic glycoside, phenol A, isolated from T. chinensis flowers via spectroscopic methods, has revealed both its structural composition and pharmacological actions, including anti-inflammatory and antibacterial properties [17]. Furthermore, T. chinensis encompasses eight trace elements: Fe, Mg, Cu, Zn, Mn, Cr, Pb, and As. Research indicates minimal variations in Ca and Fe levels across T. chinensis from different regions, while more pronounced differences exist in Mn, Cu, and Zn levels [62,63]. For a comprehensive overview, consult Table 6 and Figure 6.

7. Pharmacological Effects

7.1. Antiviral Effect

A study exploring the antiviral properties of T. chinensis revealed that its five active components—Vitexin, Orientin, Trolline, Veratric acid, and Vitexin-2″-O-β-l-galorientin—exert their effects by modulating Toll-like receptors (a critical class of protein molecules associated with non-specific immunity/natural immunity). Specifically, the T. chinensis soft capsule demonstrated in vitro inhibition of human coronavirus OC43 replication, accomplished through the regulation of TLRs to suppress elevated expression of host cell cytokines such as IL-1B, IL-6, and IFN-a mRNA induced by viral infection. These findings substantiate the inhibitory mechanism of the T. chinensis soft capsule against the virus [65]. Examining 26 active components such as Rutin, Luteolin-7-O-glucoside, Kaempferol, Genistin, Apigenin, Scutellarin, Orientin, Daidzin, Vitexin, 3′-Hydroxy Puerarin, Puerarin, Daidzein, 3′-Methoxypuerarin, 2″-O-Beta-l-Galactoside, Rosmarinic acid, Progloboflowery acid, Caffeic acid, Protocatechuic acid, Ferulic acid, Veratric acid, Indirubin E, Oleracein E, Trollioside, Carbenoside I, 2″-O-(2‴-methyl butanol)isodangyloxanthin, 2″-O-(2‴-methylbutyryl) Vitexin, and glucose veratrate in T. chinensis, were observed to bind to the Mpro protein (2019-nCoV novel coronavirus pneumonia hydrolase Mpr0 protein) primarily through hydrogen bonds. This binding showcased Mpro protein-binding activity, affirming the potential of T. chinensis against novel coronaviruses [29]. Influencing pivotal anti-inflammatory and immunomodulatory targets, T. chinensis, when combined with multiple inflammatory and immunomodulatory pathways such as tumor necrosis factor-α (TNF-α), HIF-1, and Toll-like receptors (TLR), exhibits anti-influenza viral effects, particularly against influenza A [66]. The antiviral action of T. chinensis has been scrutinized through cyberpharmacology. While cyberpharmacological analyses offer valuable insights into pharmacological research, their reliance on network interactions between biomolecules and extensive databases introduces challenges related to data quality and reliability. Furthermore, the intricate nature of biological systems, limited experimental data, and the evolving understanding of drugs and targets require cautious consideration of credibility, necessitating further validation through pharmacological experiments [67].
Chicken embryos served as the medium for influenza virus cultivation, with the inhibitory effect of T. chinensis alcohol extract on viral proliferation in chicken embryo allantoic fluid evaluated through a chicken erythrocyte agglutination test. The results substantiated the direct inactivation of the influenza A virus by T. chinensis alcohol extract in vitro. In a parallel experiment involving influenza A virus inoculation into chicken embryos, the T. chinensis alcohol extract effectively curbed the proliferation of the virus within the embryos [68]. In a mouse model infected with influenza A (H1N1) virus, the study categorized the subjects into the control group, TGC group (T. chinensis crude extract gavage group), VI1~3 groups (virus infection model 1~3 groups), and VI + TGC 1~3 groups (treatment 1~3 groups), each comprising 10 mice. Notably, the aqueous extract of T. chinensis exhibited the potential to enhance the antiviral ability of mice. Subsequent comparative analyses validated the initial findings, establishing that aqueous extracts of T. chinensis augmented antiviral capacity in mice. Conversely, alcoholic extracts of T. chinensis directly deactivated the influenza A virus [69]. Furthermore, the aqueous extract of T. chinensis demonstrated potent inhibitory activity against the Cox B3 virus, achieving an inhibitory concentration of 0.318 mg/mL. The total flavonoids in this study displayed varying inhibitory activity against the respiratory syncytial virus, influenza A virus, and parainfluenza virus, with inhibitory concentrations of the viruses being 20.8 μg/mL and 11.7 μg/mL for Vitexin and Orientin, respectively [70]. Notably, 60% ethanolic extracts of T. chinensis and total flavonoids exhibited weak effects, with Protopanaxanthic acid among the organic acids demonstrating the weakest antiviral ability. While T. chinensis showed effectiveness against the influenza A virus, its impact on the influenza B virus was not significant [10,39,53]. Comparative assessments revealed that the alcoholic extract solution of T. chinensis soup displayed greater antiviral effects than the aqueous decoction of T. chinensis soup. Additionally, higher-purity T. chinensis soup extract exhibited a more robust inhibitory effect on the influenza virus. Specifically, 80% T. chinensis soup extract and secondary 95% T. chinensis soup extract demonstrated superior antiviral effects compared with 60% T. chinensis soup extract [71]. A study delved into the material basis of the UPLC-DAD-TOF/MS fingerprinting profile (ultra-performance liquid chromatography-tandem diode array detector-time-of-flight mass spectrometry) of T. chinensis, establishing its potential as the active agent against EV71 (enterovirus 71). The key active ingredients of T. chinensis in combating EV71 included Guaijaverin acid, an unidentified alkaloid, P-hydroxybenzene-malic acid, and 2″-O-acetyl Orientin [25]. In the broader context, T. chinensis flowers emerged as a valuable contributor to the anti-influenza virus activity of the overall formula, exhibiting relatively few side effects. The synergistic effect of T. chinensis, particularly in formulations like T. chinensis soup, has proven effective as a treatment for influenza virus [27,72].
In recapitulation, the findings indicate that the antiviral mechanism of T. chinensis predominantly revolves around impeding the virus-receptor binding process and restraining the cytokines/chemokines response. The unrefined flower extract derived from T. chinensis shields the host from inflammatory damage by intervening in the TLRs, encompassing TLR3, TLR4, and TLR7. This intervention leads to a reduction in the secretion of inflammatory factors, ultimately manifesting antiviral effects [73,74].

7.2. Antioxidant Effect

The varied pharmacological impacts of Orientin in T. chinensis, particularly its potent antioxidant effect, surpass those attributed to Vitexin. This discrepancy may be attributed to the structural disparity between Orientin and Vitexin. The oxidative activity of flavonoids with an o-diphenol hydroxyl group on the B-ring is notably more robust compared with those flavonoids possessing a singular phenol hydroxyl group attached to the B-ring [75].
To assess the antioxidant capacity of Orientin and Vitexin in T. chinensis concerning D-galactose-induced subacute senescence in mice, D-galactose was administered intraperitoneally [75]. The experimental outcomes revealed that Orientin and Bauhinia glycosides in T. chinensis effectively elevated the total antioxidant capacity (T-AOC), superoxide dismutase (SOD), glutathione peroxidase (GPGP), and glutathione peroxidase (GPP) in the tissues of the kidneys, livers, and brains of senescent mice. Additionally, these compounds increased SOD, glutathione peroxidase (GSH-Px), Na+-K+-ATPase, and Ca2+-Mg2+-ATPase activities in kidney, liver, and brain tissues. Notably, Orientin demonstrated superior efficacy over Oryza sativa in augmenting T-AOC activity within the organism [75]. The former mitigates impaired sodium ion transport and associated metabolic disorders [76], while the latter, elevated levels of Ca2+, adversely impact the cytoskeleton and membrane structure of neuronal cells, culminating in diminished stability and heightened membrane permeability, thereby contributing to the senescence process [16,75].
In contrast, the glycosides of Orientin and Vitexin pruriens act as antioxidants by positively modulating the activity of membrane transporter enzymes within tissue cells. Remarkably, Orientin exhibited greater efficacy than Vitexin in enhancing the activity of these tissue cell membrane transporter enzymes [15]. The robust antioxidant potential of Orientin, exceeding that of poncirin and further surpassing total flavonoids, has been corroborated in various studies. Both Orientin and Vitexin demonstrate the ability to scavenge superoxide anion, hydroxyl radical, and DPPH radical, effectively safeguarding the erythrocyte membrane. Specifically, Orientin displayed notable scavenging efficacy within the concentration range of 2.0–12.0 μg/mL. In contrast, Vitexin exhibited hydroxyl radical scavenging within the concentration range of 0–1.0 μg/mL, achieving maximum scavenging efficiency at 1.0 μg/mL, followed by a decline in scavenging effect with increasing Vitexin concentration [77].
The pharmacological mechanism underlying the antioxidant action of T. chinensis encompasses several key facets: (1) Scavenging of free radicals: The active constituents in T. chinensis, particularly flavonoids, exhibit potent free radical scavenging capabilities. This capacity enables the neutralization of free radicals both inside and outside the cell, thereby mitigating oxidative stress-induced damage [78]. (2) Stimulation of antioxidant enzyme activity: the active ingredients in T. chinensis stimulate the activity of antioxidant enzymes by stimulating the intracellular antioxidant enzymes such as superoxide dismutase, glutathione peroxidase, etc. [79]. This stimulation enhances the efficacy of the antioxidant system, fortifying cells against oxidative damage. In conclusion, T. chinensis safeguards cells from oxidative damage through the dual mechanisms of scavenging free radicals and enhancing antioxidant enzyme activity. These combined actions underscore the efficacy of T. chinensis as a potent antioxidant therapeutic agent.

7.3. Anti-Inflammatory Effect

The anti-inflammatory prowess of T. chinensis primarily targets the upper segment of the triple energizer, encompassing the area above the diaphragm within the human body. This region predominantly involves organs such as the stomach and throat, extending through the diaphragm and chest, including the heart, lungs, viscera, head, and face. Both the aqueous extract and 95% ethanol extracts of T. chinensis manifest robust anti-inflammatory activities. Notably, within the repertoire of compounds contained in T. chinensis, flavonoids such as Robinin, Quercetin, Vitexin, and Orientin exhibit heightened anti-inflammatory efficacy. Particularly, Vitexin and Orientin, due to their anti-inflammatory and soothing properties, along with peptide anti-histamine attributes, are deemed suitable for managing acute allergic skin conditions such as rash and eczema, as well as respiratory allergic diseases [80].
Current domestic research on T. chinensis underscores its potential in treating upper respiratory tract infectious diseases, including nasal mucosal diseases, by deploying an anti-inflammatory mechanism that engages multiple metabolites, targets, and pathways. Among the identified core targets, TNF and mitogen-activated protein kinase 1(MAPK1) take precedence, with the cancer factor pathway emerging as a pivotal route [81]. Additionally, Toll-like receptors 3, 4, and 7 (TLR3/4/7) have been proposed as promising common anti-inflammatory targets for T. chinensis constituents. This includes Vitexin, Orientin, Trolline, Veratric acid, and Vitexin-2″-O-galactoside, as discerned through the integration of network pharmacology and molecular docking techniques [82].
Respiratory inflammation, arising from diverse pathogens, microbial infections, influenza, nitroative stress, and compromised immune systems, can be effectively addressed by T. chinensis [83]. Its therapeutic spectrum extends beyond treating nasal mucosa inflammation to positively impacting upper respiratory infections. Leveraging data mining, an enriched analysis of the top 20 pathways linked to the targets and metabolites of T. chinensis in upper respiratory tract infection treatment identified quercetin as a highly probable compound. This conclusion was derived from the “metabolite-target-signaling pathway” network analysis [81]. Moreover, T. chinensis preparations exhibit therapeutic potential against upper respiratory tract infections by reducing serum inflammatory factors in patients. These factors include IL-8, IL-6, TNF-alpha, C-reactive protein, and procalcitonin, along with the modulation of T-cell subpopulation ratios [77,78,79,80]. Additionally, Orientin-2″-O-β-l-galactoside and Veratric acid have been identified for their anti-inflammatory effects [84]. In the clinical realm, the combination of amoxicillin, sodium, and potassium clavulanate has demonstrated the potential to reduce treatment duration and enhance therapeutic efficacy in children with acute tonsillitisn ratios [85,86,87,88].
In summary, T. chinensis harbors a repertoire of anti-inflammatory compounds, including Vitexin, Orientxin, Trolline, Veratric acid, and Vitexin-2″-O-galactoside. Notably, Quercetin may also contribute significantly to its anti-inflammatory activity [82,89]. Specifically, Orientin demonstrates efficacy in attenuating LPS-induced inflammation by impeding the production of inflammatory mediators and suppressing the expression of Cyclooxygenase 2 (COX-2) and Inducible nitric oxide synthase (iNOS) [90,91]. Vitexin-2″-O-galactoside exhibits substantial inhibitory effects on lipopolysaccharide (LPS)-induced inflammation, as evidenced by its impact on key factors such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), iNOS, and COX-2 expression. Additionally, it mitigates the production of reactive oxygen species and exerts an anti-neurogenic role by inhibiting the NF-κB and extracellular signal-regulated kinase (ERK) signaling pathways, leading to anti-neuroinflammatory activity. However, the pharmacological mechanisms underlying the anti-inflammatory effects of the other components remain elusive.

7.4. Antitumour

Flavonoids derived from T. chinensis exhibit notable inhibitory effects on active cancer cells. Specifically, the total flavonoids from T. chinensis demonstrate the capacity to impede the proliferation of tumor cells by activating the mitochondrial pathway [92]. T. chinensis extracts exerted strong inhibitory effects on Leukemia K562 cells (K562), and HeLT. chinensis extracts manifest robust inhibitory influences on various cancer cell lines, including Leukemia K562 cells (K562), HeLa cells (He La), esophageal cancer cellsEc-109 (Ec-109), lung cancer cells NCI-H446 (NCI-H446), human non-lung cancer cells NCI-H446 (NCI-H446), human non-small cell lung cancer cell line A549 (A549), and human carcinoma cells HT-29 (HT-29), MCF-7, and HepG2, among others [92]. Moreover, the total flavonoid extract of T. chinensis significantly retards the growth and proliferation of MCF-7 cells. This involvement is characterized by the activation of caspase-3 and caspase-9, leading to induced cell apoptosis within a concentration range of 0.0991 to 1.5856 mg/mL [93]. Non-alcoholic fatty liver disease (NAFLD) stands as a clinical pathologic syndrome [94,95], with its incidence in China reaching a significant 29.2%, demonstrating an annual increase [96]. The complex interplay of metabolic disorders, such as dyslipidemia, hypertension, hyperglycemia, and persistent abnormalities in liver function tests, is closely associated with NAFLD [97]. Elevated lipid levels induce expression changes in HepG2 cells (hepatoma cells) [98]. In an investigation into the impact of total flavonoids from T. chinensis on HepG2 cell function induced by high sugar levels, it was observed that oxidative stress levels in hepatocytes and the metabolic balance of reactive oxygen species (ROS) in HepG2 cells were intricately linked to intracellular fat accumulation. The study conclusively demonstrated that total flavonoids from T. chinensis exhibit a specific therapeutic effect on HepG2 cells by influencing disease-associated processes. Tissue cultures were employed to compare the effects of high glucose concentrations and varying doses of total flavonoids from T. chinensis on HepG2 cells. The proliferative tendencies of lipid substances are directly correlated with ROS levels; higher lipid accumulation corresponds to elevated ROS levels. Elevated glucose concentrations intensified ROS levels, while total flavonoids from T. chinensis effectively attenuated ROS levels, thereby influencing HepG2 cells. In vitro, total flavonoids from T. chinensis demonstrated a capacity to reduce lipid substance accumulation, presenting a promising avenue for the improved treatment of NAFLD [96].
The ethanol extract derived from the total flavonoids of T. chinensis has been observed to induce apoptosis in HT-2 cells through the endogenous mitochondrial pathway. In addition, specific constituents of T. chinensis, namely Orientin and Vitexin, have demonstrated inhibitory effects on human esophageal cancer EC-109 cells. The apoptotic induction of EC-109 cells by both Orientin and Vitexin was found to correlate with increased drug action time and elevated drug concentrations. Significantly, Orientin surpassed Vitexin in effectively inhibiting the growth and apoptosis of EC-109 cells [99]. At the administration dose of 80 μM, Orientin demonstrated a more potent apoptotic effect on EC-109 cells compared with Vitexin at the same concentration, registering apoptotic rates of 28.03% and 12.38%, respectively, within the concentration range of 0.91 to 1.5856 mg/mL.
Elucidating the pharmacological mechanism underlying Orientin’s action, specifically in the context of esophageal cancer cells (EC-109), involves the up-regulation of P53 expression and concomitant down-regulation of Bcl-2 expression. This dual modulation positions Orientin as a prospective therapeutic agent for esophageal cancer. Utilizing the total flavonoids of T. chinensis as a model drug, our exploration delved into the molecular-level relationship and mechanism of these flavonoids, shedding light on their antitumor activity. A pertinent discovery was that Orientin affected HeLa, augmenting the Bax/Bcl-2 protein ratio. This manifested as an increase in Bax protein levels coupled with a decrease in Bcl-2 protein levels, thereby triggering apoptotic protease activation. Consequently, this inhibition of HeLa cell proliferation underscores the therapeutic potential of Orientin in cervical cancer treatment.
While the notable anti-tumor activity of T. chinensis extract is evident, the specific mechanistic intricacies remain elusive. Putatively, this metabolite’s impact on the signaling pathways within tumor cells plays a pivotal role. T. chinensis is observed to down-regulate anti-apoptotic genes Bcl and Bcl-xL while concurrently up-regulating pro-apoptotic genes such as Bax, caspase-9, and caspase-3 at the mRNA levels. This concomitant suppression of COX-2 gene expression in tumor cells is linked to inhibiting the proliferation of diverse tumor cell lines. The inhibitory effect extends to the HT-29 of human colon cancer cells, with T. chinensis flavonoids proving efficacious in restraining cell proliferation. The concentration-dependent inhibition of human non-small cell lung cancer A549 cells, the induction of apoptosis in lung cancer A549 cells, and the anti-lung cancer role demonstrated by these flavonoids underscore their potential therapeutic relevance. Moreover, the ability of T. chinensis flavonoids to impede the progression of K562 cells, retaining them in the Go/G1 phase, elucidates their protective role against leukemia. Additionally, beyond the total flavonoid components, the total saponins of T. chinensis showcase robust antitumor activity, albeit without significant advantages over other pharmaceutical agents [100].

7.5. Antibacterial Effect

T. chinensis manifests broad-spectrum bacteriostatic activity against both Gram-positive cocci and Gram-negative Bacilli, including Pseudomonas aeruginosa, Staphylococcus aureus, Diplococcus pneumoniae, and Shigella dysenteriae. The pivotal antibacterial constituents of T. chinensis are its flavonoids, notably Orientin and Vitexin [101,102,103,104]. In vitro assessments utilized Minimum Inhibitory Concentration (MIC) and Minimum Bactericidal Concentration (MBC) as benchmarks for analyzing Escherichia coli, Salmonella, Staphylococcus aureus, Bacillus subtilis, Streptococcus mutans, Streptomyces, Rhodotorula, Aspergillus niger, and Candida albicans. The 30% ethanolic extract of T. chinensis exhibited notable antibacterial efficacy, particularly inhibiting Streptococcus mutans, suggesting a potential therapeutic avenue for dental caries. T. chinensis total flavonoids, along with Orientin and Vitexin, exhibited notable inhibitory effects on Gram-positive cocci while demonstrating no discernible impact on Gram-negative Bacilli and fungi. Their most pronounced inhibitory activity was observed against Staphylococcus aureus, with the order of inhibitory strength being Orientin = Total flavonoids > Vitexin. Specifically, the lowest inhibitory and bactericidal concentrations were determined to be 0.15625 mg·mL−1 and 0.625 mg·mL−1 for Orientin and total flavonoids, respectively. Additionally, these components demonstrated considerable inhibitory activity against Streptococcus mutans, with the antibacterial efficacy ranking as Orientin > Total flavonoids > Vitexin. Notably, the lowest inhibitory concentration and bactericidal concentration of Orientin were 0.15625 mg·mL−1 and 0.625 mg·mL−1, surpassing the efficacy of Vitexin [15]. In investigations exploring the bacteriostatic activity of various T. chinensis preparations, the Staphylococcus aureus solution clarified at concentrations of 225 mg/mL for Jinlianhua Tablets, 56.25 mg/mL for Jinlianhua Jiaonang, 450 mg/mL for Jinlianhua Granules, and 56.25 mg/mL for T. chinensis oral solution. For Bacillus subtilis, clarification occurred at concentrations of 56.25 mg/mL for Jinlianhua tablets, 14.0625 mg/mL for T. chinensis capsule, 225 mg/mL for T. chinensis granules, and 28.125 mg/mL for T. chinensis oral solution. Notably, the T. chinensis oral solution displayed no inhibitory effect against Escherichia coli. These experiments revealed that the antibacterial activities of the four T. chinensis preparations followed the order of strength as Bacillus subtilis > Staphylococcus aureus > Escherichia coli, with varying minimum inhibitory concentrations (MICs) against Staphylococcus aureus and Bacillus subtilis for different T. chinensis preparations, ranked from strongest to weakest as Jinlianhua capsules, Jinlianhua mixture, Jinlianhua tablets, and Jinlianhua granules [105]. In the in vitro bacteriostatic efficacy assessment, the total flavonoids extracted from T. chinensis exhibited robust inhibitory effects against common pathogenic organisms, including Staphylococcus epidermidis, Staphylococcus aureus, Escherichia coli, Streptococcus viridans, Salmonella paratyphi A, and Salmonella paratyphi B. Notably, the total demonstrated considerable protective effects in Staphylococcus aureus-infected mice, showcasing a dose-dependent reduction in the 48-h mortality of the infected mice [106]. The yellow pigment of T. chinensis, composed of xantho-phyll epoxyde and trollixanthin, also displayed bacteriostatic properties, with varying degrees of inhibition against Staphylococcus aureus, Bacillus subtilis, and Escherichia coli, showing increased activity with escalating concentrations. Tecomin, a glucose ester of Veratric acid, exhibited effective inhibition against Staphylococcus aureus and Pseudomonas aeruginosa, with MICs of 0.256 and 0.128 mg/mL, respectively [107]. Progloboflowery acid has emerged as an effective treatment for Pseudomonas aeruginosa-induced inflammatory skin reactions. Inhibitory effects were observed for proglobeflowery acid, Vitexin, and Orientin against Bacillus subtilis, Staphylococcus epidermidis, Staphylococcus aureus, and Micrococcus luteus. T. chinensis total flavonoids, Vitexin, Orientin, and proglobeflowery acid displayed inhibitory effects on Staphylococcus aureus and Staphylococcus epidermidis, with MICs of 50 and 25 μg/mL, 100 and 25 μg/mL, 25 and 25 μg/mL, and 200 and 200 μg/mL. For Micrococcus luteus and Bacillus subtilis, the MICs were higher than 200 μg/mL [108]. In the investigation, T. chinensis extract and its three metabolites exhibited potent inhibitory effects on four Gram-positive cocci. Total flavonoids and Vitexin, having the highest content, demonstrated strong inhibition, especially Orientin, against Staphylococcus aureus and Staphylococcus epidermidis, while PA demonstrated relatively weak inhibition against these two bacteria [100,106]. The study further revealed that PA had robust inhibitory action against Pseudomonas aeruginosa and Staphylococcus aureus, with MIC rates of 16 and 200 mg/L, respectively. Additionally, PA exhibited modest antiviral activity (IC50 of 184.2 μg/mL) against Para 3. Conversely, GA displayed significant antiviral efficacy against influenza A, as evidenced by its IC50 value of 42.1 μg/mL. With a MIC rate of 128 mg/L, TS demonstrated moderate inhibitory activity against Streptococcus pneumonia [43].
The antibacterial pharmacological mechanism underlying the action of T. chinensis predominantly revolves around impeding regular bacterial growth processes. This is accomplished by elevating extracellular nucleic acid and soluble protein levels within bacteria. The resultant damage to the cell membrane influences membrane permeability, inducing the efflux of vital metabolic substances crucial for cellular viability or the influx of detrimental medicinal fluids. Such interactions significantly impact bacterial growth, thereby realizing the intended inhibitory effects. The drug concentration exhibits a positive correlation with both the rate of inhibition of bacterial growth and the rate of inhibition of biofilm formation [105,109].

7.6. Others

The main active components of T. chinensis, total flavonoids, also have analgesic and antipyretic effects. Studies have shown that flavonoids can significantly reduce ET (the lipid and polysaccharide substances produced by the cell wall of G-bacteria-ET are a standard model for screening antipyretic drugs and exploring antipyretic mechanisms). Total flavonoids can also reduce the contents of endogenous heat sources TNF-α and IL-1β in the serum of febrile rabbits and then inhibit the production and release of PGE2 in the cerebrospinal fluid of rabbits by inhibiting the production or release of TNF-α and IL-1β induced by ET to reduce fever, increase heat loss, and restore body temperature to normal. Reducing the production of endogenous pyrogens such as IL-1 and TNF-α is the pharmacological basis of the antipyretic effect of total flavonoids [110].
The experiment was divided into two parts: the blank group, the positive group, the water extract from stem and leaf (low), the water extract from stem and leaf (high), the alcohol extract from stem and leaf (low), and the alcohol extract from stem and leaf (high). The control group was used to investigate the anti-inflammatory effect of T. chinensis. A part of the control group was divided into the blank group (distilled water 20 mL/kg), positive group (100 mg/kg), low (12 g/kg), and high (24 g/kg) water extract groups, and low (12 g/kg) and high (24 g/kg) alcohol extract groups as the control group to verify the analgesic effect of T. chinensis. The extracts of T. chinensis stem and leaf have anti-inflammatory and analgesic effects [111]. One study further investigated the antitussive, anti-inflammatory, and analgesic effects of T. chinensis [112]. The study showed that all dose groups of total flavonoid extract of T. chinensis could significantly prolong the therapeutic effect of antitussive; with the increase in total flavonoid extract dose, the incubation period of cough in mice was prolonged, and the number of coughs was reduced—the more significant the tracheal phenol red excretion, the more pronounced the antitussive and expectorant effects. The antitussive and expectorant effects were more evident in the high-dose group of total flavone extract of T. chinensis than in patent medicine cough syrup. In addition, the high-dose group treated with the whole flavonoid extract of T. chinensis showed significantly reduced ear swelling caused by xylenes, reduced reaction times, and an improved hot plate pain threshold.
Moreover, statistical analysis showed that the total flavonoid extract of this drug had effects similar to those of nonsteroidal anti-inflammatory drugs commonly used in clinics. It was confirmed that the total flavone extract had sound anti-inflammatory and analgesic effects and that the total flavones of T. chinensis were helpful in myocardial ischemia-reperfusion injury. Experimental studies have shown that its mechanism of action is to inhibit the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px), reduce the content of malondialdehyde (MDA), reduce the area of myocardial infarction, inhibit the release of myocardial enzymes, reduce the apoptosis of myocardial cells, and play a corresponding therapeutic and relieving role [113]. In addition, Orientin and Vitexin in T. chinensis could improve membrane transport in d-galactose-induced aging mice, which may be helpful for clinical applications in treating acute respiratory distress syndrome [102,114] (Table 7, Figure 7).

8. Quality Control

8.1. Analysis Methods

Currently, the market for Chinese herbal medicine T. chinensis has not been unified into varieties, in addition to Trollius chinensis Bunge. as the primary source of medicinal botanical drugs, Trollius ledebourii Reichenbach. Trollius macropetalus Fr. et al. have also done more research on resource exploitation and utilization for medicinal use. Hence, the quality of T. chinensis on the market is confusing, and it is difficult to distinguish the good from the bad. The 1977 edition of the Chinese Pharmacopoeia analyzes the quality of botanical drugs from two perspectives: physical identification and chemical identification. The 1998 edition of the Beijing Standards for Chinese Materia Medica (1998) also includes a microscopic identification method for determining authenticity. The 2019 edition of the Anhui Provincial Standard for the Preparation of Chinese Medicinal Tablets (2019) records the method of identification by thin-layer chromatography, in which the chromatograms of the test article obtained by experimental treatment and the chromatogram of the control botanical drug show spots of the same color at the corresponding positions of the thin-layer plate. The evaluation method in the 2018 edition of the Hubei Quality Standard for Traditional Chinese Medicinal Materials (2018) specifies that the moisture content of T. chinensis should not exceed 13.0%. The total ash content should not exceed 9.0%. The leachate content shall not be less than 35.0%. The content of Orientin (C21H20O11) must not be less than 1.0% when measured by high-performance liquid chromatography and calculated on the dry product. In addition to the identification methods recorded in pharmacopeia and local standards, the fluorescence reaction identification method, micro-sublimation test, FTIR identification, and DNA barcode molecular identification method of Chinese herbal medicines can also be used to identify the authenticity of T. chinensis. A micro-sublimation test can be seen on the slide of yellowish snow-like crystals [122]. The FTIR profile of T. chinensis was obtained by using FTIR identification, and the differences in peak shape, peak position, and peak intensity of the peaks in the profile can elucidate the differences in the components, compositions, and ratios of T. chinensis botanical drugs extracted from different origins, habitats, varieties, growth years, and different drying methods and extraction solvents to carry out a more accurate quality analysis to determine the authenticity of T. chinensis [123,124,125]. DNA barcode molecular identification of Chinese herbal medicines is a method to identify herbal medicines through the study of the polymorphism of the genetic material of Chinese herbal medicines, which can quickly identify the species [126]. At present, with the rapid development of molecular identification technology and in-depth plant genetic information mining, molecular identification methods in the standardization of traditional Chinese medicine identification have been widely used. For example, the early DNA molecular identification technique of T. chinensis, random amplified polymorphic DNA labeling (RAPD), was used to identify T. chinensis by observing the electrophoretic results of the DNA bands by PCR amplification, and the samples of T. chinensis could be classified according to their origins by using the RAPD technique [101]. The DNA barcode identification method of T. chinensis was established by using ITS2 sequences, and the neighbor-joining (NJ) phylogenetic tree was constructed to accurately identify T. chinensis, Trollius lilacinus Bunge, and Artemisia annua L. In addition, high-performance liquid chromatography (HPLC) coupled with mass spectrometry (MS) can be used to identify the chemical composition and characteristics of TCM. Based on the different information of protein bands of different varieties as the basis for the identification of TCM with protein as the informative substance, it is observed that the protein bands of different varieties of T. chinensis differ significantly in the number of bands, levels, and distribution [127]. In addition to the above methods, X-ray diffraction and X-ray fluorescence analysis can also be used to identify the grain characteristics of T. chinensis and establish a primary X-ray diffraction database for rapid identification of the authenticity of T. chinensis and its powder [128].

8.2. Quality Evaluation Method

To ensure the quality and therapeutic efficacy of T. chinensis, the key to quality control of the active ingredients is also to establish quality analysis methods. The quality of T. chinensis can be identified and evaluated through the establishment of content determination standards, the use of fingerprinting evaluation methods, and other methods that can provide reference for the further development and utilization of T. chinensis. At present, the quality evaluation method of T. chinensis is mainly based on chemical content determination, i.e., HPLC fingerprinting, with Orientin and Vitexin as the index components of the method [129,130]. In some studies, these two metabolites are combined with phenolic acid or alkaloid and other metabolites as quality evaluation indexes to improve the comprehensiveness of evaluation, and HPLC is the main evaluation method at present [8,39,59,131].

9. Conclusions and Future Perspectives

Based on ancient texts and modern research, this paper reviews the herbal testimonies, traditional uses, phytochemistry, pharmacological activities, and quality standards of T. chinensis to provide new ideas for future research on T. chinensis. According to ancient texts, T. chinensis can reduce inflammation, eliminate heat and toxins, and enhance visual clarity. It is particularly effective in managing sore throats, swollen gums, and oral gingival pain caused by heat. Based on recent phytochemical and pharmacological studies, T. chinensis possesses anti-inflammatory, antiviral, antitumor, antibacterial, and antimicrobial effects, which are especially good for treating virus-induced colds and various types of inflammation, such as respiratory inflammation. It was initially recorded as an ornamental plant in various ancient books. Since its initial inclusion in the Compendium of Materia Medica as a traditional Chinese medicine in 1765 during the Qing Dynasty, T. chinensis has been widely developed for its medicinal properties and employed in health care products and various dosage forms following current processing technology. Over 180 compounds from T. chinensis have been isolated and identified. The main active components of T. chinensis are flavonoids, alkaloids, and organic acids. Objective evaluations are emphasized in recent studies of T. chinensis, where the focus is mainly on the flavonoids Orientin and Vitexin. These two compounds are the most important and representative of T. chinensis, with less research on the other active components. Various domestic and international investigations indicate that flavonoids account for most of the pharmacological effects of T. chinensis.
First of all, regarding the medicinal employment of T. chinensis, historical records specify that its dried flower is the primary constituent. Additionally, contemporary experimental research concentrates on the flower of T. chinensis; however, chemical makeup and pharmacology evaluations of its roots, stems, and leaves are limited. Moreover, most research on the phytochemical metabolites of T. chinensis concentrates on crude extracts and flavonoids, including Vitexin, Orientin, and Orientin-2″-O-β-l-galactopyranoside. However, there is a lack of studies on the alkaloids and organic acids present in T. chinensis, with only a limited number of articles on this topic.
Second, studies have shown that both crude extracts and active constituents of T. chinensis have a wide range of pharmacological activities, and these modern pharmacological studies support most of the traditional uses of T. chinensis as a folk medicine. However, there is still a gap in the systematic research on T. chinensis. Many pharmacological studies on its crude extracts or active constituents are not in-depth enough, and fewer in vitro experiments exist. These pharmacological activities must be further confirmed by in vivo animal experiments and combined with clinical applications. This direction will provide a solid foundation for developing novel drug-lead compounds. For example, relevant animal experiments did not verify the antitumor effect of T. chinensis.
Third, most studies on the pharmacological activities of T. chinensis have focused on uncharacterized crude extracts, making it difficult to clarify the link between the isolated compounds and their biological activities. Systematic pharmacological studies on compounds isolated from T. chinensis are considerable. In addition, many pharmacological activities of crude extracts or compounds of T. chinensis, such as the anti-inflammatory pharmacological effects of T. chinensis, are currently focused on network pharmacology and molecular docking techniques, with only very few relevant in vitro experiments for further validation, and the exact mechanism of the inhibitory activity is still unclear; therefore, further studies to reveal better the precise molecular mechanism of the pharmacological activity of the drug appear to be necessary.
Fourth, in some ancient texts, T. chinensis was used with other botanical drugs, thereby treating chronic inflammation. However, almost no studies have been carried out to investigate the formulae of T. chinensis or to reveal the effects of synergistic or antagonistic actions. The area of this piece is almost blank. Therefore, drug interactions between certain botanical drugs and T. chinensis seem to be a new direction worth further exploration.
Fifth, T. chinensis was included in the 1977 edition of the Chinese Pharmacopoeia, but this variety was not included in the 1985–2020 edition. Although this paper summarizes the identification methods of T. chinensis in other pharmacopeias, the provisions on authenticity identification and quality evaluation methods of T. chinensis are not comprehensive compared with other Chinese medicinal materials. For example, Trollius ledebourii Rchb. It is an alternative source of T. chinensis. However, the different base plants of T. chinensis have not been included in the pharmacopeia like other Chinese botanical drugs, which limits the further development and utilization of T. chinensis. In addition, although other plants of the same genus have been used as substitutes for T. chinensis in some places, there is no unified standard in the market for evaluation, confusing product types, specifications, and grades of Chinese medicinal materials in the medicinal materials market, which easily leads to problems in efficacy and safety. At present, the commonly used identification methods for T. chinensis are different. Microscopic identification and character identification make it difficult to distinguish the difference between T. chinensis and different species of T. chinensis. Molecular identification technology still needs to be further improved, and new DNA molecular marker technology must be developed. By analyzing and comparing the ribosomal DNA of biological species, species identification methods such as ITS barcode technology still need to collect more T. chinensis from different places and species to improve relevant studies and further verify the applicability of this method.
In summary, T. chinensis serves not only as an ornamental plant and a tea source but also as a significant medicinal and food crop, possessing wide-ranging pharmacological and nutritional value. Nonetheless, more in-depth and comprehensive clinical utility studies are needed to establish the plant’s safety and effectiveness. Various compounds have been identified in T. chinensis, although the work done so far has been insufficient. Furthermore, additional research is necessary to determine the precise molecular mechanisms of these active ingredients in specific diseases. Future investigations should emphasize active metabolites other than flavonoids to uncover novel compounds and pharmacological effects. Thus, systematic studies on the phytochemistry and bioactivity of T. chinensis are essential for future research endeavors. This review is intended to serve as a valuable reference for developing and applying T. chinensis.

Author Contributions

L.H.: Writing—review and editing; Writing—original draft. Z.W.: Writing—original draft, sorted the references. J.L.: Writing—original draft, edited the paper’s language. C.Q.: Revise the first draft and draw the chemical composition map. J.H.: Writing—original draft. W.R.: Writing—review and editing; conceived and designed the structure of the review. X.L.: Writing—review and editing of the final draft. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Key Research and Development Program of China grant number 2021YFD1600901. This research was funded by a talent training project supported by the central government for the reform and development of local colleges and universities grant number ZYRCB2021008. This research was funded by the Heilongjiang Touyan Innovation Team Program grant number HLJTYTP2019001. And The APC was funded by Xiubo Liu.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No data were used for the research described in the article.

Conflicts of Interest

We declare that we have no financial and personal relationships with other individuals or organizations that could inappropriately influence our work, and no professional or other personal interests of any nature or kind in any product, service, and/or company that could be interpreted to influence the position presented in the paper or the review of the paper. In addition, all authors have read the manuscript and approved its submission to the Journal of Ethnopharmacology. The author has no conflict of interest.

Glossary

SARS-CoVSevere acute respiratory syndrome coronavirus
ACE2Angiotensin-converting enzyme II
IL-6Interleukin-6
OC43HCoV-OC43
HIF-1Hypoxia-inducible factor
UPLC-DAD-TOF/MSUltra-performance liquid chromatography-tandem diode array detector-time-of-flight mass spectrometry
TNFTumor necrosis factor
MAPK1 Mitogen-activated protein kinase 1
NAFLDNon-alcoholic fatty liver disease
CCK-8Cell Counting Kit-8
Annexin-FITC/PI-FCMAnnexinV-FITCApoptosisDetectionKit
PGE2Prostaglandin(PG) E2
GSH-Pxglutathione peroxidase
RAW264.7mononuclear macrophage leukemia
MICMinimal Inhibitory Concentration
TCMTraditional Chinese Medicine
iNOSInducible nitric oxide synthase
IL-1βinterleukin-1β
ERKextracellular signal-regulated kinase
K562Leukemia K562 cells
A549Human non-small cell lung cancer cells A549
MBCMinimum Bactericidal Concentration
A549Human non-small cell lung cancer cells A549
GAGlobeflowery acid
COVID-19Corona Virus Disease 2019
TMPRSS2Transmembrane serine two protease
IFN-αInterferon-α
TNF-αTumor necrosis factor alpha
TLRToll-like receptors
EV71Enterovirus 71
ROSReactive oxygen species
HepG2Human hepatocellular carcinomas
EC-109Human esophageal cancer
Hoechest33258BisBenzimide H 33258
ETGram-negative bacteria ET
SODSuperoxide dismutase
MDAmalondialdehyde
LPSlipopolysaccharide
MCF-7Human Breast Cancer
COX-2Cyclooxygenase 2
NF-κBnuclear factor-κB
NLRP3nucleotide-bindingdomain-(NOD-)like receptor protein 3
He LaHeLa cells
HT-29Human Carcinoma Cells HT-29
NCI-H446Lung cancer cells NCI-H446
PAProglobeflowery acid
TSTrolloside

References

  1. Liang, Y.; Liu, X.; Hu, J.; Huang, S.; Ma, X.; Liu, X.; Wang, R.; Hu, X. The Crude Extract from the Flowers of Trollius chinensis Bunge Exerts Anti-Influenza Virus Effects through Modulation of the TLR3 Signaling Pathway. J. Ethnopharmacol. 2023, 300, 115743. [Google Scholar] [CrossRef]
  2. Hou, R.; Yang, L.; Wuyun, T.; Chen, S.; Zhang, L. Genes Related to Osmoregulation and Antioxidation Play Important Roles in the Response of Trollius chinensis Seedlings to Saline-Alkali Stress. Front. Plant Sci. 2023, 14, 1080504. [Google Scholar] [CrossRef]
  3. Lei, R.; Feng, L.; Liu, Y.; Duan, J. Research progress of Trollius chinensis. J. Chin. Med. Mater. 2015, 38, 1085–1091. [Google Scholar]
  4. Li, H.; Fan, R.; Zhao, J.; Su, L. Research situation of Trollius plants. Chin. J. Exp. Tradit. Med. Formulae 2020, 26, 239–250. [Google Scholar]
  5. Li, J.; Du, Y.; Xie, L.; Jin, X.; Zhang, Z.; Yang, M. Comparative Plastome Genomics and Phylogenetic Relationships of the Genus Trollius. Front. Plant Sci. 2023, 14, 1293091. [Google Scholar] [CrossRef]
  6. Witkowska-Banaszczak, E. The Genus Trollius—Review of Pharmacological and Chemical Research. Phytother. Res. 2015, 29, 475–500. [Google Scholar] [CrossRef]
  7. Liu, Y.; Ding, X.; Zheng, L.; Zhang, Y.; Zhou, H. Herbal Drink Formulation Optimization of Trollius chinensis Bunge by Sensory Fuzzy Comprehensive Evaluation. Acta Sci. Pol. Technol. Aliment. 2020, 19, 185–194. [Google Scholar]
  8. Cai, H.; Liu, H.; Zheng, G.; Zhan, Z.; Hu, J. Research progress on chemical composition and pharmacological activity of Trollius chinensis Bunge. World Sci. Technol.-Mod. Tradit. Chin. Med. 2021, 23, 2340–2352. [Google Scholar]
  9. Yuan, M.; Wang, R.; Wu, X.; An, Y.; Yang, X. Investigation on Flos Trollii: Constituents and Bioactivities. Chin. J. Nat. Med. 2013, 11, 449–455. [Google Scholar] [CrossRef]
  10. Li, Y.-L.; Ma, S.-C.; Yang, Y.-T.; Ye, S.-M.; But, P.P.-H. Antiviral Activities of Flavonoids and Organic Acid from Trollius chinensis Bunge. J. Ethnopharmacol. 2002, 79, 365–368. [Google Scholar] [CrossRef] [PubMed]
  11. Fan, R.; Liu, R.; Zhang, T.; Wu, T. A New Natural Ceramide from Trollius chinensis Bunge. Molecules 2010, 15, 7467–7471. [Google Scholar]
  12. Liu, J.-Y.; Li, S.-Y.; Feng, J.-Y.; Sun, Y.; Cai, J.-N.; Sun, X.-F.; Yang, S.-L. Flavone C-Glycosides from the Flowers of Trollius chinensis and Their Anti-Complementary Activity. J. Asian Nat. Prod. Res. 2013, 15, 325–331. [Google Scholar] [CrossRef]
  13. Tian, H.; Zhou, Z.; Shui, G.; Lam, S.M. Extensive Profiling of Polyphenols from Two Trollius Species Using a Combination of Untargeted and Targeted Approaches. Metabolites 2020, 10, 119. [Google Scholar] [CrossRef] [PubMed]
  14. Guo, M.-L.; Xu, H.-T.; Yang, J.-J.; Chou, G.-X. Diterpenoid Glycosides from the Flower of Trollius chinensis Bunge and Their Nitric Oxide Inhibitory Activities. Bioorg. Chem. 2021, 116, 105312. [Google Scholar] [CrossRef] [PubMed]
  15. Liu, Y.; Liu, C.; Li, X.; Kang, Y.; Chen, Y.; Guo, S.; Li, Y.; Zhang, J.; Liu, S.; Xue, T.; et al. Study on antibacterial and antioxidant activities of flavonoids from Trollius chinensis Bge. J. Pharm. Res. 2023, 42, 228–231+242. [Google Scholar]
  16. Sun, P.; Li, X.; Xue, T.; Xin, J.; Chen, Y.; Guo, S.; Zhang, B. Progress of pharmacological effects and clinical application of goldenseal flower (Rosa canina). China Pharm. 2022, 33, 507–512. [Google Scholar]
  17. Ru, J.; Li, P.; Wang, J.; Zhou, W.; Li, B.; Huang, C.; Li, P.; Guo, Z.; Tao, W.; Yang, Y.; et al. TCMSP: A Database of Systems Pharmacology for Drug Discovery from Herbal Medicines. J. Cheminform. 2014, 6, 13. [Google Scholar] [CrossRef]
  18. Wu, Q.; Li, P. Chinese Herbal Medicine; Series of Chinese Herbal Medicine Resource Dictionary; China Medical Science and Technology Press: Beijing, China, 2020. [Google Scholar]
  19. Huang, J.; Feng, J.; Liu, Z.; Su, L.; Fan, R. Decipherment of ancient literature about Trollius chinensis. J. Liaoning Univ. Tradit. Chin. Med. 2023, 25. [Google Scholar]
  20. Li, L. The Geographical Distribution of Subfam. Helleboroideae (Ranunculaceae). J. Syst. Evol. 1995, 33, 537–555. [Google Scholar]
  21. Zhu, D.; Ding, W.; Chen, S. Research progress of Trollius plants. World Sci. Technol./Mod. Tradit. Chin. Med. Mater. Medica 2006, 8, 26–33. [Google Scholar]
  22. Zhao, X. Supplement to Compendium of Materia Medica; China Press of Traditional Chinese Medicine Co., Ltd.: Beijing, China, 1998; ISBN 978-7-80089-671-2. [Google Scholar]
  23. Editorial Committee of Flora of China, Chinese Academy of Sciences. Flora of China; Science Press: Beijing, China, 1979; Volume 27, ISBN 978-7-03-048169-6. [Google Scholar]
  24. Shen, Z. Chinese Medicinal Source and Application of Trollius chinensis Bunge. Lishizhen Med. Mater. Medica Res. 2000, 12, 1110. [Google Scholar]
  25. Zhang, T.; Guo, W.; Zhao, H.; LI, S.; Wang, L. Study on Anti-enterovirus 71 Material Basis of Jinlianhua (Trollius chinensis Bge. Based on Spectrum-effect Correlation). J. Shandong Univ. Tradit. Chin. Med. 2022, 46, 386–392. [Google Scholar]
  26. National Pharmacopoeia Committee. Pharmacopoeia of the People’s Republic of China (One Part); China Medical Science and Technology Press: Beijing, China, 2020. [Google Scholar]
  27. Wang, T. Fundamental Studies on the Anti-Influenza Virus and Pharmacodynamic Substances in Golden Lotus Flower Soup. Master’s Thesis, Beijing Unviersity of Chinese Medicime, Beijing, China, 2018. [Google Scholar]
  28. Zheng, Y. Summary on “Handbook of Folk Chinese Herbal Medicines Commonly Used in Guangxi”(1,2 Set); National Committee on the Assessment of the Protected Tradtional Chinese Medicinal Products: Beijing, China, 2009.
  29. Geng, D.; Pang, Y.; Tao, Z.; Wang, S.; Liu, X.; Wang, R. Investigation on Potential of Jinlianhua Decoction against Novel Coronavirus(2019-nCoV)Based on Molecular Docking. Mod. Chin. Med. 2020, 22, 522–532. [Google Scholar]
  30. Liu, Q.; Wang, Y.; Li, J.; Ma, D.; Wang, L.; Liang, Y.; Liu, J. Chemical components and pyrolysis regularity study of medicine and foodhomology Trollius chinensis Bge. by UPLC-MS. China Food Addit. 2022, 33, 51–61. [Google Scholar]
  31. Fu, X.; Li, L. Research progress on the extraction and application of yellow pigment of Trollius chinensis. Agric. Technol. 2022, 42, 4–6. [Google Scholar]
  32. Hebei People’s Publishing House. Guang Qun Fang Pu; Beijing Ancient Books Publishing House Co., Ltd.: Beijing, China, 1989; ISBN 978-7-20200-448-7. [Google Scholar]
  33. Cha, X. A Sea Record of Cha Shenxing; Beijing Ancient Books Publishing House Co., Ltd.: Beijing, China, 1989; ISBN 978-7-53000-002-1. [Google Scholar]
  34. Science Press. Hebei Traditional Chinese Medicine Manual; Hebei Province Revolutionary Committee Commercial Bureau Medical Supply Station Co., Ltd.: Beijing, China, 1970.
  35. Meng, Z. Mongolian medicine training class. In Compilation of Mongolian Medicine Prescriptions; Beijing Ancient Books Publishing House Co., Ltd.: Beijing, China, 2004; ISBN 9787538012521. [Google Scholar]
  36. Inner Mongolia Revolutionary Committee Health Bureau. Inner Mongolia Chinese Herbal Medicine; Inner Mongolia Autonomous Region Press, Ltd.: Hohhot, China, 1989. [Google Scholar]
  37. Wang, Z.; Yang, S.; Gao, Y.; Huang, J. Extraction and Purification of Antioxidative Flavonoids from Chionanthus retusa Leaf. Front. Bioeng. Biotechnol. 2022, 10, 1085562. [Google Scholar] [CrossRef]
  38. Wu, X.; Zhao, Y. Progress on natural flavonoid carbon glycosides and their activities. Pharm. J. Chin. People’s Lib. Army 2005, 21, 135–138. [Google Scholar]
  39. Sun, P.; Li, X.; Xue, T.; Xin, J.; Liu, Y.; Chen, Y.; Guo, S.; Zhang, B. Research progress of authenticity identification and quality evaluation methods of Trollius chinensis. Mod. Chin. Med. 2022, 24, 2048–2054. [Google Scholar]
  40. Witkowska-Banaszczak, E. Flavonoids from Trollius Europaeus Flowers and Evaluation of Their Biological Activity. J. Pharm. Pharmacol. 2018, 70, 550–558. [Google Scholar] [CrossRef]
  41. Li, D.-Y.; Wei, J.-X.; Hua, H.-M.; Li, Z.-L. Antimicrobial Constituents from the Flowers of Trollius chinensis. J. Asian Nat. Prod. Res. 2014, 16, 1018–1023. [Google Scholar] [CrossRef]
  42. Li, Z.; Li, D.; Hua, H.; Chen, X.; Kim, C. Three New Acylated Flavone C-Glycosides from the Flowers of Trollius chinensis. J. Asian Nat. Prod. Res. 2009, 11, 426–432. [Google Scholar] [CrossRef]
  43. Qin, Y.; Liang, Y.; Ren, D.; Qiu, X.; Li, X. Separation of Phenolic Acids and Flavonoids from Trollius chinensis Bunge by High Speed Counter-Current Chromatography. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2015, 1001, 82–89. [Google Scholar] [CrossRef] [PubMed]
  44. Wei, J.-X.; Li, D.-Y.; Li, Z.-L. Acylated Flavone 8-C-Glucosides from the Flowers of Trollius chinensis. Phytochem. Lett. 2018, 25, 156–162. [Google Scholar] [CrossRef]
  45. Wu, L.-Z.; Zhang, X.-P.; Xu, X.-D.; Zheng, Q.-X.; Yang, J.-S.; Ding, W.-L. Characterization of Aromatic Glycosides in the Extracts of Trollius Species by Ultra High-Performance Liquid Chromatography Coupled with Electrospray Ionization Quadrupole Time-of-Flight Tandem Mass Spectrometry. J. Pharm. Biomed. Anal. 2013, 75, 55–63. [Google Scholar] [CrossRef] [PubMed]
  46. Shi, Y.; Sang, L.; Yan, R.; Zhao, Q.; Zhang, Y.; Zhao, G.; Li, Y.; Chen, X.; Zhang, C.; Qiao, H.; et al. Two New Compounds from Trollius chinensis Bunge. J. Nat. Med. 2017, 71, 281–285. [Google Scholar]
  47. Song, Z.; Wang, H.; Ren, B.; Zhang, B.; Hashi, Y.; Chen, S. On-Line Study of Flavonoids of Trollius chinensis Bunge Binding to DNA with Ethidium Bromide Using a Novel Combination of Chromatographic, Mass Spectrometric and Fluorescence Techniques. J. Chromatogr. A 2013, 1282, 102–112. [Google Scholar] [CrossRef] [PubMed]
  48. Cai, S.-Q.; Wang, R.; Yang, X.; Shang, M.; Ma, C.; Shoyama, Y. Antiviral Flavonoid-Type C-Glycosides from the Flowers of Trollius chinensis. Chem. Biodivers. 2006, 3, 343–348. [Google Scholar] [CrossRef]
  49. Zhang, J.; Yan, R.; Zhang, P.; Shi, S.; Chen, X.; Zhang, G. Isolation and identification of flavonoid chemical components in Lotus japonica. J. Shenyang Pharm. Univ. 2018, 35, 344–347+373. [Google Scholar]
  50. Yan, R.; Cui, Y.; Deng, B.; Bi, J.; Zhang, G. Flavonoid Glucosides from the Flowers of Trollius chinensis Bunge. J. Nat. Med. 2019, 73, 297–302. [Google Scholar] [CrossRef]
  51. Wu, W. Intestinal Absorption of Chemical Constituents from T. chinensis Based on Caco-2 Cell Modeling. Master’s Thesis, Beijing University of Chinese Medicine, Beijing, China, 2020. [Google Scholar]
  52. Du, R.; Shi, Z.; Zhan, Z.; Hu, J.; Zheng, G. Optimization of the extraction process of total phenolic acid from Trollii Flos by response surface methodlogy and study on its whitening activity. Nat. Prod. Res. Dev. 2023, 35, 915–924. [Google Scholar]
  53. Wang, R.; Yang, X.; Ma, C.; Chai, S.; Xu, T. Analysis of Fatty Acids from the Flowers of Trollius chinensis. J. Chin. Med. Mater. 2010, 33, 1579–1581. [Google Scholar]
  54. Wu, X.-W.; Wang, R.-F.; Liu, L.-J.; Guo, L.-N.; Zhao, C. Absorbability, Mechanism and Structure-Property Relationship of Three Phenolic Acids from the Flowers of Trollius chinensis. Molecules 2014, 19, 18129–18138. [Google Scholar] [CrossRef]
  55. Qiu, Y. Study on the Extraction Process and Antibacterial Activity Evaluation of Trollius chinensis Bunge. Master’s Thesis, South China University of Technology, Guangzhou, China, 2022. [Google Scholar]
  56. Shi, S.; Zhang, J.; Liu, T.; Ma, Q.; Liu, C.; Zhang, G. Isolation and structural identification of chemical constituents from Trollius chinensis Bunge. J. Shenyang Pharm. Univ. 2017, 34, 297–301+316. [Google Scholar]
  57. Zhao, D. Study on Chemical Composition and Biological Activity of Trollius chinensis Bunge. Master’s Thesis, Xiamen University, Xiamen, China, 2019. [Google Scholar]
  58. Zuo, J. Extraction of Flavonoids from Short-Petaled Goldenseal Flower by Natural Low-Eutectic Solvents and Molecular Blotting Technique. Master’s Thesis, Inner Moncolia University, Hohhot, China, 2022. [Google Scholar]
  59. Liu, S.; Zhao, C.; Ding, P.; Tao, Z.; Geng, D.; Fan, R. Study on the Metabolites of Goldenseal Extract. Mod. Chin. Med. 2020, 22, 1027–1047. [Google Scholar]
  60. Liu, Y.; Guo, Q.; Zhang, S.; Bao, Y.; Chen, M.; Gao, L.; Zhang, Y.; Zhou, H. Polysaccharides from Discarded Stems of Trollius chinensis Bunge Elicit Promising Potential in Cosmetic Industry: Characterization, Moisture Retention and Antioxidant Activity. Molecules 2023, 28, 3114. [Google Scholar] [CrossRef] [PubMed]
  61. Yu, L. Distribution of T. chinensis resources, current cultivation status, main chemical constituents, and pharmacological effects of T. chinensis in Mongolian medicine. J. Med. Pharm. Chin. Minor. 2016, 22, 39–41. [Google Scholar]
  62. Zhang, L.; Wu, D.; Zhang, A. ICP-AES for primary morphological analysis of inorganic elements in Trollius chinensis Bunge and its dissolution characterization. Chin. J. Spectrosc. Lab. 2011, 28, 739–742. [Google Scholar]
  63. Li, G.; Liu, J. Determination of trace elements in Trollius chinensis Bunge of different origins. Lishizhen Med. Mater. Medica Res. 2014, 25, 2533–2534. [Google Scholar]
  64. Zhang, H.; Wang, L.; Chai, C.; Wang, W.; Song, C. Study on the ultrasonic extraction process of nasturtium pigment optimized by response surface method. China Condiment 2012, 37, 102–107. [Google Scholar]
  65. Mao, X.; Gu, S.; Liu, S.; Huang, W. The Inhibitory Effect of Jinlianhua Soft Capsule Against Human Coronavirus OC43. J. Pharm. Today 2021, 31, 914–917. [Google Scholar]
  66. Zhou, Q.; Yang, C.; Zhang, Y.; Jin, Z.; Zhou, J.; Zhang, G. Exploring the anti-influenza virus mechanism of action of Golden Lotus based on network pharmacology and molecular docking. J. Shenyang Pharm. Univ. 2022, 39, 1100–1110. [Google Scholar]
  67. Xie, C.; Ren, J. Network pharmacology research and reflection on the efficacy of traditional Chinese medicine and compound formulas. Chin. J. Exp. Tradit. Med. Formulae 2024, 30, 198–207. [Google Scholar]
  68. Zhao, H.; Zhao, Y. Studies on the in vitro anti-influenza A virus effect of an alcoholic extract of Amaryllis flowers. China Pharm. 2010, 19, 10–11. [Google Scholar]
  69. Liu, X.; Liang, Y.; Wang, R.; Hu, X. Experimental study on the protection of the lungs of mice modeled with H1N1 virus infection by extracts of Auricularia auriculae. Chin. Pharmacol. Bull. 2017, 33, 1034–1035. [Google Scholar]
  70. Wen, Y.; Lin, Y.; Huang, H.; Liu, X.; Wei, G.; Fu, F.; Wu, P.; Yang, C. Experimental study on the antiviral activity of aqueous extracts of goldenseal flowers. Chin. J. Microbiol. Immunol. 1999, 01, 25. [Google Scholar]
  71. Liang, Y.; Liu, X.; Zhang, L.; Wang, R.; Hu, X. Effects of different extracts of Golden Lotus Flower Soup on cell proliferation and in vitro antiviral activity studies. J. Med. Res. 2018, 47, 100–106. [Google Scholar]
  72. Wang, T.; Liu, S.; Ding, P.; LIU, S.; Wang, Q.; Pang, Y.; Hu, X.; Wang, R. Study on the anti-influenza virus activity of Golden Lotus Flower Soup. Mod. Chin. Med. 2020, 22, 202–206. [Google Scholar]
  73. Liu, L.-J.; Li, D.-I.; Fang, M.-Y.; Liu, S.-Y.; Wang, Q.-Q.; Liang, Y.-X.; Hu, X.-H.; Wang, R.-F. The Antiviral Mechanism of the Crude Extract from the Flowers of Trollius chinensis Based on TLR 3 Signaling Pathway. Pak. J. Pharm. Sci. 2021, 34, 1743–1748. [Google Scholar]
  74. Shi, D. Study on the Anti-Influenza Virus H1N1 Mechanism of Goldenseal Based on TLRs Pathway. Master’s Thesis, Beijing University of Chinese Medicine, Beijing, China, 2018. [Google Scholar]
  75. Qu, H.; Yang, G.; Jiang, W.; Yuan, D.; Wang, S.; An, F. Dynamic effects of Orientin and Bauhinia glycosides from Goldenseal on serum and tissue antioxidant activities in D-galactose-induced senescent mice. Chin. J. Gerontol. 2015, 35, 443–446. [Google Scholar]
  76. Liang, W.; Yang, Z.; Huang, Y. Mitochondrial Ca2+ transport and regulation of cellular metabolism. Prog. Physiol. Sci. 2000, 04, 357–360. [Google Scholar]
  77. Yang, G.; Rao, N.; Tian, J.; An, F.; Wang, S. Studies on the antioxidant effects of Orientin and Oryza sativa glycosides in goldenseal flowers. Lishizhen Med. Mater. Medica Res. 2011, 22, 2172–2173. [Google Scholar]
  78. Tian, J.; Yuan, B.; Zhu, D.; Wang, S.; An, F. Dynamic antioxidant effects of Orientin and Bauhinia glycosides from Goldenseal on D-galactose-induced senescence in mice. Chin. J. Gerontol. 2014, 34, 5512–5513. [Google Scholar]
  79. Cai, H. Study on the Commodity Specification Grade of Goldenseal Flower. Master’s Thesis, Hubei University of Chinese Medicine, Wuhan, China, 2022. [Google Scholar]
  80. Liu, C. Study on the Anti-Inflammatory and Antibacterial Potency Sites of T. chinensis Bunge Decoction. Master’s Thesis, Beijing University of Chinese Medicine, Beijing, China, 2016. [Google Scholar]
  81. Zhong, L.; Yang, J.; Feng, M.; Dai, J.; Wang, Z.; Jiang, Q.; Pan, C. The mechanism of Jinlianhua series preparations in the treatment of upper respiratory tract infection based on data mining. J. Chengdu Univ. (Nat. Sci.) 2021, 40, 1–7. [Google Scholar]
  82. Wang, R.; Geng, D.; Wu, X.; An, Y. Studies on the anti-inflammatory activities of four major components of goldenseal flowers. Lishizhen Med. Mater. Medica Res. 2012, 23, 2115–2116. [Google Scholar]
  83. Yang, N.; Dong, Z.; Tian, G.; Zhu, M.; Li, C.; Bu, W.; Chen, J.; Hou, X.; Liu, Y.; Wang, G.; et al. Protective Effects of Organic Acid Component from Taraxacum Mongolicum Hand.-Mazz. against LPS-Induced Inflammation: Regulating the TLR4/IKK/NF-κB Signal Pathway. J. Ethnopharmacol. 2016, 194, 395–402. [Google Scholar] [CrossRef]
  84. Lin, Q.; Zhao, N.; Sun, Q.; Wang, L.; Meng, F. Clinical efficacy of Trollius chinensis preparation against upper respiratory tract infection: A Meta analysis. Chin. J. Pharmacovigil. 2022, 19, 893–896+907. [Google Scholar]
  85. Gu, Z.; Jin, Y. Observation on the effect of gold lotus granules combined with cefixime granules in the treatment of pediatric acute respiratory tract infections. J. Clin. Med. Pract. 2019, 23, 79–82. [Google Scholar]
  86. Li, H. Efficacy of gold lotus granules combined with ribavirin in the treatment of pediatric acute upper respiratory tract infection and its effect on serum inflammatory markers. China Med. Pharm. 2019, 9, 21–24. [Google Scholar]
  87. Su, L.; Cai, L.; Wang, J. Study on the clinical efficacy of gold lotus granules in the treatment of patients with upper respiratory tract infections. Strait Pharm. J. 2019, 31, 227–228. [Google Scholar]
  88. Zeng, X.; Lin, L.; Mai, L. Clinical study of gold lotus granules combined with cefixime in the treatment of pediatric acute respiratory tract infections. Drugs Clin. 2018, 33, 84–87. [Google Scholar]
  89. Guo, Y.; Liu, W.; Ju, A.; Ma, W. Study on the anti-inflammatory mechanism of the active ingredients of goldenseal flower based on network pharmacology. Acta Chin. Med. Pharmacol. 2020, 48, 25–28. [Google Scholar]
  90. Wan, S.; Liu, L.; Liu, M.; Huang, X. Studies on the pharmacological mechanism of action of Orientin. J. Med. Res. 2018, 47, 183–186. [Google Scholar]
  91. Xiao, Q.; Qu, Z.; Yang, L.; Zhao, Y.; Gao, P. Orientin Ameliorates LPS-Induced Inflammatory Responses through the Inhibitory of the NF-κB Pathway and NLRP3 Inflammasome. Evid.-Based Complement. Altern. Med. 2017, 2017, 2495496. [Google Scholar] [CrossRef] [PubMed]
  92. Sun, L.; Liu, F.; Liu, H.; Luo, Q.; An, F. The effects of Trollius flavonoids on human breast cancer cells. Chin. J. Gerontol. 2009, 29, 1098–1099. [Google Scholar]
  93. Wang, S.; Tian, Q.; An, F. Growth Inhibition and Apoptotic Effects of Total Flavonoids from Trollius chinensis on Human Breast Cancer MCF-7 Cells. Oncol. Lett. 2016, 12, 1705–1710. [Google Scholar] [CrossRef]
  94. Li, S.; Wu, X.; Ma, Y.; Zhang, H.; Chen, W. Prediction and Verification of the Active Ingredients and Potential Targets of Erhuang Quzhi Granules on Non-Alcoholic Fatty Liver Disease Based on Network Pharmacology. J. Ethnopharmacol. 2023, 311, 116435. [Google Scholar] [CrossRef] [PubMed]
  95. Cui, W.; Yang, J.; Chen, X.; Mao, Q.; Wei, X.; Wen, X.; Wang, Q. Triterpenoid-Rich Fraction from Ilex Hainanensis Merr. Attenuates Non-Alcoholic Fatty Liver Disease Induced by High Fat Diet in Rats. Am. J. Chin. Med. 2013, 41, 487–502. [Google Scholar] [CrossRef]
  96. Fan, R.; Li, W.; Liu, Y.; Li, H. Effects of Total Flavonoids from Flos Trolliion the Abnormal Function o of in vitro HEPGCells Induced by High Concentration of Glucos. J. Med. Res. 2021, 50, 124–129+112. [Google Scholar]
  97. Pouwels, S.; Sakran, N.; Graham, Y.; Leal, A.; Pintar, T.; Yang, W.; Kassir, R.; Singhal, R.; Mahawar, K.; Ramnarain, D. Non-Alcoholic Fatty Liver Disease (NAFLD): A Review of Pathophysiology, Clinical Management and Effects of Weight Loss. BMC Endocr. Disord. 2022, 22, 63. [Google Scholar] [CrossRef]
  98. Wang, X.; Lin, C.; Meng, L.; Zhou, J.; Liang, X.; Yang, H.; Qin, Y. Effect of high free fatty on E2F1 expression and invasion and migration of HepG2 cells. J. Guangxi Med. Univ. 2022, 39, 276–281. [Google Scholar]
  99. Zhu, D.; An, F.; Wang, S. Effects of orientin and vitexin in Trollius chinensis Bunge on growth and apoptosis of human esophageal cancer cells. Chin. J. Gerontol. 2013, 33, 4472–4475. [Google Scholar]
  100. Liu, R. Research on the Key Technology of Containerized Seedling and Grassland Transplanting of Amaryllis Japonica. Master’s Thesis, Ninner Mongolia Agricultural University, Hohhot, China, 2022. [Google Scholar]
  101. Li, Y.; Ding, W. Genetic diversity Analysis of partial Chinese Trollius populations Based onRAPD markers. J. Plant Genet. Resour. 2010, 11, 535–539. [Google Scholar]
  102. Tian, J.; Yang, G.; Rao, N.; An, F.; Wang, S. Effect of Orientin and Vitexin in Trollius chinesis on cell membrane transport activity of aging mice induced by D-galactose. Chin. J. Gerontol. 2012, 32, 3945–3947. [Google Scholar]
  103. Ye, S.; Li, Y.; Yang, Y.; Cen, Y. Extraction technology of Trollius chinensis Bunge. China J. Chin. Mater. Medica 2002, 6, 66–67. [Google Scholar]
  104. Ye, Y.; Peng, Y.; Fu, G.; Miu, J. New progress in the research of medicinal goldenseal flower. Mod. Chin. Med. 2007, 3, 29–33. [Google Scholar]
  105. Tian, P.; Liu, R.; Deng, Y.; Zhong, L.; Lei, S.; Wu, L. Re-evaluation of goldenseal series of preparations based on the mechanism of antibacterial effect. World Notes Antibiot. 2023, 44, 263–268. [Google Scholar]
  106. Liu, P.; Chen, G.; Deng, S.; Liu, Y.; Dong, J. Experimental study on the antimicrobial effect of total flavonoids of Amaryllis flowers. Chin. J. Exp. Tradit. Med. Formulae 2013, 19, 207–210. [Google Scholar]
  107. Peng, Y.; Liu, L.; Zhao, C.; Guo, L.; Wang, R. First isolation of a phenolic acid compound and its anti-inflammatory and bacteriostatic activities from Lotus corniculatus flower. Chin. Arch. Tradit. Chin. Med. 2015, 33, 1349–1351. [Google Scholar]
  108. Lin, Q.; Feng, S.; Li, Y.; Cen, Y.; Yang, Y.; Wang, L. Study on the antibacterial and antiviral activity compositions of Trollius chinensis Bunge. J. Zhejiang Univ. (Sci. Ed.) 2004, 31, 412–415. [Google Scholar]
  109. Wang, X.; Guo, R.; Nie, X.; Chen, H.; Zhi, W.; Tian, M.; Liu, F. Studies on the antibacterial activity of Forsythia and the mechanism of action of its pharmacodynamic component hesperidin against Staphylococcus aureus. Chin. J. Antibiot. 2021, 46, 437–441. [Google Scholar]
  110. Liu, P.; Hu, N.; Chen, G.; Wang, Y.; Liu, Y.; Tong, J. Antipyretic Effect of Flavonoids from Trollius ledebouri Reichb on Endotoxin-induced Fever and Levels of TNF-α, IL-1β and PGE2 in Rabbits. Chin. J. Exp. Tradit. Med. Formulae 2014, 20, 189–191. [Google Scholar]
  111. Su, L.; Zhao, W.; Nan, Y.; Xing, R. Experimental study on anti-inflammatory and analgesic effects of extracts from stems and leaves of Trollius chinensis. Chin. J. Tradit. Med. Sci. Technol. 2012, 19, 31. [Google Scholar]
  112. You, S.; Liu, X.; NaiMaiTi, A.; Chen, W.; Zhu, Q.; Qi, X.; Zhao, J.; Liu, T. Experimental study on the antitussive, expec etorant, anti-inflammatory and analgesic effects of total flavonoids extracted from Trollius chinensis Bunge. J. Xinjiang Med. Univ. 2019, 42, 462–466. [Google Scholar]
  113. Fang, J.; Meng, X.; Liu, Y. Protective effects of flavone of Trollius chinensis Bunge on myocardial ischemia reperfusion injury in rats. J. Chongqing Med. Univ. 2014, 39, 1391–1395. [Google Scholar]
  114. Fan, C.; LI, J.; Sun, H. Study on the technology of supercritical CO2 extraction of total flavonoids from Trollius chinensis Bunge and the antioxidant effect of the extract. J. Chin. Med. Mater. 2016, 39, 2828–2832. [Google Scholar]
  115. Liu, X.; Liang, Y.; Wang, R.; Hu, X. Studies on the fatty acid composition of Auricularia auricula. J. Chin. Med. Mater. 2017, 33, 1034–1035. [Google Scholar]
  116. Shi, D.; Chen, M.; Liu, L.; Wang, Q.; Liu, S.; Wang, L.; Wang, R. Anti-Influenza A Virus Mechanism of Three Representative Compounds from Flos Trollii via TLRs Signaling Pathways. J. Ethnopharmacol. 2020, 253, 112634. [Google Scholar] [CrossRef]
  117. Li, H.; Zhang, M.; Ma, G. Radical Scavenging Activity of Flavonoids from Trollius chinensis Bunge. Nutrition 2011, 27, 1061–1065. [Google Scholar] [CrossRef]
  118. An, F.; Yang, G.; Tian, J.; Wang, S. Antioxidant Effects of the Orientin and Vitexin in Trollius chinensis Bunge in D-Galactose-Aged Mice. Neural Regen. Res. 2012, 7, 2565–2575. [Google Scholar]
  119. Liu, L.-J.; Hu, X.-H.; Guo, L.-N.; Wang, R.-F.; Zhao, Q.-T. Anti-Inflammatory Effect of the Compounds from the Flowers of Trollius chinensis. Pak. J. Pharm. Sci. 2018, 31, 1951–1957. [Google Scholar] [PubMed]
  120. An, F.; Wang, S.; Tian, Q.; Zhu, D. Effects of Orientin and Vitexin from Trollius chinensis on the Growth and Apoptosis of Esophageal Cancer EC-109 Cells. Oncol. Lett. 2015, 10, 2627–2633. [Google Scholar] [CrossRef]
  121. Jiang, M.; Yan, L.; Li, K.-A.; Ji, Z.-H.; Tian, S.-G. Evaluation of Total Phenol and Flavonoid Content and Antimicrobial and Antibiofilm Activities of Trollius chinensis Bunge Extracts on Streptococcus Mutans. Microsc. Res. Tech. 2020, 83, 1471–1479. [Google Scholar] [CrossRef]
  122. Bai, Y.; Qi, X.; Gao, Y. Pharmacognostical studies on Trollius chinensis Bunge. J. Shanxi Med. Univ. 2001, 1, 27–28. [Google Scholar]
  123. Nie, B.; Zhang, G.; Sun, S.; Zhou, Q.; Ding, W. IR identification study of different nasturtium medicinal materials. J. Chin. Med. Mater. 2006, 04, 323–326. [Google Scholar]
  124. Pan, Y.; Xiao, P.; Zhang, G.; Lu, J.; Sun, S. Fourier infrared spectroscopy identification and characterization of pharmacodynamic components of nasturtium. China J. Chin. Mater. Medica 2006, 12, 1024–1026. [Google Scholar]
  125. Wang, H.; Ye, H.; Xu, Q.; Liu, J.; Xie, Z. Study on the Quality of Nasturtium Chinese Medicine Tablets. Chin. J. Mod. Appl. Pharm. 2020, 37, 963–966. [Google Scholar]
  126. Yuan, Q.; Zhang, W.; Jiang, D. On the Methods and Principles of Molecular Identification of Chinese Herbs. Plant Divers. 2012, 34, 607. [Google Scholar] [CrossRef]
  127. Zhang, L. Identification, Characterization and Activity Study of Goldenseal Proteins; Beijing University of Chinese Medicine: Beijing, China, 2006. [Google Scholar]
  128. Guan, Y.; Ding, X.; Wang, W.; Di, L.; Wang, X. X-Ray Fluorescence Analysis and X-ray Diffraction of Goldenseal Flowers. Chin. J. Pharm. Anal. 2006, 26, 1623–1625. [Google Scholar]
  129. Huang, R.; Zhang, G.; Pan, Y.; Cui, H.; Zhao, Y. Analysis and Identification on Characterization of Chemical Components in Trollii Flos by HPLC-MS. Zhongcaoyao Chin. Tradit. Herb. Drugs 2012, 43, 670–672. [Google Scholar]
  130. Sun, Y.; Li, Q.; Zhang, Q. Determination of Orientin, Vitexin and Total Flavonoids in Trollius chinensis from Different Areas and Optimization of Extraction Process. Northwest Pharm. J. 2019, 34, 596–601. [Google Scholar]
  131. Song, Z.; Hashi, Y.; Sun, H.; Liang, Y.; Lan, Y.; Wang, H.; Chen, S. Simultaneous Determination of 19 Flavonoids in Commercial Trollflowers by Using High-Performance Liquid Chromatography and Classification of Samples by Hierarchical Clustering Analysis-ScienceDirect. Fitoterapia 2013, 91, 272–279. [Google Scholar] [CrossRef]
Figure 1. Distribution of T. chinensis. (The green shading represents the distribution of T. chinensis, white is the area where T. chinensis almost does not exist, and the bottom half of the image is the typical height legend of T. chinensis (map approval number: GS(2019)1822)).
Figure 1. Distribution of T. chinensis. (The green shading represents the distribution of T. chinensis, white is the area where T. chinensis almost does not exist, and the bottom half of the image is the typical height legend of T. chinensis (map approval number: GS(2019)1822)).
Molecules 29 00421 g001
Figure 2. The plant of Trollius chinensis Bunge (Ranunculaceae). (A,C,D) flowers (B) the medicinal part after processing.
Figure 2. The plant of Trollius chinensis Bunge (Ranunculaceae). (A,C,D) flowers (B) the medicinal part after processing.
Molecules 29 00421 g002
Figure 3. Parent nucleus structure of flavonoid chemicals in T. chinensis.
Figure 3. Parent nucleus structure of flavonoid chemicals in T. chinensis.
Molecules 29 00421 g003
Figure 4. Parent nucleus structure of Phenolic acids chemicals in T. chinensis.
Figure 4. Parent nucleus structure of Phenolic acids chemicals in T. chinensis.
Molecules 29 00421 g004
Figure 5. Alkaloids isolated from T. chinensis.
Figure 5. Alkaloids isolated from T. chinensis.
Molecules 29 00421 g005
Figure 6. Other chemical components isolated from T. chinensis.
Figure 6. Other chemical components isolated from T. chinensis.
Molecules 29 00421 g006
Figure 7. The pharmacological properties of T. chinensis.
Figure 7. The pharmacological properties of T. chinensis.
Molecules 29 00421 g007
Table 1. A total of 26 species of Trollius genus.
Table 1. A total of 26 species of Trollius genus.
NumLatin NameDistribution AreaAltitude
1T. chinensis BungeShanxi, N. Henan, Hebei, E. Inner Mongolia, W. Liaoning and Jilin provinces of China1000–2200 m
2Trollius altaicus C. A. Mey.N. Xinjiang (Tacheng, Altai, etc.), China; W. Inner Mongolia, China; Siberia, Russia; People’s Republic of Mongolia1200–2650 m
3Trollius asiaticus L.Heilongjiang, China (Shangzhi); Xinjiang, China (Hami); Siberia, Russia; MongoliaNot applicable
4 Trollius buddae Schipcz.N. Sichuan, China; S. Gansu, China; S. Shaanxi, China1780–2400 m
5Trollius buddae f. dolichopetalus P. L. Liu and C. DuNot applicableNot applicable
6Trollius dschungaricus RegelTianshan and Zhaosu, Xinjiang, China; Central Asia, Russia1800–3100 m
7Trollius farreri StapfQinghai, China2000–4700 m
8Trollius farreri Stapf var/. major W. T. WangNW Yunnan, China (Deqin); SE Tibet, China (Tsatsumi)3500–4200 m
9Trollius japonicus Miq.Changbai Mountain, Jilin, China; Sakhalin Island (Kuril Islands); Japan1200–2300 m
10Trollius ledebourii Rchb.Heilongjiang, China; NE Inner Mongolia, China; E. Siberia, Russia; Far East110–900 m
11Trollius macropetalus Fr.Liaoning, China; Jilin, China; Heilongjiang, China, etc.; Russian Far East; N Korea;450–600 m;
12Trollius micranthus Hand.-Mazz.NW Yunnan (Deqin); E. Tibet (Motuo)3900–4200 m
13Trollius pumilus D. DonSouthern Tibet, China; Nepal; Sikkim4100–4800 m
14Trollius pumilus D. Don var. foliosus (W. T. Wang) W. T. WangMin County, S. Gansu, China3000–3400 m
15Trollius pumilus D. Don var. tanguticus BrühlNE Tibet, China; NW Sichuan, China; S. and E. Qinghai, China; SW Gansu, China.2300–3700 m
16Trollius pumilus D. Don var. tehkehensis (W. T. Wang) W. T. WangDege, Sichuan, ChinaNot applicable
17Trollius ranunculoides Hemsl.NW Yunnan, E Xizang, W Sichuan, S and E Qinghai, S Gansu, China.2900–4100 m
18Trollius taihasenzanensis Masam.Taiwan, China3400–3900 m
19Trollius vaginatus Hand.-Mazz.NW Yunnan (Zhongdian), China; SW Sichuan (Muli), China.3000–4200 m
20Trollius yunnanensis (Franch.) Ulbr.W. and NW Yunnan, China; W. Sichuan, China.2700–3600 m
21Trollius yunnanensis (Franch.) Ulbr. var. anemonifolius (Brühl) W. T. WangW. Sichuan and S. Gansu, China.3050–3800 m
22Trollius yunnanensis (Franch.) Ulbr. var. eupetalus (Stapf) W. T. WangGonshan and Deqin, NW Yunnan, Sichuan, China3300–3900 m
23Trollius yunnanensis (Franch.) Ulbr. var. peltatus W. T. WangEmei area, Sichuan, China1900 m
24Trollius lilacinus BungeTian Shan, Xinjiang, China; W. Siberia, USSR; Central Asia2600–3500 m
25Trollius laxusthe United States in Conn.(Connecticut), Del.(Delaware)NJ.(New Jersey)N.Y.(New York)Pa.(Pennsylvania), Ohio.(Ohio)Not applicable
26Trollius europaeusN. Europe, Central Europe and W. AsiaNot applicable
Not applicable means that no relevant information is to be found.
Table 2. The ethnopharmacological use of T. chinensis.
Table 2. The ethnopharmacological use of T. chinensis.
NOEthnopharmacological UseReferences
1Ornamental: The whole flower is golden yellow. It blooms in June. In autumn, the flowers are dry, and the fruit is like millet.Guang Qun Fang Pu Kangxi (AD. 1708) [32]
2Drink: Dry long-term preservation, to spend some tea, a pot of one, boiled water.
Medicinal use: taste smooth and bitter, non-toxic, cold, cure sore throat, heat flotation tooth declaration, ear pain, eye pain, and fry this generation of Ming.
A sea record of Cha Shenxing Kangxi (AD. 1713) [33]
3Medicinal use: bitter taste, cold, non-toxic, treating mouth sore throat swelling, ear pain, eye pain, sore throat, fever from a cold, eyesight.Bencao Gangmu Shiyi: A Supplement to Compendium of Materia Medica (AD. 1765) [22]
4Medicinal use: treatment of furunculosis big poison, bias wind, wind heat, wind hysteria, and wind arthralgia, et al.Mountain sea grass letter (Qing dynasty) [22]
5Medicinal use: clearing away heat and toxic materials, treatment of chronic/acute tonsillitis, acute otitis media, acute tympanitis, acute conjunctivitis, and acute lymphangitis.Hebei Traditional Chinese Medicine Manual (1970) [34]
6Medicinal use: for treating blade wounds and pulse wound sores; for swollen lymph glands and sore throats.Compilation of Mongolian medical formulas (In 2004) [35]
7Medicinal use: cure fever from an ear infection or eye disease.Inner Mongolia Herbal Medicine (1972) [36]
8Jinlianhua Mixture: clearing heat and removing toxins for upper respiratory tract infections, pharyngitis, and tonsillitis.2020 Edition of Chinese Pharmacopoeia (CP) (2020) [26]
9Jinlianhua tablets: clearing heat and removing toxins for upper respiratory tract infections, pharyngitis, and tonsillitis.2020 Edition of Chinese Pharmacopoeia (CP) (2020) [26]
10Jinlianhua capsules: Clearing heat and removing toxins, relieving pharynx and swelling. Suitable for treating inflammation of the upper Jiao, etc.2020 Edition of Chinese Pharmacopoeia (CP) (2020) [26]
11Jinlianhua granules: Treats upper respiratory tract infections, pharyngitis, and tonsillitis. Relieves inflammation and pain.2020 Edition of Chinese Pharmacopoeia (CP) (2020) [26]
12Jinlianhua Runhou tablets: clearing heat, removing toxins, reducing swelling, relieving pain, and improving the taste of the throat.2020 Edition of Chinese Pharmacopoeia (CP) (2020) [26]
13Jinlianhua granules: It is effective in clearing heat and removing toxins, promoting the production of body fluids, improving the pharynx, and relieving cough and expectoration. It is suitable for symptoms of heat and toxicity caused by colds, including high fever, thirst, and dry throat, and for the above symptoms caused by influenza and upper respiratory tract infections.2020 Edition of Chinese Pharmacopoeia (CP) (2020) [26]
This table summarizes the uses and origins of T. chinensis from ancient to modern times.
Table 3. Flavones isolated from T. chinensis.
Table 3. Flavones isolated from T. chinensis.
NoNamesMolecular FormulaParent NucleusSubstituentCASMolecular WeightRefs.
13″-O-AcetylquercetinC28H32O14Inothingnothing592.50[8]
2IsorhamnetinC21H20O10IInothing480-19-3432.38[40]
3IcariinC21H20O10IIIR1 = OH; R2 = H; R3 = H; R4 = H; R5 = H; R6 = OH489-32-7432.38[8]
4ApigeninC21H20O11IIIR1 = OH; R2 = OH; R3 = H; R4 = H; R5 = H; R6 = OH520-36-5448.41[40]
5IsoswertisinC22H22O10IIIR1 = OCH3; R2 = H; R3 = H; R4 = H; R5 = H; R6 = OH6980-40-1446.40[40]
6IsoswertiajaponinC22H22O11IIIR1 = OCH3; R2 = OH; R3 = H; R4 = H; R5 = H; R6 = OHnothing462.40[8]
7Trollisin IC22H22O10IIIR1 = OCH3; R2 = H; R3 = H; R4 = H; R5 = H; R6 = OHnothing446.40[8]
8Cyanidin 2″-O-(β-d-xyranosyl)-β-d-glucosideC26H28O16IIIR1 = OH; R2 = OH; R3 = D-xyl; R4 = H; R5 = H; R6 = OHnothing596.49[8]
9Cyanidin 2″-O-(β-d-pyranosyl)-β-d-glucosideC26H27O15NIIIR1 = OH; R2 = OH; R3 = D-glu; R4 = H; R5 = H; R6 = OHnothing593.50[8]
10Cyanidin 2-prime-O-beta-pyranosyl-arabinosideC26H28O16IIIR1 = OH; R2 = OH; R3 = D-ara; R4 = H; R5 = H; R6 = OHnothing596.50[8]
11Cyanidin 2″-O-beta-l-rhamnosideC27H30O16IIIR1 = OH; R2 = OH; R3 = L-gal; R4 = H; R5 = H; R6 = OHnothing609.15[8]
12Cyanidin 3-O-beta-d-glucoside-6″-O-alpha-l-rhamnosideC26H27O15NIIIR1 = OH; R2 = OH; R3 = H; R4 = H; R5 = D-glu; R6 = OHnothing593.50[8]
136″-O-Acetyl cyanidinC32H27O11N3IIIR1 = OH; R2 = OH; R3 = H; R4 = H; R5 = Ac; R6 = OHnothing629.58[8]
143″-O-Acetyl cyanidinC32H27O11NIIIR1 = OH; R2 = OH; R3 = H; R4 = Ac; R5 = H; R6 = OHnothing629.58[8]
152″-O-Acetyl cyanidinC32H27O11N3IIIR1 = OH; R2 = OH; R3 = Ac; R4 = H; R5 = H; R6 = OHnothing629.58[8]
16Quercetin 2″-O-(β-d-xyranosyl)-β-d-glucosideC26H28O15IIIR1 = OH; R2 = H; R3 = D-xyl; R4 = H; R5 = H; R6 = OHnothing580.50[8]
17Quercetin 2″-O-(β-d-arabinopyranoside)C26H28O15IIIR1 = OH; R2 = H; R3 = D-ara; R4 = H; R5 = H; R6 = OHnothing580.50[8]
18Rhamnetin 2″-O-β-l-rhamnosideC27H30O16IIIR1 = OH; R2 = H; R3 = L-gal; R4 = H; R5 = H; R6 = OHnothing610.53[8]
19Kaempferol 2″-O-β-d-glucopyranosideC27H30O15IIIR1 = OH; R2 = H; R3 = D-glu; R4 = H; R5 = H; R6 = OHnothing609.15[8]
20Kaempferol 6″-O-glucopyranosideC26H27O14NIIIR1 = OH; R2 = H; R3 = H; R4 = H; R5 = D-glu; R6 = OHnothing577.50[8]
216″-O-acetylkaempferolC32H27O10N3IIIR1 = OH; R2 = H; R3 = H; R4 = H; R5 = Ac; R6 = OHnothing613.58[8]
223″-O-acetylkaempferolC32H27O10N3IIIR1 = OH; R2 = H; R3 = H; R4 = Ac; R5 = H; R6 = OHnothing613.58[8]
232″-O-acetylkaempferolC32H27O10N3IIIR1 = OH; R2 = H; R3 = Ac; R4 = H; R5 = H; R6 = OHnothing613.58[8]
24Genistein-7-O-β-d-pyranosylglucosideC22H22O10IIIR1 = H; R2 = H; R3 = H; R4 = H; R5 = H; R6 = OCH3nothing446.41
284.26
[8]
253″-O-(2‴-methylbutanoyl)resveratrolC26H28O12IVR1 = OH; R2 = OHnothing532.15[8]
263″-O-(2‴-methylbutanoyl)quercetinC27H30O11IVR1 = OCH3; R2 = Hnothing530.52[41]
273″-O-(2‴-methylbutanoyl) luteolinC26H28O11IVR1 = OH; R2 = Hnothing517.17[42]
283″-O-(2‴-methylbutanoyl) chrysoeriolC27H30O12IVR1 = OCH3; R2 = OHnothing547.18[42]
29IsoorientinC21H20O11Vnothing28608-75-5448.38[40]
302″-O-feruloylharpagosideC24H31O18VIR1 = H; R2 = OH; R2 = OCH3nothing607.14[43]
312″-O-feruloylverbascosideC31H27O14VIR1 = OH; R2 = OH; R3 = OCH3nothing623.13[43]
322″-O-feruloylisovitexinC31H27O11VIR1 = H; R2 = CH3 O; R3 = OCH3nothing575.16[44]
332″-O-(3‴-methoxycaffeoyl)luteolinC30H26O13VIR1 = H; R2 = OH; R3 = OHnothing594.14[44]
342″-O-feruloylgenistinC32H30O13VIR1 = OH; R2 = CH3 O; R3 = OCH3nothing622.17[8]
352″-O-(2‴-methylbutanoyl)quercetinC26H28O11VIIR1 = OH; R2 = H; R3 = OH; R4 = CH2 CH3nothing515.15[45]
362″-O-(2‴-methylbutanoyl)kaempferolC26H28O12VIIR1 = OH; R2 = OH; R3 = OH; R4 = CH2 CH3nothing531.14[45]
374′-methoxy-2”-O-(2‴-methylbutanoyl)luteolinC32H30O13VIIR1 = OH; R2 = H; R3 = OCH3; R4 = CH2CH3nothing623.17[44]
384′-methoxy-2″-O-(2‴-methylbutanoyl)apigeninC32H30O14VIIR1 = OH; R2 = OH; R3 = OCH3; R4 = CH2CH3nothing639.17[44]
392″-O-(2‴-methylbutanoyl)isogenistinC20H33 O17VIIR1 = OCH3; R2 = OH; R3 = OH; R4 = CH2CH3nothing545.17[43]
402″-O-(2‴-methylbutanoyl)isokanamycin AC27H29 O11VIIR1 = OCH3; R2 = H; R3 = OH; R4 = CH2CH3nothing529.17[43]
412″-O-isopropylbenzoyl-isokanamycin AC32H31O14VIIR1 = OCH3; R2 = OH; R3 = OH; R4 = CH3nothing639.17[46]
423″-O-veratroyl orientinC29H26O14VIIIR = OHnothing599.13[44]
433″-O-veratroyl vitexinC31H28O13VIIIR = Hnothing608.56[44]
442″-O-benzoylisorhamnetinC29H26O12IXnothingnothing567.14[44]
453″-O-AcetylquercetinC31H28O13Xnothingnothing609.16[44]
462″-O-VanilloylquercetinC29H26O14XIR1 = OH; R2 = OH; R3 = OH; R4 = OHnothing599.13[44]
472″-O-(3‴,4‴-dimethoxybenzoyl)isorhamnetinC31H30O14XIR1 = OCH3; R2 = OH; R3 = OH; R4 = OCH3nothing626.16[45]
482″-O-(3‴,4‴-dimethoxybenzoyl) isoswertisinC31H30O13XIR1 = OCH3; R2 = H; R3 = OH; R4 = OCH3nothing610.17[45]
492″-O-(3‴,4‴-dimethoxybenzoyl)isodaidzeinC30H28O13XIR1 = OH; R2 = H; R3 = OH; R4 = OCH3nothing595.14[47]
502″-O-(3‴,4‴-dimethoxybenzoyl)quercetinC30H28O14XIR1 = OH; R2 = OH; R3 = OH; R4 = OCH3nothing611.14[47]
512″-O-vanilloylisorhamnetinC30H28O13XIR1 = OCH3; R2 = H; R3 = OH; R4 = OHnothing597.16[48]
522″-O-vanilloylquercetinC29H26O13XIR1 = OH; R2 = H; R3 = OH; R4 = OHnothing582.5[45]
53SalvigeninC18H16O6XIIR1 = H; R2 = OCH3; R3 = OCH3; R4 = OH; R5 = H; R6 = H; R7 =OCH3; R8 = H19103-54-9328.31[8]
54AcacetinC16H12O5XIIR1 = H; R2 = OH; R3 = H; R4 = OH; R5 = H; R6 = H; R7 = OCH3; R8 = H480-44-4284.26[8]
55ApigeninC15H10O5XIIR1 = H; R2 = OH; R3 = H; R4 = OH; R5 = H; R6 = H; R7 = OH; R8 = H520-36-5270.24[8]
56PectolinarigeninC17H14O6XIIR1 = H; R2 = OH; R3 = OCH3; R4 = OH; R5 = H; R6 = H; R7 = OCH3; R8 = H520-12-7314.29[8]
57CirsimaritinC17H14O6XIIR1 = H; R2 = OCH3; R3 = OCH3; R4 = OH; R5 = H; R6 = H; R7 = OH; R8 = H6601-62-3314.29[8]
58LuteolinC15H10O6XIIR1 = H; R2 = OH; R3 = H; R4 = OH; R5 = H; R6 = OH; R7 = OH; R8 = H491-70-3286.24[8]
59QuercetinC15H10O7XIIR1 = H; R2 = OH; R3 = H; R4 = OH; R5 = OH; R6 = OH; R7 = OH; R8 = H73123-10-1302.23[8]
60NaringeninC15H12O5XIIR1 = H; R2 = OH; R3 = H; R4 = OH; R5 = H; R6 = H; R7 = OH; R8 = H480-41-1272.25[8]
61ChrysoeriolC16H12O6XIIR1 = H; R2 = OH; R3 = H; R4 = OH; R5 = H; R6 = OCH3; R7 = OH; R8 = H491-71-4300.26[8]
62DiosmetinC16H12O6XIIR1 = H; R2 = OH; R3 = H; R4 = OH; R5 = H; R6 = OH; R7 = OCH3; R8 = H520-34-3300.26[8]
63FarnisinC16H12O5XIIR1 = H; R2 = OH; R3 = H; R4 = H; R5 = H; R6 = OH; R7 = OCH3; R8 = H54867-60-6284.26[8]
64KaempferolC15H10O6XIIR1 = H; R2 = OH; R3 = H; R4 = OH; R5 = OH; R6 = H; R7 = OH; R8 = H520-18-3286.24[8]
65MyricetinC15H10O8XIIR1 = H; R2 = OH; R3 = H; R4 = OH; R5 = OH; R6 = OH; R7 = OH; R8 = OH529-44-2318.23[8]
66NeodiosminC28H32O15XIIR1 = H; R2 = O-rutinoside; R3 = H; R4 = OH; R5 = H; R6 = H; R7 = OCH3; R8 = H38665-01-9608.54[8]
678-C-β-d-pyranosyl catechinC21H20O10XIIR1 = D-xyl; R2 = H; R3 = H; R4 = OH; R5 = H; R6 = OH; R7 = OCH3; R8 = Hnothing432.38[9]
687-O-viciafuranosyl quercetinC28H32O14XIIR1 = H; R2 = O-rutinoside; R3 = H; R4 = OH; R5 = H; R6 = OH; R7 = OCH3; R8 = Hnothing593.19[47]
697-O-naringenin rutinosideC28H32O14XIIR1 = H; R2 = O-neohesperidoside; R3 = H; R4 = OH; R5 = H; R6 = OH; R7 = OCH3; R8 = H20633-93-6607.16[45]
70Quercetin-3-O-β-l-rhamnosideC21H20O11XIIR1 = H; R2 = OH; R3 = H;
R4 = OH;
R5 = O-β-l-rhamnoside;
R6 = OH; R7 = OH; R8 = H
522-12-3448.38[9]
71Quercetin-3-O-β-d-glucopyranosideC21H20O11XIIR1 = H; R2 = OH; R3 = H;
R4 = OH; R6 = OH; R7 = OH;
R5 = O-β-d-glucopyrano- side; R8 = H
21637-25-2448.37[9]
725-Hydroxy-4′,7,8-trimethoxyflavoneC18H16O6XIIR1 = OCH3; R2 = OCH3; R3 = H; R4 = OH; R5 = H; R6 = H; R7 = OCH3; R8 = H57096-03-4328.09[8]
734′,5-Dihydroxy-7,8-dimethoxyflavoneC17H14O6XIIR1 = OCH3; R2 = OCH3; R3 = H; R4 = OH; R5 = H; R6 = H; R7 = OH; R8 = H6608-33-9314.08[8]
746‴-(3-hydroxy-3-methylbutanoyl)-2″-O-β-d-pyranosyl-hongcaosideC33H38O20XIIIR = OHnothing777.18[12]
756‴-(3-hydroxy-3-methylbutanoyl)-2″-O-β-d-pyranosylmatrineC33H38O19XIIIR = Hnothing761.19[12]
762″-O-veratroylvitexinC30H28O13XIVnothingnothing596.15[8]
77Isodaphnetin-2″-O-(6-O-feruloyl)-β-l-lactosideC37H38O18XVnothingnothing771.21[49]
78HyperosideC21H20O12XVIR1 = OH; R2 = OH; R3 = CH2OH482-36-0464.40[8]
79Naringenin 3-(6″-ethyl glucuronide)C23H24O10XVIR1 = OH; R2 = H; R3 = COOCH2CH3nothing460.14[8]
80AstragalinC21H20O11XVIR1 = OH; R2 = H; R3 = CH2OH480-10-4448.40[8]
81EriodictyolC15H12O6XVIInothing552-58-9288.25[8]
822″-O-(2‴-O-methybutyryl)-glucopyranosyl isoswertisinC33H40O16XVIIInothingnothing691.22[50]
832″-O-(6‴-O-veratroyl)-galactopyranosyl vitexinC36H38O18XIXnothingnothing759.21[50]
842″-O-(6‴-O-caffeoyl)-galactopyranosyl vitexinC36H36O18XXR1 = H; R2 = OH; R3 = H; R4 = OHnothing757.19[50]
852″-O-(6‴-O-feruloyl)-galactopyranosyl orientinC37H38O19XXR1 = OH; R2 = OCH3; R3 = H; R4 = OHnothing787.20[50]
86Trollichinenside A(3″-O-veratroylvitexin)C36H35O18XXR1 = OH; R2 = OH; R3 = H; R4 = OHnothing755.18[8]
87Trollichinenside B (3″-O- feruloylvitexin)C38H40O20XXR1 = OH; R2 = OCH3; R3 = OCH3; R4 = OHnothing816.21[8]
88Trollichinenside C (6″-O-veratroylvitexin)C38H40O19XXR1 = OH; R2 = H; R3 = OCH3; R4 = OCH3nothing800.22[8]
89DaidzinC21H20O9XXIR1 = H; R2 = OH552-66-9416.41[8]
90Kaempferol-7-O-β-d-glucosideC22H22O10XXIR1 = OH; R2 = OCH3nothing446.12[8]
91GlucosylorientinC27H30O17XXIInothing76135-83-6626.5[8]
926″-MalonylcosmosiinC24H22O13XXIIInothing86546-87-4518.4[8]
93(-)-GallocatechiC15H14O7XXIVnothingnothing306.27[8]
94Quercetin-2″-O-β-l-arabinopyranosideC27H30O16XXVnothing861691-37-4610.15[8]
95Apigenin-8-C-(2″-O-feruloyl)-β-d-glucosideC31H28O13XXVInothingnothing608.15[8]
96(2E)-2-methyl-1-O-vanilloyl-4-β-d-glucopyrano-side-2-buteneC19H26O10XXVIInothingnothing437.14[46]
97NeocarlinosideC26H28O15XXVIIInothing83151-89-7580.5[8]
984′,5-dihydroxy-3′,7-dimethoxy-isoflavoneC17H14O6XXIXR1 = OCH3; R2 = H; R3 = OH; R4 = OCH3nothing314.08[8]
99GlyciteinC16H12O5XXIXR1 = OH; R2 = OCH3; R3 = H; R4 = H40957-83-3284.26[8]
100DaidzeinC15H10O4XXIXR1 = OH; R2 = H; R3 = H; R4 = H486-66-8254.23[8]
Table 4. Phenolic acids isolated from T. chinensis.
Table 4. Phenolic acids isolated from T. chinensis.
NoNamesMolecular FormulaParent NucleusSubstituentCASMolecular WeightRefs.
101TrolliosideC19H26O9XXXR1 = O-β-d-glucopyranosyl;
R2 = OCH3
nothing399.16[9]
102Proglobellowery acidC7H6O2XXXR1 = OH; R2 = OCH3nothing235.00[9]
1034-(β-d-glucopyranosyloxy)-3-
(3-methyl-2-butenyl)benzoic acid
C18H24O8XXXR1 = O-β-d-glucopyranosyl; R2 = Hnothing368.38[9]
1044-Hydroxybenzoic acidC7H6O3XXXIR1 = COOH; R2 = H; R3 = OH; R4 = H99-96-7138.03[9]
1053,4-dihydroxybenzoic acid methyl esterC8H8O2XXXIR1 = COOCH3; R2 = OH; R3 = OH; R4 = H2150-43-8152.05[9]
106MethylparabenC6H4O3XXXIR1 = COOCH3; R2 = H; R3 = OH; R4 = H35816-31-0152.05[9]
107Protocatechuic acidC7H6O4XXXIR1 = COOH; R2 = OH; R3 = OH; R4 = H99-50-3154.12[9]
108Methyl veratrateC10H12O4XXXIR1 = COOCH3; R2 = OCH3; R3 = OCH3; R4 = H2150-38-1196.20[9]
109Benzoic acidC7H6O2XXXIR1 = COOH; R2 = H; R3 = H; R4 = H117500-35-3122.12[9]
110Veratric acidC9H10O4XXXIR1 = COOH; R2 = OCH3; R3 = OCH3; R4 = H93-07-2182.17[9]
111Vanillic acidC8H8O4XXXIR1 = COOH; R2 = OCH3; R3 = OH; R4 = H121-34-6168.14[8]
112Gallic acidC7H6O5XXXIR1 = COOH; R2 = OH; R3 = OH; R4 = OH149-91-7170.12[8]
1134-Hydroxy-2,6-dimethoxybenzaldehydeC9H10O4XXXIR1 = CHO; R2 = OCH3; R3 = OH; R4 = OCH322080-96-2182.17[8]
114MonotropeinC16H22O11XXXIInothing5945-50-6390.33[8]
115Ascorbic acidC6H8O6XXXIIInothing299-36-5176.13[8]
1163-(6-hydroxy-7-methoxy-2H-1,3-benzodioxol-5-yl)propanoic acidC11H12O6XXXIVnothingnothing240.06[8]
117(2R,3S)-Piscidic acidC11H12O7XXXVnothing469-65-8256.06[8]
118Isochlorogenic acid AC25H24O12XXXVInothing2450-53-5516.46[8]
119TecominC15H20O9XXXVIInothing31002-27-4344.31[8]
120Shikimic acidC7H10O5XXXVIIInothing138-59-0174.15[8]
121Phlorizin dihydrateC21H26O11XXXIXnothing7061-54-3454.43[8]
122Globeflowery acidC13H16O4XLnothing4041-28-5236.26[8]
123Ursolic acidC30H48O3XLInothing77-52-1456.71[8]
124SalviaflasideC24H26O13XLIInothing178895-25-5522.46[8]
125RhynchophyllineC14H6O8XLIIInothing76-66-4302.19[8]
126Methyl dodecanoateC13H26O2XLIVR = H111-82-0214.34[53]
127Methyl tridecanoateC14H2802XLIVR = CH2CH31731-88-0228.37[53]
128Methyl tetradecanoateC15H30O2XLIVR = (CH2)3CH3124-10-7242.40[53]
129Methyl pentadecanoateC16H32O2XLIVR = (CH2)4CH37132-64-1256.42[53]
130Methyl hexadecanoateC17H34O2XLIVR = (CH2)7CH112-39-0270.45[53]
131Methyl heptadecanoateC18H36O2XLIVR = (CH2)8CH31731-92-6284.47[53]
132Methyl octadecanoateC19H38O2XLIVR = (CH2)9CH3112-61-8298.50[53]
133Methyl eicosanoateC21H42O2XLIVR = (CH2)11CH322589-04-4326.55[53]
134Methyl docosanoateC23H46O2XLIVR = (CH2)13CH3929-77-1354.61[53]
135Methyl tetracosanoateC25H50O2XLIVR = (CH2)15CH32442-49-1382.66[53]
136Methyl decanoateC11H22O2XLIVR = CH2CH3110-42-9186.29[53]
137Methyl octanoateC9H18O2XLIVR = H111-11-5158.23[53]
138Methyl benzeneacetateC9H10O2XLVnothing143390-89-0150.17[53]
139Methyl benzoateC8H8O2XLVIR1 = H, R2 = H36712-21-7136.15[53]
1403,4-dimethoxybenzoic acid methyl esterC10H12O4XLVIR1 = OCH3; R2 = OCH32150-38-1196.2[53]
141Dimethyl 3-hydroxy-2-methyl-glutarateC8H1405XLVIInothingnothing190.19[53]
142N-hexadecanoic acidC16H32O2XLVIIIR1 = H; R2 = H57-10-3256.42[53]
143Hexadecanoic acid,10,16-dihydroxyC16H32O4XLVIIIR1 = OH; R2 = OH3233-90-7288.42[53]
144Dimethyl butanedioateC6H10O4XLIXnothing106-65-0146.14[53]
145Dimethyl octanedioateC10H18O4Lnothing1732-09-8202.24[53]
146Dimethyl nonanedioateC11H26O4LInothing1732-10-1216.27[53]
1472-hydroxyhexadecanoic acid methyl esterC17H34O3LIIR1 = OH; R2 = H; R3 = H78330-57-1286.45[53]
1483-hydroxyhexadecanoic acid methyl esterC17H34O3LIIR1 = H; R2 = OH; R3 = H51883-36-4286.45[53]
14910-hydroxyhexadecanoic acid methyl esterC17H34O3LIIR1 = H; R2 = H; R3 = OH56247-30-4286.45[53]
1502-hydroxy-benzaldehyde oximeC7H7NO2LIIInothing94-67-7137.14[53]
1514-hydroxy-acetophenoneC8H8O2LIVnothing99-93-4136.15[53]
1522-methoxydocosyl methanoateC25H50O3LVnothingnothing398.66[53]
1533-phenylprop-2-enoic acid methyl esterC11H12OLVIR1 = H; R2 = H103-26-4160.00[53]
1543-(4-hydroxyphenyl)prop-2-enoic acid methyl esterC10H10O3LVIR1 = H; R2 = OH61240-27-5178.00[53]
155(4-hydroxy-3-methoxyphenyl)-2-propenoic acid methyl esterC11H12O4LVIR1 = OCH3, R2 = OH34298-89-0208.00[53]
1562,3-DihydrobenzofuranC8H8OLVIInothing496-16-2120.15[53]
1574-phenyl-2-butenoic acid methyl esterC11H12OLVIIInothing54966-43-7176[53]
1589-(propoxybenzene)-nonanoic acid methyl esterC19H30O2LIXnothingnothing290[53]
159(E)-11-eicosenoic acid methyl esterC21H40O2LXnothingnothing324[53]
160(Z)-9-hexadecenoic acid methyl esterC17H32O2LXIR = (CH2)4CH31120-25-8268.43[53]
161(Z)-9-octadecenoic acid methyl esterC19H36O2LXIR = (CH2)6CH3112-62-9296.48[53]
162(Z,Z)-9,12-octadecadienoic acid methyl esterC19H32O4LXIR = CH = CH(CH2)4CH3168482-44-8294.47[53]
163(Z,Z,Z)-9,12,15-Octadecatrienoic acid methyl esterC19H32O2LXIR = CH = CHCH2CH = CHCH2CH3301-00-8292.46[53]
164(Z,Z,Z)-9,12,15-ctadecatrien-1-olC18H32OLXIInothingnothing264[53]
Table 5. Alkaloids isolated from T. chinensis.
Table 5. Alkaloids isolated from T. chinensis.
NoNamesMolecular FormulaParent NucleusSubstituentCASMolecular WeightRefs.
165SenecionineC18H25NO5LXIIINothing130-01-8335.4[56]
166IntegerrimineC18H25NO5LXIVNothing480-79-5335.4[46]
167TrollineC12H13NO3LXVNothing1021950-79-7219.24[9]
168(R)-Cyanomethyl-3-hydroxyindoleC10H7O2N2LXVINothingNothing187.05[46]
169AdenineC5H5N5LXVIINothing73-24-5135.13[8]
Table 6. Other chemical components isolated from T. chinensis.
Table 6. Other chemical components isolated from T. chinensis.
NoNamesMolecular FormulaParent NucleusSubstituentCharacterization MethodMolecular WeightRefs.
168DaucosterolC35H60O6LXVIIIInothing474-58-8576.85[8]
169Trolliusol AC17H16O6LXIXnothingnothing316.30[8]
170EsculetinC9H6O4LXXnothing305-01-1178.14[8]
171β-SitosterolC29H50OLXXInothing5779-62-4414.71[8]
172TrolliamideC42H82NO5LXXIInothingnothing680.62[11]
173L-RhamnoseC6H14O6LXXIIInothing6155-35-7182.17[8]
174L-ArabinoseC5H10O5LXXIVnothing5328-37-0150.13[8]
175D-GalactoseC6H12O6LXXVnothing59-23-4180.15[8]
176VanillylamineC8H11NO2LXXVInothing1196-92-5153.18[8]
1772-(3,4-Dihydroxyphenyl)ethyl-O-β-d-pyranoglucoseC8H10O3LXXVIIR = H10597-60-1154.16[8]
178Homovanillyl alcoholC9H12O3LXXVIIInothing2380-78-1168.19[8]
1792-(3,4-dihydroxyphenyl)-ethyl-O-β-d-glucopyranosideC14H20O8LXXIXnothingnothing315.10[8]
1803,5-dihydroxyphenethyl alcohol 3-0-β-d-glucopyranosideC14H20O8LXXXnothing52674-86-9315.10[8]
1814′-O-(6″-O-Vanilloylajugol-β-d-glucopyranoyl)phenylethanolC22H26O10LXXXInothing27606-08-2450.44[8]
182Xantho-phyll-epoxydeC40H56O3LXXXIInothingnothing584.87[64]
183TrollixanthinC40H56N4LXXXIIInothing14660-91-4592.90[64]
Table 7. Main pharmacological effects of TC.
Table 7. Main pharmacological effects of TC.
Pharmacological EffectsExtracts/CompoundsAnimals/CellsDosage/ConcentrationEffects/MechanismsReferences
AntiviralThe crude extract from the flowers of T. chinensisICR mice0.2 mg/g/dThe T. chinensis crude extract treatment resulted in a significant increase in the body weight percentage, a decrease in the number of white blood cells, and a lowered lung index among mice infected with influenza virus A/FM/1/47 (H1N1) virus.[115]
OrientinHep 2 cell0.1 mL of maintenance medium containing serial two-fold dilutions of the tested compounds
0.1 mL of maintenance medium without the test compound was added
The flavonoids isolated from T. chinensis, Orientin, and Vitexin, possess strong anti-viral activities against Para 3. Proglobeflowery acid showed weak antiviral activity against Para 3.[10]
Vitexin
Proglobeflowery acid
The crude extract from the flowers of T. chinensisICR mice0.2 mg/g/dThe crude extract from the flowers of T. chinensis was found to inhibit the increased expression of TLR3, TBK1, TAK1, and IRF3 induced by the high-dose influenza virus and treat mice infected with influenza virus by activating the TLR3 signaling pathway.[1]
Veratric acidRAW264.7 cell50, 100, 200, 400, and 800 μmol/LThe three representative compounds play a role in anti-H1N1 viral effects by regulating the TLR 3, 4, and 7 pathways, counteracting the inflammatory damage caused by excessive production of NO, IL-1, IL-6, and TNF induced by viral infection, and promoting the production of IFN- to eliminate the virus.[116]
Vitexin
Trolline
Piscidic acidEV71-infected RD cellsThe T. chinensis mother liquor was diluted by a factor of 20, 2−1, 2−2, 2−3, 2−4, and 100 μL was dispensed into each well of the cell culture plateThe viral inhibition rate of T. chinensis ranges from 49.64% to 73.69%. It exhibits a clear inhibitory effect on the EV71 virus, and the three compounds form the foundation of T. chinensis’ anti-EV71 material.[25]
2″-Oacetylorientin
2-(4-hydroxybenzyl) malic acid
AntioxidantOrientinNot applicable46/5.64/5.19/3.97 mg/mLUnder in vitro conditions, phenolic and flavonoid compounds could efficiently scavenge a variety of ROS or DPPH-free radicals.[117]
Vitexin
Proglobeflowery acid
OrientinKM mice40/20/10 mg/kgSlowing down d-galactose-induced aging by enhancing the activity of antioxidant enzymes, eliminating excessive oxygen free radicals, and mitigating damage to cells and tissues.[118]
Vitexin
AntiinflammatoryTrolliusditerpenosides A-Q (1–17)RAW 264.7 cells mediated by LPSNot applicableThe inhibitory effects on LPS-induced NO (pro-inflammatory mediator nitric oxide) release in RAW 264.7 cells by diterpenoid glycosides from T. chinensis.[14]
OrientinRAW264.7 cells0, 25, 50, 100, 200, and 400 µmol/L−1The production of NO, IL-6, and TNF-stimulated cells decreased.[119]
Vitexin
Quercetin
Isoquercetin
Veratric acid
Proglobeflowery acid
Trollioside
2″-O-β-l-galactopyranosy-orientin
LuteolinRAW264.7 cells0, 12.5, 25, 50, 100, 200, and 400 µmol·L−1The production of NO, IL-6, and TNF-stimulated cells decreased.[119]
Trolline
Aqueous extract of the stem and leaves of T. chinensisKM miceDistilled water: 20 mL/kg
Positive drug: 100 mg/kg
Aqueous extract of stem and leaves of T. chinensis (low/high): 12 g/kg/24 g/kg
Alcohol extract of the stem and leaves of T. chinensis (low/high): 12 g/kg/24 g/kg
T. chinensis has some anti-inflammatory effects on stem and leaf extracts.[111]
Alcohol extract of the stem and leaves of T. chinensis
AntitumorTotal flavonoidsMCF-7 cells0/0.0991/0.1982/0.3964/0.7928/1.5856 mg/mLFlavonoids were found to suppress growth and induce apoptosis in MCF-7 cells.[93]
OrientinEC-109 cells5.0, 10.0, 20.0, 40.0, and 80.0 µMOrientalin and Vitexin reduce apoptosis in human esophageal cancer EC-109 cells by regulating oncogenes and tumorigenic genes.[120]
Vitexin
AntibacterialTrolliusol AM. albicans
E. coli
P. aeruginosa
B. subtilis
S. aureus
Drug concentrations: 1:4, 1:8, 1:16, 1:32, 1:64, 1:128, 1:256, 1:512, 1:1024, 1:2048, and 1:4096Minimal Inhibitory Concentration (MIC) was achieved by the microbroth method to achieve inhibition efficiency.[41]
1-(3′,4′-dihydroxyphenyl)-6,7-dihydroxyisochroman
(S)-1-(3′,4′-dihydroxyphenyl)-1-hydroxypropan-2-one
3,4-dihydroxyphenylethanol
2″-O-(2‴-methylbutyryl)isoswertisin
3″-O-(2‴-methylbutyryl)isoswertisin
Isoswertisin
Orientin
Water extracts from T. chinensisMicroorganism S. mutans50/25/12.5/6.25/3.125 (mg/mL)T. chinensis has antibacterial and anti-inflammatory effects and can be used against mutant baculoviruses. Thirty percent ethanol extractexhibited the best antibacterial and antibiofilm effects.[121]
30% ethanol extracts fromT. chinensis
60% ethanol extracts from T. chinensis
90% ethanol extracts from T. chinensis
AntiagingOrientinKM mice40/20/10 mg/kgIt can enhance the activity of antioxidant enzymes, eliminate excessive oxygen-free radicals, and reduce the damage to cells and tissues so as to delay the senescence caused by D-galactose.[118]
Vitexin
AntipyreticFlavonoidsNew Zealand rabbitsFlavonoids 200 mg·kg−1 group
Flavonoids 100 mg·kg−1 group
Flavonoids 50 mg·kg−1 group
Asprin 100 mg·kg−1 group
By inhibiting the expression of TNF-α and IL-1β in serum and PGE2 in cerebrospinal fluid.[110]
AnalgesicAqueous extract of stem and leaves of T. chinensisKM miceDistilled water: 20 mL/kg
Positive drug: 100 mg/kg
Aqueous extract of stem and leaves of T. chinensis (low/high): 12 g/kg/24 g/kg
Alcohol extract of the stem and leaves of T. chinensis (low/high): 12 g/kg/24 g/kg
T. chinensis extracts from stems and leaves have been shown to have some analgesic effects.[111]
Alcohol extract of stem and leaves of T. chinensis
The total flavones in T. chinensisKM mice125, 250, and 2500 mg/kgIt may increase the pain threshold of the hot plate in mice and have analgesic effects.[112]
Antitussive and ExpectorantThe total flavones in T. chinensisKM mice125, 2250, and 2500 mg/kgThe total flavonoid extract of T. chinensis has obvious anti-tussive and expectorant effects.[112]
Myocardial ischemia/reperfusion injury (MI/RI)The total flavones in T. chinensisSD rats50 mg/(kg·d)–100 mg/(kg·d)The total flavones in T. chinensis protect the myocardium from MI/RI.[113]
Not applicable means not described in detail in the literature.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

He, L.; Wang, Z.; Lu, J.; Qin, C.; He, J.; Ren, W.; Liu, X. Trollius chinensis Bunge: A Comprehensive Review of Research on Botany, Materia Medica, Ethnopharmacological Use, Phytochemistry, Pharmacology, and Quality Control. Molecules 2024, 29, 421. https://doi.org/10.3390/molecules29020421

AMA Style

He L, Wang Z, Lu J, Qin C, He J, Ren W, Liu X. Trollius chinensis Bunge: A Comprehensive Review of Research on Botany, Materia Medica, Ethnopharmacological Use, Phytochemistry, Pharmacology, and Quality Control. Molecules. 2024; 29(2):421. https://doi.org/10.3390/molecules29020421

Chicago/Turabian Style

He, Lianqing, Zhen Wang, Jiaxin Lu, Chen Qin, Jiajun He, Weichao Ren, and Xiubo Liu. 2024. "Trollius chinensis Bunge: A Comprehensive Review of Research on Botany, Materia Medica, Ethnopharmacological Use, Phytochemistry, Pharmacology, and Quality Control" Molecules 29, no. 2: 421. https://doi.org/10.3390/molecules29020421

Article Metrics

Back to TopTop