Next Article in Journal
Recent Progress on Perovskite-Based Electrocatalysts for Efficient CO2 Reduction
Previous Article in Journal
On the Question of Zwitterionic Intermediates in the [3+2] Cycloaddition Reactions between Aryl Azides and Ethyl Propiolate
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Review on the Morphology, Cultivation, Identification, Phytochemistry, and Pharmacology of Kitagawia praeruptora (Dunn) Pimenov

by
Qi Wang
,
Lulu Ding
,
Ruihong Wang
* and
Zongsuo Liang
*
Zhejiang Province Key Laboratory of Plant Secondary Metabolism and Regulation, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
*
Authors to whom correspondence should be addressed.
Submission received: 27 November 2023 / Revised: 11 December 2023 / Accepted: 11 December 2023 / Published: 18 December 2023

Abstract

:
Kitagawia praeruptora (Dunn) Pimenov, commonly known as Qianhu in China, is a widely used folk Chinese herbal medicine. This article reviews its botanical traits, ethnopharmacology, cultivation techniques, identification, phytochemical compositions, and pharmacological effects. Over 70 coumarin compounds, including simple coumarins, pyranocoumarins, and furanocoumarins, have been isolated within this plant. Additionally, K. praeruptora contains other components such as flavonoids, fatty acids, benzoic acids, and sterols. This information highlights the importance of utilizing active ingredients and excavating pharmacological effects. With its remarkable versatility, K. praeruptora exhibits a wide range of pharmacological effects. It has been found to possess expectorant and bronchodilator properties, cardiovascular protection, antimicrobial and antioxidant activities, anti-tumor effects, and even antidiabetic properties. It is recommended to focus on the development of new drugs that leverage the active ingredients of K. praeruptora and explore its potential for new clinical applications and holistic utilization.

Graphical Abstract

1. Introduction

Kitagawia praeruptora (Dunn) Pimenov (= Peucedanum praeruptorum Dunn), also known as “Qianhu” (前胡) in Chinese, belongs to the Apiaceae family and is a perennial herb that is widely utilized in traditional Chinese medicine (TCM) formulations with a domestication history of more than 1500 years [1]. The classification of Kitagawia has been a topic of controversy for a long time, and its delineation and morphological delimitation is not well-defined. Despite the creation of Kitagawia, numerous scientists still classify certain species as part of Peucedanum sensu lato, and Kitagawia has been considered a synonym of Peucedanum sensu lato [2,3,4,5]. Until 2017, Pimenov had examined the Chinese Apiaceae-type specimens and proposed five new nomenclatural combinations for Kitagawia [6]. At present, there are ten known species within Kitagawia, with eight species native to China [7]. The dried roots of K. praeruptora are recorded as Peucedani Radix in Chinese Pharmacopoeia for reducing fever and resolving phlegm and commonly used to treat various respiratory disorders, including anemopyretic cold, cough with abundant phlegm, and congested chest [8]. Over the past few years, the medicinal benefits and economic worth of K. praeruptora, as well as the specific environmental conditions required for its cultivation, combined with excessive human exploitation have made it a highly prized and valuable species in China [9]. It is artificially cultivated for its roots on a large scale in several Southeastern and Mid-China regions including Zhejiang, Anhui, Hubei, and Jiangxi for its unique medicinal value, especially in the authentic production area of Zhejiang Province. K. praeruptora was included in the list of one of the famous authentic medicinal materials “Zhebawei” announced by multiple departments in Zhejiang Province in 2018. Chun’an, Lishui, and other places in Zhejiang Province are traditional authentic cultivation areas of K. praeruptora and currently have a cultivation area of over 12,000 acres, with an annual output of more than 2000 tons, accounting for 15% of the national total.
We introduce the research achievements from various perspectives in recent years both domestically and internationally as a comprehensive review article. In the beginning, we provide a brief overview of the botanical characteristics and traditional uses. Subsequently, we delve into topics of cultivation and identification methods including early bolting issues. Furthermore, we discuss numerous isolated compounds and pharmacological activities. Lastly, we share our insights regarding the potential future development prospects of K. praeruptora. To the best of our knowledge, there has been no comprehensive review of all aspects of K. praeruptora. Thus, we aim to enhance the understanding of this neglected plant and provide valuable evidence for the rational use of K. praeruptora as a traditional medicine through this paper.
To conduct a comprehensive literature review, we initially defined the review topic. Our search strategy first involved exploring diverse scientific databases such as Google Scholar, PubMed, Springer Link, ScienceDirect, ScienceFinder, Web of Science, Baidu Scholar, and the Chinese National Knowledge Infrastructure. Employing specific keywords and search terms, we narrowed our focus to five key aspects of K. praeruptora (Peucedanum praeruptorum): morphological characteristics, cultivation methods, physicochemical identification, compounds isolation, and pharmacology. Furthermore, we set a specific time frame for the literature search, extending up to the end of February 2023. Furthermore, the criteria guiding the inclusion and exclusion of studies encompass several factors: relevance to the research topic, academic credibility with a preference for literature from peer-reviewed academic journals, conference papers, professional books, and methodological diversity. Specifically, we sought diversity in chemical composition and pharmacological effects, including variations in compound isolation methods, animal modeling, and cytokine detection. Finally, we ensured the exclusion of multiple versions of the same study in our review.

2. Botany

K. praeruptora is a perennial herb that grows up to 1 m in height and has a solitary stem that branches above with puberulous branchlets. The basal leaf blade is triangular-ovate and 2–3-ternate, with long-petiolulate pinnae and pinnules that are long-ovate, 3–5-lobed, and measure 1.5–6 × 1.2–4 cm (Figure 1). Both surfaces of the leaves are glabrous, occasionally puberulous, with prominent abaxial nerves, a cuneate base, irregular serrate margins, and an acuminate apex. The 8–12 ovate-lanceolate bracteoles are shorter than the flowers and have rough-puberulous surfaces, with 15–20-flowered umbellules. The calyx teeth are obsolete, the petals are white, and the styles are short. The fruit is ovoid and measures approximately 4 × 3 mm, with sparse pubescence. The root is conical with forked end. The entire growth cycle of K. praeruptora can be categorized into three stages: seedling stage, vegetative growth stage, and reproductive growth stage [10]. During the period of reproductive growth, the pregnancy bud stage is generally around mid-June, the flower time is about two months long, usually from August to September, and the fruit time is from October to November [3]. Wild populations of K. praeruptora are widely distributed throughout Central and Eastern regions of China, westward to Sichuan and Guizhou, stretching northward to Henan and Gansu, as well as southward to Guangxi with the preference for habitat of forest margins and grassy slopes at the altitude of 250–2000 m [11] (Figure 1E). Northwestern Zhejiang, southeastern Anhui, and northeastern Jiangxi are authentic production areas of K. praeruptora. Root of K. praeruptora produced in Anhui and Zhejiang is commonly known as “Ningqianhu” and “Chunqianhu”, which accounts for about 80% of the market share of Chinese medicinal materials. The root of K. praeruptora produced in Hubei, Hunan, Guizhou, and other places is called “Xinqianhu”, which accounts for about 20% of the market share [10].

3. Ethnobotany

K. praeruptora is a traditional medicine commonly used in China with a long history of application and has been recorded in many ancient books and modern pharmacopeia [12]. It was first recorded in Ming Yi Bie Lu and Ben Cao Jing Ji Zhu, resolving sputum and relieving asthma. During the Tang and Song dynasties, it was described in many other medical books, such as Xin Xiu Ben Cao, Ben Cao Tu Jing, and Zheng Lei Ben Cao, which were mainly used for the treatment of making expectoration easy. In the Ming and Qing dynasties, it was used for the treatment of typhoid fever, which was recorded in Ben Cao Meng Quan and Ben Cao Feng Yuan. In Ben Cao Gang Mu, a classic book written by Shizhen Li, it was recorded to treat vomiting and regurgitation as well as reducing fever and resolving phlegm.

4. Cultivation

K. praeruptora is suitable for a cool and moist climate and possesses biological characteristics such as cold resistance, heat resistance, drought resistance, barren soil resistance, and disease and insect resistance [13]. For a long time, materials of K. praeruptora medicinal herbs have relied on wild resources collection without classification of commodity specifications and grades, and non-renewable harvesting has led to a sharp decline in the reserves of wild K. praeruptora resources [14]. Due to its high medicinal value and large market demand, the cultivation area of K. praeruptora is constantly increasing. Currently, there are mainly four types of cultivation modes for K. praeruptora, including wild planting, simulated wild planting on barren slopes, planting on newly opened forest land, and field planting [15]. The cultivation technology research focusing on K. praeruptora site selection and land preparation, breeding and planting, field management, harvesting and processing, and storage have attracted the attention of many scholars [16]. Current studies proved that the cultivation mode has the greatest impact on the content of coumarin of K. praeruptora [17]. K. praeruptora grown in wild environments such as valley streams or sunny sparse forest was reported to take several years to produce flowering stems, but artificially cultivated K. praeruptora can produce flowering stems in the second year [18].
In recent years, the planting scale of K. praeruptora has been continuously expanding nationwide; the problems of early bolting, flowering, lignification, and the content of active ingredients in K. praeruptora below standard greatly affect the production of K. praeruptora [19]. Early bolting refers to the process of transitioning from vegetative growth to reproductive growth by blooming before the vegetative mass is fully developed. Early bolting has always been a key factor that severely restricts the production and industrialization of Apiaceae medicinal herbs, including Angelica dahurica (Fisch. ex Hoffm.) Benth. et Hook. f. ex Franch. e, Saposhnikovia divaricata (Turcz.) Schischk. [20], Angelica sinensis (Oliv.) Diels [21,22], and K. praeruptora [23]. During early bolting, endogenous hormones will change to regulate growth and development, which has a greater impact on the root structure and coumarin content of K. praeruptora. After bolting, the cross-sectional area of secondary xylem in the root is greatly increased, and there are many wood fibers and vessels (Figure 2). The pericycle and thin-walled cross-sectional area of tissue and phloem decrease sharply, and the content of coumarin components decrease significantly [24]. The roots of K. praeruptora that have gone through early bolting often lose their medicinal efficacy and cannot be used as medicinal materials because the content of coumarin components is greatly reduced and does not meet the standards of pharmacopoeia [25]. Therefore, due to the influence of genetics, ecological environment, and other factors, the vegetative growth and reproductive growth of different individuals in the same period are inconsistent, and early bolting has become one of the key issues that seriously restrict the production of traditional Chinese medicine. Effective control of bolting can significantly improve the medicinal quality and yield of K. praeruptora.
To decrease the rate of early bolting and increase the content of effective ingredients of K. praeruptora, high altitude (900–1200 m) and shading degree of 60% was suitable for K. praeruptora [26]. The change of altitude gradient in the same area will cause changes in various environmental factors such as light, humidity, and temperature and directly affect the physiological and biochemical structure and quality. There was a big difference in yield of K. praeruptora through different treatment methods; the highest yield of roots was achieved by using silver film covering and float bed seedling technology, and the highest net income was achieved by using silver film covering alone [19]. Many field management experiments mentioned that topping and pruning 3–4 times a year can reduce the occurrence of flowering stems and increase yield, but there are no specific data to support this in the literature.
Most current studies were mainly focused on cruciferous vegetables and large vegetables with straw properties, such as Brassica oleracea L. [27], Allium cepa L. [28], and Brassica rapa L. [29]. The phenomenon of bolting is primarily a quantitative trait that is controlled by multiple genes and regulated by various pathways of environmental signals and endogenous factors [30]. Numerous molecular markers or quantitative trait locus (QTL) loci [31] linked to bolting and flowering traits have been discovered in cruciferous vegetables, and there is a considerable collinearity in genome arrangement with significant variations in the copy number and gene structure of homologous genes [32,33]. Consequently, the genetics and molecular regulatory mechanisms of bolting and flowering in cruciferous crops have always been a hot topic. The process of flowering induction of the model plant Arabidopsis thaliana (L.) Heynh. has gradually become clear (Figure 3). It contains several key genes that control flowering [34,35]: CONSTANS (CO), LEAFY (LFY), FRIGIDA (FRI), FLOWERING LOCUS C (FLC), and VERNALIZAION (VRN). But there is still a lack of systematic study in this area. At present, this study has confirmed that some exogenous hormones have a certain inhibitory effect on the early bolting of plants and at the same time can improve the quality of medicinal materials, which has been confirmed in plants such as A. sinensis [36], A. dahurica [37], and B. rapa [38]. In the study of genes related to bolting in K. praeruptora, researchers employed a combination of transcriptome sequencing and co-expression analysis to elucidate the regulatory mechanisms of key genes involved in coumarin synthesis and the formation of other active ingredients before and after bolting [39,40]. The results revealed that key genes in the phenylpropanoid pathway (4-coumarate-CoA ligase (4CL) and hydroxycinnamoyl transferase (HCT)), ATP-binding cassette (ABC) transporters, apoptosis-related genes, and genes involved in circadian rhythm regulation (cryptochrome circadian regulator (CRY), GIGANTEA (GI), phytochrome A (PHYA), and phytochrome-interacting factors 3 (PIF3)) may play crucial roles in regulation.

5. Identification

At present, the main identification techniques of traditional Chinese medicine include sensory evaluation, microscopic identification [41], physical and chemical identification, and molecular identification [42]. One of the traditional identification methods, thin-layer chromatography, can distinguish K. praeruptora, Angelica decursiva (Miquel) Franchet & Savatier, and S. divaricata through the number of thin-layer fluorescent spots, color, and Rf value [43]. Fourier Transform Infrared Spectroscopy (FTIR) [44] can not only quickly identify K. praeruptora and its counterfeit products but also speculate that there are differences in the composition and content of organic ester compounds, aromatic compounds, and glycoside compounds contained according to the map information [45]. Handheld near-infrared spectroscopy (NIRS) combined with chemical pattern recognition techniques is employed to identify K. praeruptora and its counterfeit products based on absorbance in the spectral region of 1405–2442 nm and standard normal variate (SNV) pretreatment [46]. In addition, the utilization of headspace-gas chromatography-ion mobility spectrometry (HS-GC-IMS) in combination with principal component analysis (PCA) [47] and other multivariate analysis methods to examine the volatile components of K. praeruptora during different harvest periods is an efficient and quantifiable approach for creating a fingerprint atlas, which aids in the identification of plants [48].
Modern molecular DNA barcoding technology is widely used in the identification of K. praeruptora and counterfeit species [49,50]. The use of random amplified polymorphic DNA (RAPD) and inter simple sequence repeat (ISSR) genetic diversity analysis, as well as internally transcribed spacer (ITS) sequencing [8,51,52], allow for the detection of interspecies differences and molecular analysis of botanicals. First, according to the RADP polymorphism of the genomic DNA, at the threshold of 0.66, A. decursiva and K. praeruptora were divided into two categories, and there were also obvious differences in the amplified bands [53]. Second, the ISSR molecular marker technique can be used to analyze the genetic diversity of K. praeruptora from different origins, and it was found that the percentage of polymorphic bands was 95.1% and the genetic similarity coefficient was between 0.201–0.822 [54]. In addition, due to the wide distribution of test sequence information sites and variation sites [55,56,57], the ITS sequence is an ideal DNA barcode to identify K. praeruptora, A. decursiva, and adulterants [58]. The specific PCR primer pairs QH-CP19s/QH-CP19a and ZHQH-CP3s/ZHQH-CP3a were designed for K. praeruptora and A. decursiva, respectively, which was a quick and effective method to distinguish between them [59]. DNA barcoding technology based on ITS4 and ITS5 sequences can successfully identify 13 regional substitutes and 23 adulterants of K. praeruptora [8]. Some scholars have also found a specific primer region for identifying camphor in A. sylvestris, K. praeruptora, and Anthriscus sylvestris (L.) Hoffm. through the comparison of rDNA-ITS sequences, and the primer region has produced a 273 bp camphor-specific DNA chain to A. sylvestris. As a result of the RAPD analysis, a Sequence Characteristic Amplified Region (SCAR) marker is developed, and PCR combined with primers can be used to establish a SCAR marker that could simultaneously identify the three plants [54]. The advent of DNA molecular identification technology has led to the discovery of numerous potential DNA barcodes, thereby providing additional feasible identification sites for the species identification of K. praeruptora.
With the development of high-throughput techniques, the use of complete chloroplast genomes as one super-barcode should bring better resolution effects than the use of one or even several universal or specific DNA barcodes [60,61]. The complete chloroplast genome of K. praeruptora was generated using high-throughput sequencing [62]. Comprehensively comparative and phylogenomic analyses of plastid genomes for seven Peucedanum plants including K. praeruptora were performed; the results showed both conservation and diversity and supported the thesis that Peucedanum is not a monophyletic group [14]. These findings can serve as a basis for future studies on the taxonomy and evolution of Peucedanum.

6. Phytochemistry

The main chemical components of K. praeruptora include coumarin, volatile oil, phenanthrenequinone, organic acids, and sterols [63,64,65]. Coumarin was first isolated from the legume Dipteryx ordorata (Aubl.) Forsyth f., which can be found in many plants as secondary metabolites of the roots [66,67], stems [68], leaves [69], flowers, fruits, and seeds. Coumarins were reported to exhibit tremendous biological activities, such as anti-inflammatory, antioxidant, and antidiabetic actions. According to related reports, more than 70 kinds of coumarin compounds have been isolated in the root of K. praeruptora [70].
Modern pharmacological studies have found that the most important pharmacodynamic components of K. praeruptora are divided into simple coumarin, furanocoumarin and pyranocoumarin compounds [71,72,73]; the structural formula of some compounds is displayed in Figure 4. Phenylalamine ammonia lyase (PAL) in plants can convert L-phenylalanine into trans-cinnamic acid, which is activated by cinnamic acid 2-hydroxylase (cinnamic acid under the action of 2-hydroxylase, C2H) and cinnamic acid 4-hydroxylase (cinnamic acid under the action of 4-hydroxylase, C4H). The ortho-hydroxylation and para-hydroxylation reactions occur, which convert it into o-coumaric acid and p-coumaric acid. The p-hydroxylation reaction of trans-cinnamic acid is prior to the ortho-hydroxylation reaction [74]. If there is a lack of C4H in the chloroplast, C2H can convert cinnamic acid into o-coumaric acid [75]. The simple framework of the coumarin biosynthetic pathway is shown in Figure 5 [76,77,78].

6.1. Coumarins

According to literature reports, various methods have been employed for the qualitative analysis of chemical components in K. praeruptora. These methods include Nuclear Magnetic Resonance spectroscopy (NMR) [79], Mass Spectrometry (MS) [80], and Gas Chromatography-Mass Spectrometry (GC-MS) [81]. Additional techniques such as High-Performance Liquid Chromatography-Ultraviolet Detection (HPLC-UV), Nuclear Magnetic Resonance Spectroscopy (NMRS), and High-Performance Liquid Chromatography-Mass Spectrometry (HPLC-MS) have also been employed [82,83,84] (Table 1). Scholars first discovered two simple coumarin compounds, namely skimmin and scopolin, within K. praeruptora in 1989 [79]. To date, a total of eleven simple coumarin compounds have been identified from K. praeruptora, as listed in Table 1 (1–11), and some of these coumarins exhibit antitumor properties. For instance, scopolamine activates NF-κB and caspase-3, and resveratrol enhances the activity of Phase II enzymes glutathione S-transferase (GST) and quinone reductase (QR) while also inhibiting the expression of matrix metalloproteinases (MMP-7, -2 and MMP-9), aiming to achieve an antitumor effect [85]. Since the 1990s, researchers have conducted more systematic investigations into the active components of K. praeruptora [86,87]. The results indicate that coumarins are the main active constituents of K. praeruptora, and, until now, a total of 19 linear furanocoumarins and six angular furanocoumarins have been discovered (Table 1 (12–36)). From the 1970s to 1980s, researchers made the discovery that the primary active component of K. praeruptora is praeruptorin A (Pra-A), which has the highest content [88]. They also identified three other saponins derived from K. praeruptora: praeruptorin B (Pra-B), praeruptorin C (Pra-C), and praeruptorin D (Pra-D) [89,90]. Until now, more than 15 pyranocoumarin compounds have been separated from K. praeruptora (Table 1 (37–52)).

6.2. Other Constituents

K. praeruptora contains various components, including coumarin compounds, flavonoids, fatty acids, benzoic acids, and steroids. Flavonoid compounds were discovered in K. praeruptora for the first time, which were identified as 4H-1-benzopyran-4-one, 5-hydroxy-6-methoxy-2-phenyl-7-O-α-D-glucuronyl methyl ester and 4H-1-benzopyran-4-one, 5-hydroxy-6-methoxy-2-phenyl-7-O-α-D-glucuronyl acid [91]. Additionally, the volatile oil content in K. praeruptora is relatively high, which consists of alkanes, esters, ketones, and sesquiterpenes (Table 1 (53–70)). Among them, α-pinene, hinokitol, aromadendrene, terpinolene, α-farnesene, and longifolene account for more than 60% of the relative composition of the volatile oil in K. praeruptora [92]. These components, being the main constituents, can also serve as quality control standards for the oil’s analysis and assessment.
Table 1. Main chemical compounds isolated from K. praeruptora.
Table 1. Main chemical compounds isolated from K. praeruptora.
ClassificationNo.Chemical ComponentsFormulaReferencesTest MethodsCAS Identifiers
Simple coumarin1ApiosylskimminC20H24O12[26]UPLC-HR-Orbitrap-MS103529-94-8
2Eleutheroside B1C17H20O10[93]NMR/MS16845-16-2
3HymexelsinC21H26O13[94]HPLC117842-09-8
4IsofraxidinC11H10O5[95] 486-21-5
5IsoscopoletinC10H8O4[96]LC-MS776-86-3
6ScopoletinC10H8O4[96]LC-MS92-61-5
7ScopolinC16H18O9[97]UPLC-Q-TOF-HRMS531-44-2
8SkimminC15H16O8[97]HPLC93-39-0
9UmbelliferoneC9H6O3[77]DMR93-35-6
108-carboxy-7-hydroxy coumarinC11H8O5[98]NMR5112-55-0
11(−)-peucedanolC15H18O5[99]HPLC/PAD28095-18-3
Furanocoumarin compounds12AngelicinC11H6O8[80]HPLC/NMR/MS523-50-2
13ArnocoumarinC14H10O3 11037-15-3
14ApterinC20H24O10[100]LC-MS53947-89-0
15BergaptenC12H8O4[101]HPLC484-20-8
16ImperatorinC16H14O4[102]IMP482-44-0
17IsopimpinellinC13H10O5[103]HPLC482-27-9
18IsorutarinC16H21NO3[79]NMR53846-51-8
19MarmesinC14H14O4[104]NMR/MS13849-08-6
20MarmesininC20H24O9[79]NMR27497-13-8
21Marmesin-11-O-β-D-glucopyranosyl (1→6)-β-D-glucopyranoside [104]NMR/MS
22NodakenetinC14H14O4[89]NMR/MS495-32-9
23Nodakenetin tiglateC19H20O5[105]HPLC106974-21-4
24NodakeninC26H34O14[106]UPLC495-31-8
25OroselolC14H12O4[90]HPLC1891-25-4
26OxypeucedaninC16H14O5[91]NMR737-52-0
27Oxypeucedanin hydrateC16H16O16[91]NMR133164-11-1
28Peucedanoside AC20H22O10[107]NMR946122-87-8
29Peucedanoside B [107]NMR
30Praeroside I [79]NMR
31Praeroside VII [108]NMR
32PsoralenC11H6O3 66-97-7
33Qianhucoumarin G [109]NMR
34RutarinC20H24O10[79]NMR20320-81-4
35SphondinC12H8O4[110]HPLC483-66-9
36XanthotoxinC12H8O4[101]HPLC298-81-7
Pyranocoumarin compounds37Decursinol angelateC19H20O5[82]UHPLC-MS/MS130848-06-5
38Decursitin DC19H20O6[111]HPLC245446-61-1
39d-LaserpitinC19H20O6[112]UPLC-MS/MS134002-17-8
40Longshengensin AC21H22O7[113]NMR/MS
41MarmesinC14H14O4[97]UPLC-Q-TOF-HRMS13849-08-6
42Peucedanocoumarin IIC21H22O7[114]NMR130464-56-1
43Praeruptorin AC21H22O[115]HPLC/LC–MS/MS73069-25-7
44Praeruptorin BC24H26O[73]HPLC/LC–MS/MS81740-07-0
45Praeruptorin CC24H28O7[116]HPLC/LC–MS/MS72463-77-5
46Praeruptorin DC24H26O[117]HPLC
47Praeroside IIC20H24O10[118]UPLC-HR-Orbitrap-MS86940-46-7
48PteryxinC21H22O7[119]NMR13161-75-6
49SelinidinC19H20O5[74]UPLC-MS/MS19427-82-8
50SuksdorfinC21H24O7[120]HPLC53023-17-9
51(+)-samidinC21H22O7[74]HPLC477-33-8
Other constituents52AcetylatractylodinolC15H12O3[111]NMR61582-39-6
53AdenosineC10H13N5O4[121]NMR58-61-7
54AromadendreneC15H24[122]GC/MS109119-91-7
55BaihuaqianhuosideC6H12O6[86]NMR155969-61-2
56Butyric acidC4H8O2[93]NMR107-92-6
57DaucosterolC35H60O6[87]NMR74-58-8
58GalactitolC6H14O6[87]NMR608-66-2
59Gallic acidC7H6O5[86]NMR149-91-7
60HinokitolC10H12O2[123]GC/MS499-44-5
61LongifoleneC15H24[124]GC/MS475-20-7
62MannitolC6H14O6[125]HPLC69-65-8
63Palmitic acidC16H32O2[126]NMR57-10-3
64Tanshinone IC18H12O3[93]NMR/MS568-73-0
65Tanshinone IIAC19H18O3[93]NMR/MS568-72-9
66Tetracosanoic acidC24H48O2[126]NMR/MS302912-17-0
67TerpinoleneC10H16[127]HS-SPME/GC-MS586-62-9
68Vanillic acidC8H8O4[86]NMR121-34-6
69α-farneseneC15H24[128]HS-SPME/GC-MS502-61-4
70α-PineneC10H16[129]HS-SPME/GC-MS7785-26-4
71β-sitosterolC29H50O[87]NMR
722,6-dimethyl quinolineC11H11N[126]NMR/MS877-43-0
734H-1-benzopyran-4-one,5-hydroxy-6-methoxy-2-phenyl-7-O-α-D-glucuronyl acid [91]Reverse-phase silica gel column
744H-1-benzopyran-4-one,5-hydroxy-6-methoxy-2-phenyl-7-O-α-D-glucuronyl methyl ester [91]Reverse-phase silica gel column
759,10-dihydrophenanthrinic acid [130]NMR
76(−)-sclerodinC18H16O6[126]NMR104855-18-7

7. Pharmacology

K. praeruptora represents a gentle Chinese herbal remedy known for its diverse pharmacological effects and demonstrates positive therapeutic efficacy against certain diseases (Table 2). Its main functions include eliminating phlegm and relieving asthma, providing cardiovascular protection, antibacterial and anti-oxidation effects, as well as anti-tumor and anti-diabetes functions (Figure 6).

7.1. Eliminate Phlegm and Relieve Asthma

According to the 2020 edition of the Chinese Pharmacopoeia, K. praeruptora is recorded as an effective treatment for congested chest and reducing phlegm, dispelling wind, and clearing heat. It is commonly used to alleviate symptoms such as coughing with phlegm-heat, thick yellow phlegm, and wind-heat cough with excessive phlegm. In a designed study, the honey moxibustion treatment involving K. praeruptora was administered to mice [130]. The results demonstrate the potent expectorant and antitussive effects of this treatment, which are evaluated based on the release of phenol red. The findings indicate that the combination of K. praeruptora with honey moxibustion has the potential to effectively reduce coughing and facilitate expectoration. This is evident through a significant reduction in coughing and improvement in expulsion of respiratory secretions in the treated mice. Moreover, the study reveals that phenol red has a significant impact on coughing in mice. It increased the output of phenol red and inhibited coughing caused by ammonia, while also prolonging the incubation period of coughing. Additionally, phenol red exhibited similar effects on asthma induced by histamine phosphate in guinea pigs. Notably, these effects were found to be stronger than those observed with K. praeruptora alone prior to moxibustion with honey. Furthermore, a specific compound called angle-type pyranocoumarin, found in the raceme of Pra-C, was found to inhibit the contraction of rabbit tracheal smooth muscle induced by acetylcholine and potassium chloride (KCl). It particularly demonstrated efficacy in the contraction and relaxation induced by high potassium [158].

7.2. Anti-Inflammatory and Analgesic Properties

Previous literature reports suggest that Pra-A inhibits the inflammatory response of macrophages induced by lipopolysaccharide (LPS). It is speculated that this compound exerts anti-inflammatory effects in vitro by inhibiting the activation of the NF-kB signaling pathway [138]. On the other hand, Pra-C has been shown to protect neurons from excitotoxicity by down-regulating glutamate N-methyl-D-aspartate (NMDA) [153]. Additionally, it can inhibit the inflammatory response of RAW264.7 cells stimulated by LPS by blocking the NF-kB and signal transducer and activator of transcription 3 (STAT3) signaling pathways [140]. Therefore, recent studies have suggested that Pra-C may possess anti-inflammatory effects in the central nervous system, particularly on microglia. These findings raise the possibility of utilizing Pra-C for the treatment of chronic pain associated with systemic inflammation.

7.3. Cardiovascular Protection

The extract of K. praeruptora has been found to improve left ventricular diastolic function, enhance blood supply to the body, and treat heart failure [142]. This improvement is primarily attributed to the important role of two components, Pra-A and Pra-C. Pra-A acts as a calcium blocker and potassium channel opener, offering myocardial protection. On the other hand, Pra-C exhibits various beneficial effects such as anti-myocardial ischemia, anti-heart failure [159], vasodilation, blood pressure reduction [143], inhibition of calcium influx [144], and reduction in myocardial oxygen consumption [145]. Furthermore, studies have shown that the extract derived from K. praeruptora can modulate gene expression associated with apoptosis in cardiomyocytes induced by abdominal aortic stenosis. This modulation inhibits myocardial remodeling and offers a biological therapeutic impact on heart failure [141]. Notably, the extract also effectively impedes the expression of c-Jun protein, a crucial nuclear transcription factor involved in the c-Jun N-terminal kinase (JNK) pathway of cell signal transduction, thereby protecting the myocardium. Additionally, research has identified coumarin 7, extracted from the rhizome of K. praeruptora, as an inhibitory compound against the proliferation of rat aortic vascular smooth muscle cells. Further investigation and biological testing have revealed that ostrutin7, a coumarin derivative, is the principal anti-proliferation compound that can ameliorate atherosclerosis [144].
Existing studies have demonstrated that Pra-A can effectively decrease mean pulmonary artery pressure, total pulmonary resistance, and pulmonary vascular resistance. Furthermore, it can increase oxygen transport capacity, reduce pulmonary artery vascular resistance, and simultaneously enhance cardiac volume, mixed venous oxygen partial pressure, and oxygen transport capacity [160]. These findings suggest that Pra-A has selectively pulmonary vasodilatory properties, leading to the reduction in pulmonary arterial pressure, pulmonary vascular resistance, and the improvement of cardiac function and tissue oxidation. As a result, it may be utilized as a potential treatment for pulmonary hypertension [141]. In addition, Pra-C has been shown to have blood pressure-lowering effects. Long-term use of Pra-C may reduce blood pressure by increasing the concentration of vascular endothelial nitric oxide (NO). Moreover, Pra-C may also protect against myocardial hypertrophy, ischemia, and an increase in intracellular calcium ions [139]. It can upregulate phospholamban gene expression and Ca2+ concentration, which play a crucial role in ensuring normal cardiac function [161]. By inhibiting Ca2+ influx and increasing Ca2+ uptake in sarcoplasmic reticulum, Pra-C can reduce coronary artery spasm, lower afterload and blood pressure, and protect cardiomyocytes from ischemia-reperfusion injury. Interestingly, the expression of the phospholamban gene in spontaneously hypertensive (SHR) rats was found to be lower than that in normal sprague-dawley (SD) rats, while Pra-C supplementation could up-regulate the expression of the phospholamban gene. This suggest that the low expression of cardiac phospholamban mRNA in SHR may not only be caused by hypertension but also be influenced by mechanisms unrelated to blood pressure [133,162]. Overall, Pra-C has the potential to regulate calcium levels and lower blood pressure and may serve as a promising therapeutic agent for hypertension and related cardiovascular diseases.

7.4. Antineoplastic Activity

Pra-A and Pra-B exhibit anti-proliferation and cytotoxic effects against human gastric cancer cells [147]. Pra-A has demonstrated the ability to enhance the efficacy of doxorubicin (DOX) in treating gastric cancer cells, leading to a more pronounced reduction in cell growth when used in combination with DOX compared to DOX treatment alone. Notably, Pra-B shows promise as an anti-metastasis agent for renal cell carcinoma [149]. It achieves this by inhibiting the epidermal growth factor receptor extracellular-regulated kinase/mitogen-activated protein kinase (EGFR-MEK-ERK) signaling pathway, which consequently down-regulates cathepsin C (CTSC) and cathepsin V (CTSV). As a result, the migration and invasion abilities of human renal cancer cells are significantly decreased.

7.5. Other Pharmacological Effects

According to a previous study, the volatile oil samples of K. praeruptora extracted with 95% ethanol and petroleum ether at 60 °C to 90 °C demonstrated positive effects on Escherichia coli, Staphylococcus aureus, Bacillus typhi, and Shigella flexneri, exhibiting certain antibacterial or bactericidal properties [163]. Additionally, it was reported that Pra-A and Pra-C can significantly up-regulate the expression of multidrug resistance-associated protein 2 through the combined androgen receptor-mediated pathway, providing novel therapeutic approaches for the related cancers [152]. Furthermore, the chemical components of K. praeruptora have been identified using an HEK293 α1A AR cell membrane chromatography (HEK293 α1A/CMC) column and UPLC-ESI-MS/MS coupling system, revealing that Pra-A, Pra-B, and Pra-C exhibit α1 adrenergic receptor activity [155].
Furthermore, to combat the coronavirus disease 2019 (COVID-19) pandemic and showcase the benefits of traditional Chinese medicine in pre-treatment, Jilin Provincial Hospital of Traditional Chinese Medicine has taken proactive measures. The hospital has leveraged traditional Chinese medicine and prepared over 50,000 decoctions, mainly composed of traditional Chinese medicines such as Lonicera japonica Thunb, K. praeruptora, Glycyrrhiza uralensis Fisch, Stemona japonica (BI.) Miq, Perilla frutescens (L.) Britt., Ephedra sinica Stapf, and raw gypsum. It has the effects of treating congested chest and strengthening the exterior, clearing away heat and detoxification, clearing the lung and relieving the throat, and is beneficial to many people in the city [164]. The key groups in each isolation point will be intervened with traditional Chinese medicine to help them improve their immunity and scientifically assist in the prevention and control of the epidemic. Modern pharmacological studies have found that the most important pharmacodynamic components of K. praeruptora are dihydropyranoid coumarin compounds, including Pra-A, Pra-B, and Pra-C [9,91], especially Pra-A and Pra-B, which were included in the quality evaluation index of the 2020 edition of Chinese Pharmacopoeia.

8. Safety and Toxicity

General pharmacological studies revealed that oral administration of Pra-A and Pra-B did not induce behavioral effects or acute toxicity at a dose of 1 g/kg [150]. Furthermore, cytotoxicity tests on RAW264.7 cells and primary mouse peritoneal macrophages showed that Pra-A did not affect cell viability in the dose range of 1 to 100 mg/mL. Similarly, Pra-B did not exhibit cytotoxic effects in primary mouse peritoneal macrophages at doses ranging from 1 to 60 mg/mL, indicating a favorable safety profile in vitro. Nevertheless, further evaluation of the in vivo safety profile is warranted [165]. Acute toxicity tests in laboratory animals demonstrated a wide margin of safety with no observed adverse effects at recommended therapeutic doses. However, research showed that when administered to mice separately via oral gavage and intraperitoneal injection, methanol extract of K. praeruptora exhibited lower toxicity when administered orally, whereas intraperitoneal injection resulted in higher toxicity. This finding should raise caution in clinical drug administration [166]. In the investigation of the extracellular anti-tumor activity of extracts from K. praeruptora, higher concentrations of the extract treatment resulted in a decreased proliferation rate of human hepatocellular carcinomas (HepG2 cells). However, it is important to note that the cytotoxicity of the chemotherapeutic drug doxorubicin against HepG2 cells was significantly greater than that of K. praeruptora extracts [167].

9. Conclusions and Perspectives

K. praeruptora, a renowned traditional Chinese herbal medicine with diverse chemical components and extensive pharmacological effects, exhibits efficacy in treating conditions like wind-heat headache, phlegm-heat cough, and asthma. Its cardiovascular benefits, cancer prevention properties, and antimicrobial activities contribute to its prominence in ethnopharmacology. Despite its growing cultivation in China, addressing knowledge gaps is crucial.
First, cultivation systems need improvement, necessitating a deeper understanding of the bolting gene and mechanism. Current research lacks systematicity, hindering practical advancements. Second, comprehensive research into K. praeruptora’s chemical components, particularly volatile oils, phenanthrenequinone, and sterols, is essential. Utilizing advanced analytical techniques can enhance understanding. Third, physiological and molecular studies should focus on stress responses and the relationship between microorganisms and secondary metabolites. Whole-genome sequencing is imperative to address research gaps.
Regarding cardiovascular effects, research is extensive, but broader physiological impacts remain underexplored. Leveraging biotechnology for active constituent extraction holds promise. Plant-derived microRNAs also play a vital role in human health, influencing growth, development, and gut microbiota [168,169,170,171]. Acknowledging this in the context of K. praeruptora enhances its development of drug mechanisms. Lastly, while K. praeruptora shows potential for disease prevention and treatment, further clinical trials are essential. Overcoming obstacles such as traditional usage, complex composition, and cultural barriers will promote its integration into modern healthcare practices. Future research should focus on bolting characteristics, chemical composition, and bioinformatics to unravel its mechanisms and expand medical applications, solidifying its economic and medical significance.

Author Contributions

Q.W. and R.W. conceptualized the idea, made the figures and tables as well as wrote the manuscript. Q.W. and L.D. were responsible for collecting the available materials. R.W. and Z.L. provided critical feedback of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was financially supported by Natural Science Foundation of Zhejiang Province (LGH22H280005) and Key Research and Development Program of Zhejiang Province (grant no. 2023C02017).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare that they have no competing interests.

References

  1. Liu, R.; Feng, L.; Sun, A.; Kong, L. Preparative isolation, and purification of coumarins from Peucedanum praeruptorum Dunn by high-speed counter-current chromatography. J. Chromatogr. A 2004, 1057, 89–94. [Google Scholar] [CrossRef]
  2. Sheh, M.L. Peucedanum. In Flora of China; Missouri Botanical Garden Press: St. Louis, MO, USA, 1992; Volume 55, pp. 127–158. [Google Scholar]
  3. Sheh, M.L.; Watson, M.F. Peucedanum Linnaeus. In Flora of China; Missouri Botanical Garden Press: St. Louis, MO, USA, 2005; Volume 14, pp. 182–192. [Google Scholar]
  4. Yamazaki, T. Umbelliferae in Japan II. J. Jpn. Bot. 2001, 76, 283–285. [Google Scholar]
  5. Chang, C.S.; Kim, H.; Chang, K.S. Provisional Checklist of Vascular Plants for the Korea Peninsula Flora (KPF); Designpost: Pajo, Philippines, 2014. [Google Scholar]
  6. Pimenov, M.G. Updated Checklist of Chinese Umbelliferae: Nomenclature, Synonymy, Typification, Distribution. Turczaninowia 2017, 20, 106–239. [Google Scholar] [CrossRef]
  7. Plunkett, G.M.; Pimenov, M.G.; Reduron, J.P.; Kljuykov, E.V.; van Wyk, B.E.; Ostroumova, T.A.; Henwood, M.J.; Tilney, P.M.; Spalik, K.; Watson, M.F.; et al. Apiaceae. In Flowering Plants. Eudicots: Apiales, Gentianales (Except Rubiaceae); Kadereit, J.W., Bittrich, V., Eds.; The Families and Genera of Vascular Plants; Springer International Publishing: Cham, Switzerland, 2018; Volume 15, pp. 9–206. [Google Scholar]
  8. Zhou, J.; Wang, W.; Liu, M.; Liu, Z. Molecular authentication of the traditional medicinal plant Peucedanum praeruptorum and its substitutes and adulterants by DNA-barcoding technique. Pharmacogn. Mag. 2014, 10, 385–390. [Google Scholar] [CrossRef]
  9. Song, Y.; Jing, W.; Yan, R.; Wang, Y. Research progress of the studies on the roots of Peucedanum praeruptorum dunn (Peucedani radix). Pak. J. Pharm Sci. 2015, 28, 71–81. [Google Scholar] [PubMed]
  10. Shi, T.T.; Zhang, X.B.; Zhang, K.; Guo, L.P.; Huang, L.Q. Study of extracting Peucedanum praeruptorum planted area in Ningguo of Anhui province based on multi-source and multi-phase image. China J. Chin. Mater. Med. 2017, 42, 4362–4367. [Google Scholar] [CrossRef]
  11. Song, L.Y.; Liu, J.S.; Tan, Y.J.; Zha, G.S. The present situation and prospect of the research on the Chinese Traditional Medicine Peucedanum praeruptorum Dunn. J. Yichun Univ. 2020, 42, 22–25. [Google Scholar] [CrossRef]
  12. Yang, C.Q.; Wu, Z.B.; Yang, T.J.; Deng, C.F. Herbal Textual Research on Growth Habit and distribution of Producing Area about Qianhu. Lishizhen Med. Mater. Medica Res. 2019, 30, 1471–1473. [Google Scholar] [CrossRef]
  13. Du, Y.; Zhou, J.; Guo, R. Imitation wild planting technology under forest of Peucedanum praeruptorum in Fangxian county. Heilongjiang Agric. Sci. 2021, 10, 139–141. [Google Scholar] [CrossRef]
  14. Liu, C.K.; Lei, J.Q.; Jiang, Q.P.; Zhou, D.S.; He, X.J. The complete plastomes of seven Peucedanum plants: Comparative and phylogenetic analyses for the Peucedanum genus. BMC Plant Biol. 2022, 22, 101. [Google Scholar] [CrossRef]
  15. Liu, J.L.; Wang, S.S.; Li, X.F.; Lu, X.Y.; Li, H.; Hong, C.X. Investigation and Analysis on the Present Situation of Peucedanum praeruptorum Resources in Its Producing Area. Mod. Agric. Sci. Technol. 2021, 1, 89–99. [Google Scholar] [CrossRef]
  16. Liu, J.S.; Song, L.Y.; Wang, R.Y.; Xu, F.W.; Zhang, C.Z. Analysis of the influencing factors and reduction measures of early extraction of Peucedanum praeruptorum Dunn. South Chin. Agric. 2022, 16, 84–87. [Google Scholar] [CrossRef]
  17. Qiu, X.X.; Zhang, L.; Yue, J.Y.; Wang, M. Investigation on Factors Affecting Contents of 3 Coumarins in Ningqianhu. J. Chin. Med. Mater. 2016, 39, 713–716. [Google Scholar] [CrossRef]
  18. Chen, X. Research on biological characteristics and artificial cultivation technology of Ningqian Hu. Anhui Agric. Sci. Bull. 2017, 23, 116–118. [Google Scholar] [CrossRef]
  19. Zheng, Y.; Jian, Q.P.; Zhao, R.Q.; Chen, Y.L.; Huang, C.; Liu, Z.P.; Xian, Z.Q.; Wan, M.; Jia, F.M.; Wang, G.Q.; et al. Effects of Different Planting Patterns on Yield and Profit of Peucedanum praeruptorum. Tillage Cultiv. 2019, 39, 31–32. [Google Scholar] [CrossRef]
  20. Liu, S.L.; Wang, X.H.; Gao, Y.G.; Zhao, Y.; Zhang, A.H.; Xu, Y.H.; Zhang, L.X. Transcriptomic analysis identifies differentially expressed genes (DEGs) associated with bolting and flowering in Saposhnikovia divaricata. Chin. J. Nat. Med. 2018, 16, 446–455. [Google Scholar] [CrossRef] [PubMed]
  21. Li, M.; Cui, X.; Jin, L.; Li, M.; Wei, J. Bolting reduces ferulic acid and flavonoid biosynthesis and induces root lignification in Angelica sinensis. Plant Physiol. Biochem. 2022, 170, 171–179. [Google Scholar] [CrossRef]
  22. Yang, L.; Li, J.; Xiao, Y.; Zhou, G. Early Bolting, Yield, and Quality of Angelica sinensis (Oliv.) Diels Responses to Intercropping Patterns. Plants 2022, 11, 2950. [Google Scholar] [CrossRef]
  23. Xie, J.; Tang, X.; Xie, C.; Wang, Y.; Huang, J.; Jin, J.; Liu, H.; Zhong, C.; Zhou, R.; Ren, G.; et al. Comparative analysis of root anatomical structure, chemical components and differentially expressed genes between early bolting and unbolting in Peucedanum praeruptorum Dunn. Genomics 2023, 115, 110557. [Google Scholar] [CrossRef]
  24. Zhao, D.Y.; Hao, Q.X.; Kang, L.P.; Zhang, Y.; Chen, M.L.; Wang, T.L.; Guo, L.P. Advance in studying early bolting of Umbelliferae medicinal plant. China J. Chin. Mater. Med. 2016, 41, 20–23. [Google Scholar] [CrossRef]
  25. Xie, J.; Zhen, L.P.; Ren, G.-X.; Tang, X.-Y.; Liu, H.; Zhong, C.; Jin, J.; Huang, J.-H.; Zhang, S.-H. Influence of early bolting on anatomical structure and coumarins of Peucedanum praeruptorum. China J. Chin. Mater. Med. 2020, 45, 4861–4866. [Google Scholar] [CrossRef]
  26. Luo, L.; Liu, X.; Jin, X.; Liu, Y.; Ma, J.; Zhang, S.; Zhang, D.; Chen, X.; Sheng, L.; Li, Y. Simultaneous determination of skimmin, apiosylskimmin, 7-hydroxycoumarin and 7-hydroxycoumarin glucuronide in rat plasma by liquid chromatography-Orbitrap mass spectrometry and its application to pharmacokinetics. Biomed. Chromatogr. 2022, 36, e5223. [Google Scholar] [CrossRef] [PubMed]
  27. Lu, L.; Monakhos, S.G.; Lim, Y.P.; Yi, S.Y. Early Defense Mechanisms of Brassica oleracea in Response to Attack by Xanthomonas campestris pv. campestris. Plants 2021, 10, 2705. [Google Scholar] [CrossRef] [PubMed]
  28. Baldwin, S.; Revanna, R.; Pither-Joyce, M.; Shaw, M.; Wright, K.; Thomson, S.; Moya, L.; Lee, R.; Macknight, R.; McCallum, J. Genetic analyses of bolting in bulb onion (Allium cepa L.). Theor. Appl. Genet. 2014, 127, 535–547. [Google Scholar] [CrossRef] [PubMed]
  29. Wei, X.; Rahim, M.A.; Zhao, Y.; Yang, S.; Wang, Z.; Su, H.; Li, L.; Niu, L.; Harun-Ur-Rashid, M.; Yuan, Y.; et al. Comparative Transcriptome Analysis of Early- and Late-Bolting Traits in Chinese Cabbage (Brassica rapa). Front. Genet. 2021, 12, 590830. [Google Scholar] [CrossRef]
  30. Wang, J.L.; Li, X.X.; Qiu, Y.; Zhang, X.H.; Shen, D.; Wang, H.P.; Sun, Y.Y.; Duan, M.M. Research Advances on Molecular Markers and Regulation Mechanism for the Bolting and Blooming Characteristics of Brassicaceae Vegetables. J. Plant Genet. Res. 2015, 16, 1283–1289. [Google Scholar] [CrossRef]
  31. Xu, L.; Liu, H.; Kilian, A.; Bhoite, R.; Liu, G.; Si, P.; Wang, J.; Zhou, W.; Yan, G. QTL Mapping Using a High-Density Genetic Map to Identify Candidate Genes Associated with Metribuzin Tolerance in Hexaploid Wheat (Triticum aestivum L.). Front. Plant Sci. 2020, 11, 573439. [Google Scholar] [CrossRef]
  32. Lou, P.; Zhao, J.; Kim, J.S.; Shen, S.; Del Carpio, D.P.; Song, X.; Jin, M.; Vreugdenhil, D.; Wang, X.; Koornneef, M.; et al. Quantitative trait loci for flowering time and morphological traits in multiple populations of Brassica rapa. J. Exp. Bot. 2007, 58, 4005–4016. [Google Scholar] [CrossRef]
  33. Zhao, J.; Kulkarni, V.; Liu, N.; Del Carpio, D.P.; Bucher, J.; Bonnema, G. BrFLC2 (FLOWERING LOCUS C) as a candidate gene for a vernalization response QTL in Brassica rapa. J. Exp. Bot. 2010, 61, 1817–1825. [Google Scholar] [CrossRef]
  34. Kawanabe, T.; Fujimoto, R. Inflorescence abnormalities occur with overexpression of Arabidopsis lyrata FT in the fwa mutant of Arabidopsis thaliana. Plant Sci. 2011, 181, 496–503. [Google Scholar] [CrossRef]
  35. Miao, X.Y.; Qu, H.P.; Han, Y.L.; He, C.F.; Qiu, D.W.; Cheng, Z.W. The protein elicitor Hrip1 enhances resistance to insects and early bolting and flowering in Arabidopsis thaliana. PLoS ONE 2019, 14, e0216082. [Google Scholar] [CrossRef] [PubMed]
  36. Hui, N.N.; Wang, L.; Li, J.P.; Qian, T.Y. Effects of 8 exogenous hormones on bolting and yield of Angelica sinensis. Gansu Agric. Sci. Tech. 2015, 12, 27–30. [Google Scholar] [CrossRef]
  37. Hou, K.; Chen, J.W.; Zhai, J.Y.; Shen, H.; Chen, L.; Wu, W. Effects of Plant Growth Regulators on the Growth and Development, Yield and Quality of Angelica dahurica. China J. Chin. Mater. Med. 2013, 38, 2082–2085. [Google Scholar]
  38. Sun, P.X.; Zhang, L.P.; Wu, D.; Zhang, W.; Huang, C.; Cheng, S.H.; Zhu, G.P.; Liu, P.W. Effects of GA3 and MeJA treatment on growth and physiological indexes of Chinese cabbage seedlings. Chin. Cucur. Veg. 2023, 36, 56–63. [Google Scholar] [CrossRef]
  39. Song, C.; Li, X.; Jia, B.; Liu, L.; Ou, J.; Han, B. De novo Transcriptome Sequencing Coupled with Co-expression Analysis Reveal the Transcriptional Regulation of Key Genes Involved in the Formation of Active Ingredients in Peucedanum praeruptorum Dunn Under Bolting Period. Front. Genet. 2021, 12, 683037. [Google Scholar] [CrossRef] [PubMed]
  40. Song, C.; Li, X.; Jia, B.; Liu, L.; Wei, P.; Manzoor, M.A.; Wang, F.; Li, B.Y.; Wang, G.; Chen, C.; et al. Comparative Transcriptomics Unveil the Crucial Genes Involved in Coumarin Biosynthesis in Peucedanum praeruptorum Dunn. Front. Plant Sci. 2022, 13, 899819. [Google Scholar] [CrossRef]
  41. Miao, X.S.; Cui, Q.Y.; Wang, Z.Y.; Liu, X.N.; Zhao, A.B.; Qiao, Y.J.; Wu, Z.S. Advance in quality assessment of Chinese materia medica using microscopic and morphological methods. Chin. J. Nat Med. 2017, 15, 653–663. [Google Scholar] [CrossRef]
  42. Shi, S.M.; Pan, M.J.; Wang, J.; Chen, C.Q. Application of molecular identification techniques in Chinese materia medica. J. Chin. Med. Mater. 2016, 47, 3121–3126. [Google Scholar] [CrossRef]
  43. Zhou, Y. Identification of Radix Peucedani and Its Adulterant Radix Peucedani Terebinthaceum. Chin. Pharm. 2008, 17, 62–63. [Google Scholar]
  44. Qi, Z.C.; Pi, E.; Zhang, X.D.; Möller, M.; Jiang, B.; Lu, H.F. Ontogenesis of D-type stomata, and cork-warts on the leaf epidermis of Camellia japonica (Theaceae) and functional assessment. Flora 2017, 228, 24–30. [Google Scholar] [CrossRef]
  45. Huang, D.L.; Xu, Y.Q.; Chen, X.K. Rapid identification of Pencedani praeruptorum and its counterfeit Saposhmikoviae divaricata by second derivative spectroscopy. J. ShaoGuan Univ. 2009, 30, 50–53. [Google Scholar] [CrossRef]
  46. Wang, F.; Jia, B.; Song, X.; Dai, J.; Li, X.; Gao, H.; Pan, H.; Yan, H.; Han, B. Rapid Identification of Peucedanum praeruptorum Dunn and Its Adulterants by Hand-Held Near-Infrared Spectroscopy. J. AOAC Int. 2022, 105, 928–933. [Google Scholar] [CrossRef] [PubMed]
  47. Zhang, X.; Yu, Y.; Cen, Y.; Yang, D.; Qi, Z.; Hou, Z.; Han, S.; Cai, Z.; Liu, K. Bivariate Correlation Analysis of the Chemometric Profiles of Chinese Wild Salvia miltiorrhiza Based on UPLC-Qqq-MS and Antioxidant Activities. Molecules 2018, 23, 538. [Google Scholar] [CrossRef] [PubMed]
  48. Ma, L.; Luo, Y.; Chen, B.; Zheng, C.; Du, W.; Shi, X.; Guo, Z. Determination of Characteristic Volatile Component Fingerprint of Peucedanum praeruptorum Dunn at Different Harvest Periods Based on HS-GC-IMS. J. AOAC Int. 2023, 106, 445–456. [Google Scholar] [CrossRef] [PubMed]
  49. Hebert, P.D.; Cywinska, A.; Ball, S.L.; deWaard, J.R. Biological identifications through DNA barcodes. Proc. Biol. Sci. 2003, 270, 313–321. [Google Scholar] [CrossRef]
  50. Pan, J.; Li, X.; Li, J.; Liu, H.; Hou, Z.; Yang, Z.; Xu, L. Genome-wide identification of U-box gene family and expression analysis under abiotic stresses in Salvia miltiorrhiza. Biocell 2022, 46, 1751. [Google Scholar] [CrossRef]
  51. Choo, B.K.; Moon, B.C.; Ji, Y.; Kim, B.B.; Choi, G.; Yoon, T.; Kim, H.K. Development of SCAR markers for the discrimination of three species of medicinal plants, Angelica decursiva (Peucedanum decursivum), Peucedanum praeruptorum and Anthricus sylvestris, based on the internal transcribed spacer (ITS) sequence and random amplified polymorphic DNA (RAPD). Biol. Pharm. Bull. 2009, 32, 24–30. [Google Scholar] [CrossRef]
  52. Yang, Q.; Jiang, Y.; Wang, Y.; Han, R.; Liang, Z.; He, Q.; Jia, Q. SSR Loci Analysis in Transcriptome and Molecular Marker Development in Polygonatum sibiricum. Biomed. Res. 2022, 2022, 4237913. [Google Scholar] [CrossRef]
  53. Liu, T.S.; Lin, L.M.; Xiao, B.M.; Pan, Q.P.; Zhou, X.J. Identification of the Commercial Medicinal Materials “Qian-Hu” (Radix Peucedani) and Probing their Genetic Relations by RAPD Technique. China Assoc. Chin. Med. 2006, 24, 1838–1840. [Google Scholar] [CrossRef]
  54. Liu, Y.M.; Yi, Z.; Xiong, Y.X.; Xiu, Y.; Chen, K.L. Research on Genetic diversity of Peucedanum praeruptorum with ISSR markers method. Lishizhen Med. Mater. Medica Res. 2014, 25, 1982–1984. [Google Scholar] [CrossRef]
  55. Zhou, X.; Zhang, Z.C.; Huang, Y.B.; Xiao, H.W.; Wu, J.J.; Qi, Z.C.; Wei, Y.K. Conservation Genomics of Wild Red Sage (Salvia miltiorrhiza) and Its Endangered Relatives in China: Population Structure and Interspecific Relationships Revealed From 2b-RAD Data. Front. Genet. 2021, 12, 688323. [Google Scholar] [CrossRef] [PubMed]
  56. Feng, J.Y.; Jin, X.J.; Zhang, S.L.; Yang, J.W.; Fei, S.P.; Huang, Y.S.; Liu, Y.; Qi, Z.C.; Li, P. Smilax weniae, a New Species of Smilacaceae from Limestone Areas Bordering Guizhou and Guangxi, China. Plants 2022, 11, 1032. [Google Scholar] [CrossRef] [PubMed]
  57. Qi, Z.C.; Yu, Y.; Liu, X.; Pais, A.; Ranney, T.; Whetten, R.; Xiang, Q.Y. Phylogenomics of polyploid Fothergilla (Hamamelidaceae) by RAD-tag based GBS-Insights into species origin and effects of software pipelines. J. Syst. Evol. 2015, 53, 432–447. [Google Scholar] [CrossRef]
  58. Hou, D.Y.; Song, J.Y.; Yang, P.; Zhou, H.; Xin, T.Y.; Yao, H. Identification of Peucedanum praeruptorum and Peucedanum decursivum and Their Counterfeit Products Based on ITS2 Sequence. China J. Chin. Mater. Med. 2014, 39, 4186–4190. [Google Scholar] [CrossRef]
  59. Han, B.X.; Yuan, Y.; Huang, L.Q.; Zhao, Q.; Tan, L.L.; Song, X.W.; He, X.M.; Xu, T.; Liu, F.; Wang, J. Specific PCR Identification between Peucedanum praeruptorum and Angelica decursiva and Identification between Them and Adulterant Using DNA Barcode. Pharmacogn. Mag. 2017, 13, 38–45. [Google Scholar] [CrossRef]
  60. Coissac, E.; Hollingsworth, P.M.; Lavergne, S.; Taberlet, P. From barcodes to genomes: Extending the concept of DNA barcoding. Mol. Ecol. 2016, 25, 1423–1428. [Google Scholar] [CrossRef]
  61. Li, J.; Li, X.; Han, P.; Liu, H.; Gong, J.; Zhou, W.; Shi, B.; Liu, A.; Xu, L. Genome-wide investigation of bHLH genes and expression analysis under different biotic and abiotic stresses in Helianthus annuus L. Int. J. Biol. Macromol. 2021, 189, 72–83. [Google Scholar] [CrossRef]
  62. Li, Y.; Geng, M.; Xu, Z.; Wang, Q.; Li, L.; Xu, M.; Li, M. The complete plastome of Peucedanum praeruptorum (Apiaceae). Mitochondrial DNA B Resour. 2019, 4, 3612–3613. [Google Scholar] [CrossRef]
  63. Chu, S.; Chen, L.; Xie, H.; Xie, J.; Zhao, Y.; Tong, Z.; Xu, R.; Peng, H. Comparative analysis and chemical profiling of different forms of Peucedani radix. J. Pharmaceut. Biomed. 2020, 189, 113410. [Google Scholar] [CrossRef]
  64. Wang, F.Y.; You, H.Q.; Guo, Y.H.; Wei, Y.K.; Xia, P.G.; Yang, Z.Q.; Ren, M.; Guo, H.; Han, R.L.; Yang, D.F. Essential oils from three kinds of fingered citrons and their antibacterial activities. Ind. Crop. Prod. 2020, 147, 112172. [Google Scholar] [CrossRef]
  65. Chen, F.Y.; Tu, L.F.; Liu, D.P.; Ma, J.; Luo, Y.M. Chemical constituents from the roots of Peucedanum praeruptorum Dunn and their chemotaxonomic significance. Biochem. Syst. Ecol. 2021, 99, 104355. [Google Scholar] [CrossRef]
  66. Li, X.Y.; Zu, Y.Y.; Ning, W.; Tang, M.X.; Gong, C.; Niu, S.L.; Hua, H.M. A new xanthyletin-type coumarin from the roots of Peucedanum praeruptorum. J. Asian Nat. Prod. Res. 2020, 22, 287–294. [Google Scholar] [CrossRef] [PubMed]
  67. Seo, W.D.; Kim, J.Y.; Ryu, H.W.; Kim, J.H.; Han, S.I.; Ra, J.E.; Seo, K.H.; Jang, K.C.; Lee, J.H. Identification and characterisation of coumarins from the roots of Angelica dahurica and their inhibitory effects against cholinesterase. J. Funct. Foods 2013, 5, 1421–1431. [Google Scholar] [CrossRef]
  68. Brown, S.A. Biosynthesis of Coumarin and Herniarin in Lavender. Science 1962, 137, 977–978. [Google Scholar] [CrossRef] [PubMed]
  69. Sun, J.; Yue, Y.D.; Tang, F.; Guo, X.F. Coumarins from the leaves of Bambusa pervariabilis McClure. J. Asian Nat. Prod. Res. 2010, 12, 248–251. [Google Scholar] [CrossRef] [PubMed]
  70. Chen, L.L.; Chu, S.S.; Zhang, L.; Xie, J.; Dai, M.; Wu, X.; Peng, H.S. Tissue-Specific Metabolite Profiling on the Different Parts of Bolting and Unbolting Peucedanum praeruptorum Dunn (Qianhu) by Laser Microdissection Combined with UPLC-Q/TOF⁻MS and HPLC⁻DAD. Molecules 2019, 24, 1439. [Google Scholar] [CrossRef]
  71. Lee, S.O.; Choi, S.Z.; Lee, J.H.; Chung, S.H.; Park, S.H.; Kang, H.C.; Yang, E.Y.; Cho, H.J.; Lee, K.R. Antidiabetic coumarin and cyclitol compounds from Peucedanum japonicum. Arch. Pharm. Res. 2004, 27, 1207–1210. [Google Scholar] [CrossRef]
  72. Song, Y.L.; Jing, W.H.; Du, G.; Yang, F.Q.; Yan, R.; Wang, Y.T. Qualitative analysis and enantiospecific determination of angular type pyranocoumarins in Peucedani Radix using achiral and chiral liquid chromatography coupled with tandem mass spectrometry. J. Chromatogr. A 2014, 1338, 24–37. [Google Scholar] [CrossRef]
  73. Song, Y.L.; Jing, W.H.; Yang, F.Q.; Shi, Z.; Yao, M.C.; Yan, R.; Wang, Y.T. Simultaneously enantiospecific determination of (+)-trans-khellactone, (+/−)-praeruptorin A, (+/−)-praeruptorin B, (+)-praeruptorin E, and their metabolites, (+/−)-cis-khellactone, in rat plasma using online solid phase extraction-chiral LC-MS/MS. J. Pharmaceut. Biomed. 2014, 88, 269–277. [Google Scholar] [CrossRef]
  74. Shehzad, O.; Khan, S.; Ha, I.J.; Park, Y.; Tosun, A.; Kim, Y.S. Application of stepwise gradients in counter-current chromatography: A rapid and economical strategy for the one-step separation of eight coumarins from Seseli resinosum. J. Chromatogr. A 2013, 1310, 66–73. [Google Scholar] [CrossRef]
  75. Shimizu, B. 2-Oxoglutarate-dependent dioxygenases in the biosynthesis of simple coumarins. Front. Plant Sci. 2014, 5, 549. [Google Scholar] [CrossRef] [PubMed]
  76. Duan, Z.; Wu, F.; Yan, Q.; Zhang, J.Y. Research progress on plant coumarin biosynthesis pathway and genes encoding the key enzymes. Acta Prataculturae Sin. 2022, 31, 217–228. [Google Scholar] [CrossRef]
  77. Wang, R.X.; Song, J.; Sun, B.; Yan, X.; Zhang, W.Z.; Zhao, C. Research Progress of Function and Biosynthesis of Coumarins. China Biotechnol. 2022, 42, 79–90. [Google Scholar]
  78. Fernández-Peña, L.; Matos, M.J.; López, E. Recent Advances in Biologically Active Coumarins from Marine Sources: Synthesis and Evaluation. Mar. Drugs 2022, 21, 37. [Google Scholar] [CrossRef]
  79. Okuyama, T.; Takata, M.; Shibata, S. Structures of Linear Furano- and Simple-Coumarin Glycosides of Bai-Hua Qian-Hu. Planta Med. 1989, 55, 64–67. [Google Scholar] [CrossRef]
  80. Zhang, Y.; Gao, Z.; Cai, Y.; Dou, X.; Liang, Y.; Zhang, W.; Wu, G.; Ye, J. A novel strategy integrating gas phase fractionation with staggered mass range and LC-MS/MS molecular network for comprehensive metabolites profiling of Gui Ling Ji in rats. J. Pharm. Biomed. 2023, 222, 115092. [Google Scholar] [CrossRef]
  81. Kwon, Y.S.; Kobayashi, A.; Kajiyama, S.; Kawazu, K.; Kanzaki, H.; Kim, C.M. Antimicrobial constituents of Angelica dahurica roots. Phytochemistry 1997, 44, 887–889. [Google Scholar] [CrossRef]
  82. Kim, S.J.; Ko, S.M.; Choi, E.J.; Ham, S.H.; Kwon, Y.D.; Lee, Y.B.; Cho, H.Y. Simultaneous Determination of Decursin, Decursinol Angelate, Nodakenin, and Decursinol of Angelica gigas Nakai in Human Plasma by UHPLC-MS/MS: Application to Pharmacokinetic Study. Molecules 2018, 23, 1019. [Google Scholar] [CrossRef]
  83. Kimura, Y.; Okuda, H. Histamine-Release Effectors from Angelica dahurica var. dahurica Root. J. Nat. Prod. 1997, 60, 249–251. [Google Scholar] [CrossRef]
  84. Song, Y.L.; Jing, W.H.; Chen, Y.G.; Yuan, Y.F.; Yan, R.; Wang, Y.T. 1H nuclear magnetic resonance based-metabolomic characterization of Peucedani Radix and simultaneous determination of praeruptorin A and praeruptorin B. J. Pharmaceut. Biomed. 2014, 93, 86–94. [Google Scholar] [CrossRef]
  85. Piplani, P.; Jain, A.; Devi, D.; Anjali; Sharma, A.; Silakari, P. Design, synthesis and pharmacological evaluation of some novel indanone derivatives as acetylcholinesterase inhibitors for the management of cognitive dysfunction. Bioorg. Med. Chem. 2018, 26, 215–224. [Google Scholar] [CrossRef] [PubMed]
  86. Kong, L.Y.; Li, Y.; Pei, Y.H.; Yu, R.M.; Min, Z.D.; Zhu, T.R. Isolation and structure elucidation of BaiHuaQianHuoside and PD-C-I from Peucedanum praeruptorum. Acta Pharm. Sin. 1994, 29, 276–280. [Google Scholar] [CrossRef]
  87. Kong, L.Y.; Pei, Y.H.; Li, X.; Zhu, T.R.; Ao, S.C. Isolation, and structure elucidation of QianHuCoumarin A. Acta Pharm. Sin. 1993, 28, 432–436. [Google Scholar] [CrossRef]
  88. Chen, Z.X.; Huang, B.S.; She, Q.L.; Zeng, G.F. The chemical constituents of Bai-Hua-Qian-Hu, the root of Peucedanum Praeruptorum Dunn. (Umbelliferae)—Four new coumarins. Acta. Pharmacol. Sin. 1979, 14, 486–496. [Google Scholar] [CrossRef]
  89. Asahara, T.; Sakakibara, I.; Okuyama, T.; Shibata, S. Studies on Coumarins of a Chinese Drug “Qian-Hu” V. Coumarin-glycosides from “Zi-Hua Qian-Hu”1. Planta Med. 1984, 50, 488–492. [Google Scholar] [CrossRef] [PubMed]
  90. Kawasaki, C.; Okuyama, T.; Shibata, S.; Iitaka, Y. Studies on Coumarins of a Chinese Drug “Qian-Hu”; VI1, Coumarins of Angelica edulis. Planta Med. 1984, 50, 492–496. [Google Scholar] [CrossRef] [PubMed]
  91. Zhang, C.; Xiao, Y.Q.; Taniguchi, M.; Baba, K. Studies on chemical constituents in roots of Peucedanum praeruptorum V. China J. Chin. Mater. Med. 2012, 37, 3573–3576. [Google Scholar] [CrossRef]
  92. Yu, N.J.; Liu, S.J.; Liang, Y.M.; Fang, C.W.; Xie, L.L.; Ma, K.; Li, W.J. A Comparison of Chemical Components of Volatile Oils from the Decocting Pieces of Peucedanum praeruptorum in Different Regions. J. Anhui Colg. 2007, 26, 44–45. [Google Scholar] [CrossRef]
  93. Zhang, C.; Xiao, Y.Q.; Taniguchi, M.; Baba, K. Studies on chemical constituents in roots of Peucedanum praeruptorum III. China J. Chin. Mater. Med. 2009, 34, 1005–1006. [Google Scholar] [CrossRef]
  94. Ishii, H.; Okada, Y.; Baba, M.; Okuyama, T. Studies of coumarins from the Chinese drug Qianhu, XXVII: Structure of a new simple coumarin glycoside from Bai-Hua Qianhu, Peucedanum praeruptorum. Chem. Pharm. Bull. 2008, 56, 1349–1351. [Google Scholar] [CrossRef]
  95. Yamazaki, T.; Tokiwa, T. Isofraxidin, a coumarin component from Acanthopanax senticosus, inhibits matrix metalloproteinase-7 expression and cell invasion of human hepatoma cells. Biol. Pharm. Bull. 2010, 33, 1716–1722. [Google Scholar] [CrossRef] [PubMed]
  96. Meng, J.; Zhang, Y.; Wang, G.; Ji, M.; Wang, B.; He, G.; Wang, Q.; Bai, F.; Xu, K.; Yuan, D.; et al. Conduction of a chemical structure-guided metabolic phenotype analysis method targeting phenylpropane pathway via LC-MS: Ginkgo biloba and soybean as examples. Food Chem. 2022, 390, 133155. [Google Scholar] [CrossRef] [PubMed]
  97. Pang, W.H.; Zhao, X.J.; Chen, X.; Zhang, Y.H.; Wang, C.Q.; Zhao, Q.Y.; Jiao, B.N. Ultra-performance liquid chromatography-quadrupole-time-of-flight high-resolution mass spectrometry method for rapid screening of bioactive compounds in lemon fruits. Food Ferment. Ind. 2021, 47, 222–230. [Google Scholar] [CrossRef]
  98. Rodríguez-Domínguez, J.; Kirsch, G. Zirconyl chloride: A useful catalyst in the pechmann coumarin synthesis. Synth. Stuttg. 2006, 2006, 1895–1897. [Google Scholar] [CrossRef]
  99. Seo, U.M.; Zhao, B.T.; Kim, Y.H.; Kang, J.S.; Son, J.K.; Woo, M.H.; Woo, M. Simultaneous analysis of seven marker compounds from Saposhnikoviae Radix, Glehniae Radix and Peucedani Japonici Radix by HPLC/PDA. Arch. Pharm. Res. 2016, 39, 695–704. [Google Scholar] [CrossRef] [PubMed]
  100. Olennikov, D.N. Coumarins of Lovage Roots (Levisticum officinale W.D.J.Koch): LC-MS Profile, Quantification, and Stability during Postharvest Storage. Metabolites 2023, 13, 3. [Google Scholar] [CrossRef] [PubMed]
  101. Zheng, Y.; Yang, S.; Si, J.; Zhao, Y.; Zhao, M.; Ji, E. Shashen-Maidong Decoction inhibited cancer growth under intermittent hypoxia conditions by suppressing oxidative stress and inflammation. J. Ethnopharmacol. 2022, 299, 115654. [Google Scholar] [CrossRef]
  102. Zhang, H.; Ding, L.; Shi, X.; Mei, W.; Huang, Z.; Zhang, L.; Li, X.; Xu, B.; Zhang, L.; Wang, P. Imperatorin alleviated NLR family pyrin domain-containing 3 inflammasome cascade-induced synovial fibrosis and synovitis in rats with knee osteoarthritis. Bioengineered 2021, 12, 12954–12964. [Google Scholar] [CrossRef]
  103. Li, G.; Xiang, S.; Pan, Y.; Long, X.; Cheng, Y.; Han, L.; Zhao, X. Effects of Cold-Pressing and Hydrodistillation on the Active Non-volatile Components in Lemon Essential Oil and the Effects of the Resulting Oils on Aging-Related Oxidative Stress in Mice. Front. Nutr. 2021, 8, 689094. [Google Scholar] [CrossRef]
  104. Wang, K.; Nie, Z.X.; Sun, Y.P.; Liu, S.Z.; Wang, G.K. A Study on Chemical Components of Peucedanum praeruptorum Dunn. Anhui Med. J. 2018, 37, 83–85. [Google Scholar] [CrossRef]
  105. Liu, R.; Sun, Q.; Shi, Y.; Kong, L. Isolation and purification of coumarin compounds from the root of Peucedanum decursivum (Miq.) Maxim by high-speed counter-current chromatography. J. Chromatogr. A 2005, 1076, 127–132. [Google Scholar] [CrossRef] [PubMed]
  106. Zhao, Y.; Wang, M.; Sun, L.; Jiang, X.; Zhao, M.; Zhao, C. Rapid characterization of the chemical constituents of Sanhua decoction by UHPLC coupled with Fourier transform ion cyclotron resonance mass spectrometry. Rsc. Adv. 2020, 10, 26109–26119. [Google Scholar] [CrossRef] [PubMed]
  107. Chang, H.; Okada, Y.; Okuyama, T.; Tu, P. 1H and 13C NMR assignments for two new angular furanocoumarin glycosides from Peucedanum praeruptorum. Magn. Reson. Chem. 2007, 45, 611–614. [Google Scholar] [CrossRef] [PubMed]
  108. Chang, H.T.; Okada, Y.; Ma, T.J.; Okuyama, T.; Tu, P.F. Two new coumarin glycosides from Peucedanum praeruptorum. J. Asian Nat. Prod. Res. 2008, 10, 577–581. [Google Scholar] [CrossRef] [PubMed]
  109. Kong, L.Y.; Li, Y.; Min, Z.D.; Li, X.; Zhu, T.R. Coumarins from Peucedanum praeruptorum. Phytochemistry 1996, 41, 1423–1426. [Google Scholar] [CrossRef]
  110. Nitao, J.K.; Berhow, M.; Duval, S.M.; Weisleder, D.; Vaughn, S.F.; Zangerl, A.; Berenbaum, M.R. Characterization of furanocoumarin metabolites in parsnip webworm, Depressaria pastinacella. J. Chem. Ecol. 2003, 29, 671–682. [Google Scholar] [CrossRef] [PubMed]
  111. Zhang, C.; Xiao, Y.Q.; Taniguchi, M.; Baba, K. Studies on chemical constituents in roots of Peucedanum praeruptorum. China J. Chin. Mater. Med. 2005, 30, 675–676. [Google Scholar] [CrossRef]
  112. Lee, J.H.; Mei, H.C.; Kuo, I.C.; Lee, T.H.; Chen, Y.H.; Lee, C.K. Characterizing Tyrosinase Modulators from the Roots of Angelica keiskei Using Tyrosinase Inhibition Assay and UPLC-MS/MS as the Combinatorial Novel Approach. Molecules 2019, 24, 3297. [Google Scholar] [CrossRef]
  113. Huang, P.; Zheng, X.Z.; Xi, Z.M.; Zhong, X.Q. Isolation and structure elucidation of longshengensin A from Peucedanum longshengense. Acta Pharmacol. Sin. 1997, 32, 62–64. [Google Scholar] [CrossRef]
  114. Takata, M.; Shibata, S.; Okuyama, T. Structures of Angular Pyranocoumarins of Bai-Hua Qian-Hu, the Root of Peucedanum praeruptorum1. Planta Med. 1990, 56, 307–311. [Google Scholar] [CrossRef]
  115. Ding, X.; Li, L.; Wang, Y.; Chen, J.; Huang, Y.; Xu, K. Design of guanidinium ionic liquid-based microwave-assisted extraction for the efficient extraction of Praeruptorin A from Radix peucedani. J. Sep. Sci. 2014, 37, 3539–3547. [Google Scholar] [CrossRef] [PubMed]
  116. Zhou, G.; Chen, G.; Liu, H. Simultaneous Quantification of Three Pyranocoumarins of Peucedanum praeruptorum in Rat Plasma by Liquid Chromatography-Tandem Mass Spectrometry: Application to Pharmacokinetic Study. J. Chromatogr. Sci. 2015, 53, 511–518. [Google Scholar] [CrossRef] [PubMed]
  117. Liang, T.; Yue, W.; Du, X.; Ren, L.; Li, Q. Pharmacokinetics and Tissue Distribution Study of Praeruptorin D from Radix Peucedani in Rats by High-Performance Liquid Chromatography (HPLC). IJMS 2012, 13, 9129–9141. [Google Scholar] [CrossRef] [PubMed]
  118. Hisamoto, M.; Kikuzaki, H.; Ohigashi, H.; Nakatani, N. Antioxidant compounds from the leaves of Peucedanum japonicum thunb. J. Agric. Food Chem. 2003, 51, 5255–5261. [Google Scholar] [CrossRef] [PubMed]
  119. Nugara, R.N.; Inafuku, M.; Takara, K.; Iwasaki, H.; Oku, H. Pteryxin: A coumarin in Peucedanum japonicum Thunb leaves exerts antiobesity activity through modulation of adipogenic gene network. Nutrition 2014, 30, 1177–1184. [Google Scholar] [CrossRef] [PubMed]
  120. Willette, R.E.; Soine, T.O. Coumarins i. isolation, purification, and structure determination of pteryxin and suksdorfin. J. Pharm. 2010, 51, 149–156. [Google Scholar] [CrossRef]
  121. Jacobson, K.A.; van Galen, P.J.; Williams, M. Adenosine receptors: Pharmacology, structure-activity relationships, and therapeutic potential. J. Med. Chem. 1992, 35, 407–422. [Google Scholar] [CrossRef]
  122. Giuliani, C.; Pieraccini, G.; Santilli, C.; Tani, C.; Bottoni, M.; Schiff, S.; Fico, G.; Papini, A.; Falsini, S. Anatomical Investigation and GC/MS Analysis of ‘Coco de Mer’, Lodoicea maldivica (Arecaceae). Chem. Biodivers. 2020, 17, e2000707. [Google Scholar] [CrossRef]
  123. Nakanishi, K. Tetsuo Nozoe’s “Autograph Books by Chemists 1953–1994”: An essay. Chem. Rec. 2013, 13, 343–352. [Google Scholar] [CrossRef]
  124. Szmigielski, R.; Cieslak, M.; Rudziński, K.J.; Maciejewska, B. Identification of volatiles from Pinus silvestris attractive for Monochamus galloprovincialis using a SPME-GC/MS platform. Environ. Sci. Pollut. Res. Int. 2011, 19, 2860–2869. [Google Scholar] [CrossRef]
  125. Nolan, S.J.; Thornton, J.; Murray, C.S.; Dwyer, T. Inhaled mannitol for cystic fibrosis. Cochrane Database Syst. Rev. 2015, CD008649. [Google Scholar] [CrossRef] [PubMed]
  126. Zhang, C.; Xiao, Y.Q.; Taniguchi, M.; Baba, K. Studies on chemical constituents in roots of Peucedanum praeruptorum II. China J. Chin. Mater. Med. 2006, 31, 1333–1334. [Google Scholar] [CrossRef]
  127. de Sousa, D.B.; da Silva, G.S.; Serrano, L.A.L.; Martins, M.V.V.; Rodrigues, T.H.S.; Lima, M.A.S.; Zocolo, G.J. Metabolomic Profile of Volatile Organic Compounds from Leaves of Cashew Clones by HS-SPME/GC-MS for the Identification of Candidates for Anthracnose Resistance Markers. J. Chem. Ecol. 2023, 49, 87–102. [Google Scholar] [CrossRef] [PubMed]
  128. Wu, Y.Z.; Yu, H.T.; Yu, X.Y.; Zhu, L.J.; Yu, Z.F. Comparison of volatile compounds in Chrysanthemum nankingense during storage based on HS-SPME-GC-MS and E-nose. J. Food Meas. Charact. 2023, 17, 3134–3148. [Google Scholar] [CrossRef]
  129. Nerio, L.S.; Olivero-Verbel, J.; Stashenko, E. Repellent activity of essential oils: A review. Bioresour. Technol. 2010, 101, 372–378. [Google Scholar] [CrossRef]
  130. Zhang, C.; Li, L.; Xiao, Y.Q.; Li, W.; Yin, X.J.; Tian, G.F. A new phenanthraquinone from the roots of Peucedanum praeruptorum. Chin. Chem. Lett. 2010, 21, 816–817. [Google Scholar] [CrossRef]
  131. Guan, F.L.; Jin, W.B.; Zhang, X.H.; Zhao, N.C. The Effects of Praeruptorin A on the Contraction of isolated Tracheal Smooth Muscle Previously Induced by High K and Acetylcholine. J. Chin. Med. Univ. 1994, 023, 549–552. [Google Scholar]
  132. Xiong, Y.Y.; Wang, J.S.; Wu, F.H.; Li, J.; Zhou, L.; Kong, L.Y. Effects of (±)-praeruptorin A on airway inflammation, airway hyperresponsiveness and NF-κB signaling pathway in a mouse model of allergic airway disease. Eur. J. Pharmacol. 2012, 683, 316–324. [Google Scholar] [CrossRef]
  133. Xiong, Y.Y.; Wang, J.S.; Wu, F.H.; Li, J.; Zhou, L.; Kong, L.Y. The effects of (±)-Praeruptorin A on airway inflammation, remodeling and transforming growth factor-β1/Smad signaling pathway in a murine model of allergic asthma. Int. Immunopharmacol. 2012, 14, 392–400. [Google Scholar] [CrossRef]
  134. Bayrami, G.; Boskabady, M.H.; Iranshahi, M.; Gholamnezhad, Z. Relaxant effects of asafoetida extract and its constituent umbelliprenin on guinea-pig tracheal smooth muscle. Chin. J. Integr. Med. 2013. online ahead of publishing. [Google Scholar] [CrossRef]
  135. Wang, D.C.; Zhang, X.Z.; Feng, L. Antioxidative Effects of Total Coumarins from Peucedanum praeruptorum Dunn invitro. Her. Med. 2008, 27, 899–901. [Google Scholar] [CrossRef]
  136. Ozaki, H.; Nishidono, Y.; Fujii, A.; Okuyama, T.; Nakamura, K.; Maesako, T.; Shirako, S.; Nakatake, R.; Tanaka, K.; Ikeya, Y.; et al. Identification of Anti-Inflammatory Compounds from Peucedanum praeruptorum Roots by Using Nitric Oxide-Producing Rat Hepatocytes Stimulated by Interleukin 1β. Molecules 2023, 28, 5076. [Google Scholar] [CrossRef] [PubMed]
  137. Menghini, L.; Epifano, F.; Genovese, S.; Marcotullio, M.C.; Sosa, S.; Tubaro, A. Antiinflammatory activity of coumarins from Ligusticum lucidum Mill. subsp. cuneifolium (Guss.) Tammaro (Apiaceae). Phytother. Res. 2010, 24, 1697–1699. [Google Scholar] [CrossRef] [PubMed]
  138. Yu, P.J.; Ci, W.; Wang, G.F.; Zhang, J.Y.; Wu, S.Y.; Xu, W.; Jin, H.; Zhu, Z.G.; Zhang, J.J.; Pang, J.X.; et al. Praeruptorin A inhibits lipopolysaccharide-induced inflammatory response in murine macrophages through inhibition of NF-κB pathway activation. Phytother. Res. 2011, 25, 550–556. [Google Scholar] [CrossRef] [PubMed]
  139. Yang, L.; Li, X.B.; Yang, Q.; Zhang, K.; Zhang, N.; Guo, Y.Y.; Feng, B.; Zhao, M.G.; Wu, Y.M. The neuroprotective effect of praeruptorin C against NMDA-induced apoptosis through down-regulating of GluN2B-containing NMDA receptors. Toxicol Vitro 2013, 27, 908–914. [Google Scholar] [CrossRef]
  140. Yu, P.J.; Jin, H.; Zhang, J.Y.; Wang, G.F.; Li, J.R.; Zhu, Z.G.; Tian, Y.X.; Wu, S.Y.; Xu, W.; Zhang, J.J.; et al. Pyranocoumarins Isolated from Peucedanum praeruptorum Dunn Suppress Lipopolysaccharide-Induced Inflammatory Response in Murine Macrophages Through Inhibition of NF-κB and STAT3 Activation. Inflammation 2012, 35, 967–977. [Google Scholar] [CrossRef]
  141. Chang, T.H.; Liu, X.Y.; Zhang, X.H.; Wang, H.L. Effects of dl-praeruptorin A on interleukin-6 level and Fas, bax, bcl-2 protein expression in ischemia-reperfusion myocardium. Acta Pharmacol. Sin. 2002, 23, 769–774. [Google Scholar]
  142. Wang, W.J.; Lu, H.Q.; Sun, L.M.; Zhong, P.; Sun, G. Effects of praeruptorin C on blood pressure and expression of phospholamban in spontaneously hypertensive rats. Phytomedicine 2014, 21, 195–198. [Google Scholar] [CrossRef]
  143. Sun, L.; Rao, M.R.; Liu, P.Q. Effects of praeruptorin C on cardiac dysfunction, myocardial compliance, and collagen content in renovascular hypertensive rats. Acta Pharm. Sin. 1997, 32, 578–582. [Google Scholar] [CrossRef]
  144. Rao, M.R.; Liu, W.B.; Liu, P.Q. Effects of praeruptorin C on vascular hypertrophy, [Ca2+] i, collagen content and NO in renovascular and spontaneously hypertensive rats. J. Chin. Pharm. Sci. 2002, 11, 111–114. [Google Scholar] [CrossRef]
  145. Rao, M.R.; Sun, L.; Zhang, X.W. Effects of praeruptorum coumarin on heart hemodynamics, myocardial compliance, and collagen content in heart hypertrophy rats. Chin. J. Pharmacol. Toxicol. 2002, 16, 265–269. [Google Scholar] [CrossRef]
  146. Joa, H.; Vogl, S.; Atanasov, A.G.; Zehl, M.; Nakel, T.; Fakhrudin, N.; Heiss, E.H.; Picker, P.; Urban, E.; Wawrosch, C.; et al. Identification of ostruthin from Peucedanum ostruthium rhizomes as an inhibitor of vascular smooth muscle cell proliferation. J. Nat. Prod. 2011, 74, 1513–1516. [Google Scholar] [CrossRef] [PubMed]
  147. Liang, T.; Yue, W.; Li, Q. Chemopreventive Effects of Peucedanum praeruptorum Dunn and Its Major Constituents on SGC7901 Gastric Cancer Cells. Molecules 2010, 15, 8060–8071. [Google Scholar] [CrossRef] [PubMed]
  148. Wu, M.H.; Lin, C.L.; Chiou, H.L.; Yang, S.F.; Lin, C.Y.; Liu, C.J.; Hsieh, Y.H. Praeruptorin A Inhibits Human Cervical Cancer Cell Growth and Invasion by Suppressing MMP-2 Expression and ERK1/2 Signaling. Int. J. Mol Sci. 2017, 19, 10. [Google Scholar] [CrossRef]
  149. Lin, C.L.; Hung, T.W.; Ying, T.H.; Lin, C.J.; Hsieh, Y.H.; Chen, C.M. Praeruptorin B Mitigates the Metastatic Ability of Human Renal Carcinoma Cells through Targeting CTSC and CTSV Expression. IJMS 2020, 21, 2919. [Google Scholar] [CrossRef] [PubMed]
  150. Nishino, H.; Okuyama, T.; Takata, M.; Shibata, S.; Tokuda, H.; Takayasu, J.; Hasegawa, T.; Nishino, A.; Ueyama, H.; Iwashima, A. Studies on the anti-tumor-promoting activity of naturally occurring substances. IV. Pd-II [(+) anomalin, (+) praeruptorin B], a seselin-type coumarin, inhibits the promotion of skin tumor formation by 12-O-tetradecanoylphorbol-13-acetate in 7,12-dimethylbenz[a]anthracene-initiated mice. Carcinogenesis 1990, 11, 1557–1561. [Google Scholar] [CrossRef] [PubMed]
  151. Liu, C.M.; Shen, H.T.; Lin, Y.A.; Yu, Y.L.; Chen, Y.S.; Liu, C.J.; Hsieh, Y.H. Antiproliferative and Antimetastatic Effects of Praeruptorin C on Human Non–Small Cell Lung Cancer through Inactivating ERK/CTSD Signalling Pathways. Molecules 2020, 25, 1625. [Google Scholar] [CrossRef] [PubMed]
  152. Zhou, X.; Bi, H.; Jin, J.; Niu, L.; Cai, D.; Deng, R.; Li, Y.; Wang, Y.; Huang, M. Effects of praeruptorin A and praeruptorin C, a racemate isolated from Peucedanum praeruptorum, on MRP2 through the CAR pathway. Planta Med. 2013, 79, 1641–1647. [Google Scholar] [CrossRef]
  153. Yu, P.J.; Li, J.R.; Zhu, Z.G.; Kong, H.Y.; Jin, H.; Zhang, J.Y.; Tian, Y.X.; Li, Z.H.; Wu, X.Y.; Zhang, J.J.; et al. Praeruptorin D and E attenuate lipopolysaccharide/hydrochloric acid induced acute lung injury in mice. Eur. J. Pharmacol. 2013, 710, 39–48. [Google Scholar] [CrossRef]
  154. Liu, X.; Chin, J.F.; Qu, X.; Bi, H.; Liu, Y.; Yu, Z.; Zhai, Z.; Qin, A.; Zhang, B.; Dai, M. The Beneficial Effect of Praeruptorin C on Osteoporotic Bone in Ovariectomized Mice via Suppression of Osteoclast Formation and Bone Resorption. Front. Pharmacol. 2017, 8, 627. [Google Scholar] [CrossRef]
  155. Han, S.; Li, C.; Huang, J.; Wei, F.; Zhang, Y.; Wang, S. Cell membrane chromatography coupled with UHPLC-ESI-MS/MS method to screen target components from Peucedanum praeruptorum Dunn acting on α1A adrenergic receptor. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2016, 1011, 158–162. [Google Scholar] [CrossRef] [PubMed]
  156. Zhao, P.; Wang, Y.; Huang, W.; He, L.; Lin, Z.; Zhou, J.; He, Q. Toxic effects of terpinolene on Microcystis aeruginosa: Physiological, metabolism, gene transcription, and growth effects. Sci. Total Environ. 2020, 719, 137376. [Google Scholar] [CrossRef] [PubMed]
  157. Moon, H.I.; Lee, J.H.; Lee, Y.C.; Kim, K.S. Antiplasmodial and cytotoxic activity of coumarin derivatives from dried roots of Angelica gigas Nakai in vitro. Immunopharmacol. Immunotoxicol. 2011, 33, 663–666. [Google Scholar] [CrossRef] [PubMed]
  158. Shakeri, F.; Boskabady, M.H. A review of the relaxant effect of various medicinal plants on tracheal smooth muscle, their possible mechanism(s) and potency. J. Ethnopharmacol. 2015, 175, 528–548. [Google Scholar] [CrossRef] [PubMed]
  159. Liu, J.; Kam, K.W.; Zhou, J.J.; Yan, W.Y.; Chen, M.; Wu, S.; Wong, T.M. Effects of heat shock protein 70 activation by metabolic inhibition preconditioning or kappa-opioid receptor stimulation on Ca2+ homeostasis in rat ventricular myocytes subjected to ischemic insults. J. Pharmacol. Exp. Ther. 2004, 310, 606–613. [Google Scholar] [CrossRef] [PubMed]
  160. Li, Z.; Zhang, F.; Wang, S.; Xiao, H.; Wang, J.; Li, X.; Yang, H. Endothelium-dependent vasorelaxant effects of praeruptorin a in isolated rat thoracic aorta. Bioengineered 2022, 13, 10038–10046. [Google Scholar] [CrossRef] [PubMed]
  161. Jia, L.G.; Donnet, C.; Bogaev, R.C.; Blatt, R.J.; McKinney, C.E.; Day, K.H.; Berr, S.S.; Jones, L.R.; Moorman, J.R.; Sweadner, K.J.; et al. Hypertrophy, increased ejection fraction, and reduced Na-K-ATPase activity in phospholemman-deficient mice. Am. J. Physiol. Heart Circ. Physiol. 2005, 288, H1982–H1988. [Google Scholar] [CrossRef]
  162. Wang, S.F.; Chen, P.H.; Jiang, W.; Wu, L.; Chen, L.H.; Fan, X.H.; Wang, Y.; Cheng, Y.Y. Identification of the effective constituents for anti-inflammatory activity of Ju-Zhi-Jiang-Tang, an ancient traditional Chinese medicine formula. J. Chromatogr. A. 2014, 1348, 105–124. [Google Scholar] [CrossRef]
  163. Jung, J.W.; Kim, N.J.; Yun, H.; Han, Y.T. Recent Advances in Synthesis of 4-Arylcoumarins. Molecules 2018, 23, 2417. [Google Scholar] [CrossRef]
  164. Zhao, Y.; Wang, N.; Sui, Z.; Huang, C.; Zeng, Z.; Kong, L. The Molecular and Structural Basis of O-methylation Reaction in Coumarin Biosynthesis in Peucedanum praeruptorum Dunn. Int. J. Mol. Sci. 2019, 20, 1533. [Google Scholar] [CrossRef]
  165. Kong, L.Y.; Wu, X.L.; Min, Z.D. Semi-synthesis of derivatives with C-3’ and C-4’trans-configuration from (+)-praeruptorin A. Acta Pharm. Sin. 2003, 38, 358–363. [Google Scholar] [CrossRef]
  166. Ban, X.X.; Jiang, A.W.; Du, P.S.; Liu, Y.N. Study on Acute Toxicity of Methanol Extract from Peucedanum praeruptorum Dunn (Pd) in Mice. J. Tradit. Chin. Med. 2015, 21, 49–51. [Google Scholar] [CrossRef]
  167. Hu, S.; Wang, P.; Chen, Y.; Kuang, G.; Wang, C.; Luo, J.; Chen, S. In Vitro Investigation of the Anticancer Activity of Peucedanum praeruptorum Dunn Extract on HepG2 Human Hepatoma Cells. J. Food Qual. 2023, 2023, 9010344. [Google Scholar] [CrossRef]
  168. Zhu, W.J.; Liu, Y.; Cao, Y.N.; Peng, L.X.; Yan, Z.Y.; Zhao, G. Insights into Health-Promoting Effects of Plant MicroRNAs: A Review. J. Agric. Food Chem. 2021, 69, 14372–14386. [Google Scholar] [CrossRef] [PubMed]
  169. Villani, V.; Di Marco, G.; Iacovelli, F.; Pietrucci, D.; Canini, A.; Gismondi, A. Profile and potential bioactivity of the miRNome and metabolome expressed in Malva sylvestris L. leaf and flower. BMC Plant Biol. 2023, 23, 439. [Google Scholar] [CrossRef]
  170. Gismondi, A.; Di Marco, G.; Canini, A. Detection of plant microRNAs in honey. PLoS ONE 2017, 12, e0172981. [Google Scholar] [CrossRef]
  171. Samad, A.F.A.; Kamaroddin, M.F.; Sajad, M. Cross-Kingdom Regulation by Plant microRNAs Provides Novel Insight into Gene Regulation. Adv. Nutr. 2021, 12, 197–211. [Google Scholar] [CrossRef]
Figure 1. The overall appearance (A), roots (B), flowers (C), blades (D) view, and distribution (E) of K. praeruptora. (The green shading in E represents the distribution of K. praeruptora).
Figure 1. The overall appearance (A), roots (B), flowers (C), blades (D) view, and distribution (E) of K. praeruptora. (The green shading in E represents the distribution of K. praeruptora).
Molecules 28 08153 g001
Figure 2. Anatomical differences between (AC) unbolting and (DF) bolting plants of K. praeruptora at different growth stages [23]. The unbolting roots’ parenchyma cells, initially undifferentiated, appear large and loose (A). The pericycle differentiates into conduits, woody tissue, and wood rays (B). Woody tissue parenchyma cells are mildly fibrose, and secretion ducts increase (C). Bolting plants show earlier pericycle differentiation and woody parenchyma fibrosis (D). Pericycle shrinks, cracks increase, secretion ducts decrease, and fibrosis intensifies, leading to pith formation in later stages (E,F).
Figure 2. Anatomical differences between (AC) unbolting and (DF) bolting plants of K. praeruptora at different growth stages [23]. The unbolting roots’ parenchyma cells, initially undifferentiated, appear large and loose (A). The pericycle differentiates into conduits, woody tissue, and wood rays (B). Woody tissue parenchyma cells are mildly fibrose, and secretion ducts increase (C). Bolting plants show earlier pericycle differentiation and woody parenchyma fibrosis (D). Pericycle shrinks, cracks increase, secretion ducts decrease, and fibrosis intensifies, leading to pith formation in later stages (E,F).
Molecules 28 08153 g002
Figure 3. Regulatory pathways of plant bolting and flowering.
Figure 3. Regulatory pathways of plant bolting and flowering.
Molecules 28 08153 g003
Figure 4. The structures of chemical constituents isolated from K. praeruptora.
Figure 4. The structures of chemical constituents isolated from K. praeruptora.
Molecules 28 08153 g004aMolecules 28 08153 g004b
Figure 5. A simple route to the synthesis of coumarin.
Figure 5. A simple route to the synthesis of coumarin.
Molecules 28 08153 g005
Figure 6. The pharmacological properties of K. praeruptora.
Figure 6. The pharmacological properties of K. praeruptora.
Molecules 28 08153 g006
Table 2. Pharmacological activities of K. praeruptora (“↑“ Upward arrow indicates an increase in expression levels. “↓” Downward arrow indicates a decrease in expression levels.).
Table 2. Pharmacological activities of K. praeruptora (“↑“ Upward arrow indicates an increase in expression levels. “↓” Downward arrow indicates a decrease in expression levels.).
Pharmacological ActivityCompounds/ExtractsMaterial/ModelTreatmentResultsReferences
Eliminate phlegm and relieve asthmaPraeruptorin ARabbits/trachea extricated and immersed in a nutritive solution containing a mixed gasIn vivo 30 µmol/L 0.5 h↓ Ca+ and PDC[131]
Mice/chronic asthma modelIn vivo 30, 60, 120 mg/kg 5 d↓ (IL)-4, IL-5, IL-13, LTC4 and (Ig) E, eotaxin protein and mRNA expression [132]
Mice/chronic asthma modelIn vivo 30, 60, 120 mg/kg 56 d↓ IL-4 and IL-13 in BALF, IgE, TGF-β1 and pSmad2/3 expression[133]
Honey-roasted K. praeruptora piecesMice/histamine phosphate asthma modelIn vivo 2, 4, 8 g/kg 3 d↑ Secretion of phenol red [130]
Coumarin constituentRabbitsIn vivo 2, 5, 10 mg/mL↓ KCl and acetylcholine level[134]
Anti-inflammatory and analgesicTCP MiceIn vivo 31.3, 62.5, 125, 250, 500 µg/mL↓ MDA[135]
Praeruptorin A, B, and ERat hepatocytesIn vivo 1 nmol/L↓ NO synthase expression
↓ mRNAs encoding
[136]
Praeruptorin ACroton oil-induced ear dermatitis in miceIn vivo 0.3 μmol/cm2↓ 22–43% oedema reduction[137]
Macrophage cellsIn vitro 6.25, 12.5, 25 mg/mL 24 h↓ NO, IL-1β and TNF-α, mRNA expressions, iNOSand TNF-α[138]
Mice/chronic asthma modelIn vivo 30, 60, 120 mg/kg 5 d↓ (IL)-4, IL-5, IL-13, LTC4 and (Ig) E, eotaxin protein and mRNA expression [132]
Mice/chronic asthma modelIn vivo 30, 60, 120 mg/kg 56 d↓ IL-4 and IL-13 in BALF, IgE, TGF-β1 and pSmad2/3 expression[133]
Praeruptorin CCortical neuron cellsIn vitro 0, 0.1, 1, 10 µmol/L↓ NMDA and Bcl-2[139]
Praeruptorin C, D, and EMacrophage cellsIn vitro 4, 8, 16 μg/mL 18 h↓ NO, IL-6 and TNF-α, mRNA and r κB-α protein expression[140]
Cardiovascular protectionPraeruptorin AOpen-chest anesthetized ratsIn vivo 2 mg/mL 18 h↓ IL-6, Fas, bax and bcl-2 level[141]
Praeruptorin CSpontaneously hypertensive ratsIn vivo 20 mg/kg 48 d↓ PLB mRNA expression
↑ HMI and LVMI
[142]
Renovascular hypertensive rats/2K1C-RHRIn vivo 20 mg/kg 63 d↑CF/HWW, CO/HWW, LVSP, -dp/d tmax value
↓ LVEDP and T value
[143]
Renovascular and spontaneously hypertensive ratsIn vivo 20 mg/kg 63 d↑ Ca2+ and NO level[144]
Cortical neuron cells In vitro 0, 0.1, 1, 10 µmol/L↓ NMDA and Bcl-2[140]
Praeruptorin A, B, C, and DHeart hypertrophy rats/2K1C-RHRIn vivo 30 mg/kg 63 d↑-dp/d tmax value
↓ LVEDP and HYP level
[145]
OstruthinVascular smooth muscle cellsIn vitro 1, 3, 10, 30 µmol/L 20 h↓ VSMC proliferation concentration[146]
Anti-tumor effectMethanolic extractSGC7901 gastric cancer cellsIn vitro 50, 100, 200, 300 µg/mL 24 h↑ LDH
↓ Cell proliferation
[147]
Praeruptorin AHeLa and SiHa cellsIn vitro 0, 10, 20, 30, 40, 50 µmol/L 24 h↓ MMP-2, cyclin D1, Skp2, ERK1/2
↑ TIMP-2, Rb, p16, p21, p27,
[148]
Praeruptorin BRenal cell carcinomaIn vitro 0, 10, 20, 30, 40, 50 µmol/L 24 h↓ EGFR, MEK, ERK, CTSC, CTSV, mRNA and protein expression[149]
HeLa cells and C3H10T1/2 cellsIn vitro 0, 10, 20, 30, 40, 50 µmol/L 140 d↓ Tumor promotion[150]
Praeruptorin CHuman lung cancer cell linesIn vitro 0, 10, 20,30 µmol/L 24 h↓ cyclin D, cathepsin D, ERK1/2
↑ p21 protein,
[151]
Praeruptorin A and CHepG2 CellsIn vitro 10, 25, 50, 100, 200 µmol/L 18 and 24 h↓ mRNA and protein expression[152]
Praeruptorin D and EMice/acute lung injuryIn vivo 0, 20, 40, 80 mg/kg 1 h↓ MPO, IκB-α and p65 protein translocation[153]
Other pharmacological effectsPraeruptorin CBone marrow
Monocyte/macrophage (BMM) cells
In vitro 0, 5, 10, 20 µmol/L 48 h↓ kappa B and c-Jun N-terminal kinase/mitogen-activated protein kinase[154]
Praeruptorin A, B, and CHEK293 cell lineIn vitro 1 mg/mL 1 hIdentified with α1A adrenergic receptor activity[155]
TerpinoleneHarmful algal blooms (HABs)In vivo 0.551, 0.881, 1.079, 1.233, 1.470 mmol/L 4 d↓ NR and GS activities
↑ COX II, ABC transporter, CaBPs expression
[156]
MarmesininHloroquine-sensitive strain of P. falciparum (D10)In vitro 0.1, 1, 5, 25, and 100 µmol/L 24 h↓ Antiplasmodial D10
↑ Cytotoxicity SK-OV-3
[157]
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

Wang, Q.; Ding, L.; Wang, R.; Liang, Z. A Review on the Morphology, Cultivation, Identification, Phytochemistry, and Pharmacology of Kitagawia praeruptora (Dunn) Pimenov. Molecules 2023, 28, 8153. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules28248153

AMA Style

Wang Q, Ding L, Wang R, Liang Z. A Review on the Morphology, Cultivation, Identification, Phytochemistry, and Pharmacology of Kitagawia praeruptora (Dunn) Pimenov. Molecules. 2023; 28(24):8153. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules28248153

Chicago/Turabian Style

Wang, Qi, Lulu Ding, Ruihong Wang, and Zongsuo Liang. 2023. "A Review on the Morphology, Cultivation, Identification, Phytochemistry, and Pharmacology of Kitagawia praeruptora (Dunn) Pimenov" Molecules 28, no. 24: 8153. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules28248153

Article Metrics

Back to TopTop