Next Article in Journal
New Isocoumarin and Pyrone Derivatives from the Chinese Mangrove Plant Rhizophora mangle-Associated Fungus Phomopsis sp. DHS-11
Next Article in Special Issue
The Therapeutic Value of Solanum Steroidal (Glyco)Alkaloids: A 10-Year Comprehensive Review
Previous Article in Journal
Synthesis, Quality Control and Preliminary Activity Evaluation of a New Compound HM475
Previous Article in Special Issue
Yohimbine Alleviates Oxidative Stress and Suppresses Aerobic Cysteine Metabolism Elevated in the Rat Liver of High-Fat Diet-Fed Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dipsacus and Scabiosa Species—The Source of Specialized Metabolites with High Biological Relevance: A Review

1
Department of Biology and Pharmaceutical Botany, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
2
Chair and Department of Pharmaceutical Botany, Medical College, Jagiellonian University, Medyczna 9, 30-688 Kraków, Poland
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(9), 3754; https://doi.org/10.3390/molecules28093754
Submission received: 5 March 2023 / Revised: 21 April 2023 / Accepted: 22 April 2023 / Published: 27 April 2023

Abstract

:
The genera Dipsacus L. and Scabiosa L. of the Caprifoliaceae family are widely distributed in Europe, Asia, and Africa. This work reviews the available literature on the phytochemical profiles, ethnomedicinal uses, and biological activities of the most popular species. These plants are rich sources of many valuable specialized metabolites with beneficial medicinal properties, such as triterpenoid derivatives, iridoids, phenolic acids, and flavonoids. They are also sources of essential oils. The genus Dipsacus has been used for centuries in Chinese and Korean folk medicines to treat bone (osteoporosis) and joint problems (rheumatic arthritis). The Korean Herbal Pharmacopoeia and Chinese Pharmacopoeia include Dipsaci radix, the dried roots of D. asperoides C.Y.Cheng & T.M.Ai. In addition, S. comosa Fisch. ex Roem & Schult. and S. tschiliiensis Grunning are used in traditional Mongolian medicine to treat liver diseases. The current scientific literature data indicate that these plants and their constituents have various biological properties, including inter alia antiarthritic, anti-neurodegenerative, anti-inflammatory, antioxidant, anticancer, and antimicrobial activities; they have also been found to strengthen tendon and bone tissue and protect the liver, heart, and kidney. The essential oils possess antibacterial, antifungal, and insecticidal properties. This paper reviews the key biological values of Dipsacus and Scabiosa species, as identified by in vitro and in vivo studies, and presents their potential pharmacological applications.

Graphical Abstract

1. Introduction

Fruits, vegetables, and herbs provide the body with many valuable specialized metabolites, often with pro-health properties. These include polyphenols, alkaloids, terpenoids, and essential oils, all of which display specific biological activities. Plants have been utilized in folk medicine in many countries for centuries because of their long-known therapeutic benefits. Not only have the beneficial medicinal properties of plants have been known for thousands of years, consumer interest in phytotherapy/herbal medicines and natural food supplements continues to grow, and in many countries, traditional medicine is the only mode of treatment for many diseases.
Although herbal materials have many ethnomedicinal benefits, their toxicity or potential side effects remain relatively unexplored, and their medical potential frequently lacks a scientific basis. By determining the chemical composition of plant extracts, it is possible to estimate their safety and biological activities and hence their potential as natural drugs. The use of medicinal plants as natural sources of compounds with inter alia antioxidant, anti-inflammatory, and anti-diabetic properties has drawn the attention of many researchers. Oxidative stress and the inflammatory response are associated with many neurological diseases (Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis), cardiovascular diseases (atherosclerosis), hepatic conditions, gastrointestinal diseases, and cancers [1,2,3].
Recent years have seen a greater interest in the genera Dipsacus L. (teasel in English) and Scabiosa L. (pincushions in English) [4], with the latter being poorly understood. The first mention of phytochemical studies on Dipsacus spp. dates to the 1920s [5]. In 2011, Zhao and Shi [5] reviewed the chemical composition and biological properties of the specialized metabolites of some Dipsacus species. Ten years later, Tao et al. [6] published a review of the literature regarding one Dipsacus species, D. asper Wall. ex C.B.Clarke, its chemical constituents, selected pharmacological activities, and pharmacokinetics. In 2018, Pinto et al. [7] provided a review of flavonoids and terpenoid derivatives identified in some species from Scabiosa and their biological effects. In recent years, a significant number of reports have been published on the chemical composition and biological properties of the two genera.
Therefore, the present review aims to provide an overview of the current literature (until December 2022) regarding the phytochemistry, biological activities, and toxicology of selected species of Dipsacus and Scabiosa. Several online databases, including PubMed, Google Scholar, Scopus, and ScienceDirect, were searched in the current review. These two genera include many synonymous Latin species names, meaning the same species, which can lead to confusion. In the present review, the species names are cited according to the authors of publications. The natural occurrence of Dipsacus and Scabiosa species as well as synonymous species names are described based on the data from The World Flora Online, Plants of the World Online, The Global Biodiversity Information Facility, and Flora of China [4,8,9,10] (Table 1).
The genera Dipsacus and Scabiosa currently belong to the Caprifoliaceae Juss. family (honeysuckle family) [7,8]. They were previously classified taxonomically in the Dipsaceae [7]. According to Plants of the World Online database [9], the Caprifoliaceae includes 33 accepted genera. This family has been divided into six subfamilies and one genus. Both genera Dipsacus and Scabiosa are classified into the subfamily Dipsacoideae and order Dipsacales [8,54]. The genus Dipsacus is widely distributed in Europe, North Africa, and Asia in North Myanmar and comprises 21 accepted species [9]. The native range of the genus Scabiosa includes Eurasia, Macaronesia to North Africa, Eritrea, and South Africa, with 66 accepted species [9]. In particular, numerous representatives of European Scabiosa species appear in the Mediterranean region [55,56]. Table S1 (Supplementary Material) presents accepted species of the genera Dipsacus and Scabiosa. Dipsacus includes various ornamental plants used in floristry for their decorative dried inflorescences. The dried inflorescences were previously used in the textile industry to clean and lift the nap on woolen fabrics [57,58]. The name dipsacus itself is believed to be derived from the Greek word for dipsa or thirst [9]. The genus name scabiosa is derived from the Latin word scabies or itch [9]. According to Akar [59], Scabiosa plants were traditionally used to treat scabies, skin sores, and other skin infections.

2. Traditional Medicinal Uses and Pharmacopoeial Monographs

Many Dipsacus and Scabiosa species are known in various traditional medicines, including traditional Chinese medicine. So far, among over 20 species of the genus Dipsacus and 60 species of Scabiosa, the phytochemical profiles and biological properties of only a few species are known. As such, there is a need to better understand the chemical composition of these genera and their potential medicinal value.

2.1. Dipsacus spp.

Numerous, recent reports have examined the potential therapeutic effects of Dipsacus genera. However, most phytochemical and biological studies have focused on the biological activities and health-promoting factors of D. asperoides C.Y.Cheng et T.M.Ai (=D. asper Wall. ex C.B.Clarke) [6]. The oldest report on Dipsacus genus can be found in Shen Nong’s Herbal Classic (Shen Nong Ben Cao Jing in Chinese) [6,11,18]. D. asper is widespread in the southern and northern regions of China such as the Hunan, Yunnan, Gansu, and Shanxi Provinces [12,19]. The growing demand for this species and its mass harvesting has significantly weakened its population in a natural state [19,60]. D. asperoides is cultivated on a large scale, mainly in Hefeng City, Hubei Province. This species is also cultivated in other Chinese cities, including Xichang, Sichuan, Xifeng, Guizhou, Jianchuan, and Yunnan Provinces as well as Jiangxi and Guangxi Provinces in China [18,60].
The roots of D. asperoides in China are commonly known as Xu Duan or Himalayan Teasel Roots [6,11,12,13,14,15,16,17,18,20,61]. Traditionally, in China and Korea, Dipsaci radix is known as the raw material used in treating joint disease (rheumatic arthritis) and bone diseases (osteoporosis, bone fractures), lumbar and knee pain, arthralgia, traumatic hematoma, uterine bleeding, and gynecological diseases; it is also used to strengthen muscles and improve liver and kidney functions [6,12,15,26,27,28,62,63,64]. The raw material is collected in autumn. Dipsaci radix is 5–15 cm in length and 0.5–2 cm in diameter, with slightly twisted or twisted longitudinal wrinkles and furrows. It is greyish-brown or yellowish-brown in color [28]. Dipsaci radix has a spicy, bitter, slightly sweet, then astringent taste [26,28,29].
Dipsaci radix can be subjected to diaphoretic-, salt-, and wine-processing methods [6]. Some researchers suggest that the procedure of herb processing may result in differences in specialized metabolite content and biological effects. Some studies indicate that wine processing yields higher levels of key compounds (e.g., asperosaponin V and VI, dipsacoside A and B, dipsacussaponin B, loganic acid, and chlorogenic acid) than crude Dipsaci radix [65,66]. Materials processed by rice wine may promote anti-osteoporosis, anti-inflammatory, anti-coagulant, and analgesic activities or have a beneficial effect on blood circulation [6,67,68].
Dipsaci radix is commonly available in the Chinese herbal medicine market. This herb is mainly produced in China, in the Provinces of Sichuan, Yunnan, Hubei, Hunan, Xizang, Jiangxi, Guizhou, and Guangxi [18,20]. Dipsaci radix has a pharmacopeial monograph in the Korean Herbal Pharmacopoeia (Korea Food and Drug Administration 2007) and the 10th Chinese Pharmacopoeia, 2015 edition. The raw material is standardized to contain akebia saponin D whose level should be above 2% [69]. Chun et al. [30] indicate that the traditional use of D. asperoides is associated with its analgesic and anti-inflammatory properties for treating inter alia rheumatoid arthritis and bone fractures. The Chinese Pharmacopoeia, 2015 edition, recommends 9–15 g as the daily dose of D. asper for humans [15].
In the Korean herbal medicine markets, Phlomidis radix from Phlomis umbrosa Turczaninow is often sold instead of Dipsaci radix or both raw materials are mixed [70]. This mistake results from the morphological similarity of these two dried raw materials and their names [69,70]. Both species have been used in the Korean medicine Sok-dan for the treatment of bone- and arthritis-related diseases and are listed in the Korean Herbal Pharmacopoeia [70].
HPLC/UV profiles of four samples of Phlomidis radix and 17 samples of Dipsaci radix demonstrated that loganin, sweroside, dipsanoside A, 3-O-[β-d-glu-(1→4)][α-l-rha-(1→3)]-β-d-glu(1→3)-α-l-rha-(1→2)-α-l-ara-hed 28-O-β-d-glu-(1→6)-β-d-glu ester, and akebia saponin D were not detected in Phlomidis radix. The levels of these compounds in Dipsaci radix varied depending on the origin of the material and the extraction methods [69]. Akebia saponin D (a quality indicator of Dipsaci radix) predominated, with a content range of 0.73–10.96% (w/w) [69]. Both herbs showed anti-osteoarthritis ability in a Sprague-Dawley rat model induced by monosodium iodoacetate intra-articular knee injections [70]. However, Phlomidis radix was found to be more effective, suggesting that it may be used as an alternative to Dipsaci radix. It was found that the 70% ethanol extract (200 mg/kg/day) administered as oral gavage for 21 days restored the weight-bearing ability of the hind paw, suppressed histopathological changes of the osteoarthritic knee, inhibited the serum levels of inflammation mediators tumor necrosis factor α (TNF-α) and interleukin-1β (IL-1β), and inhibited the over-expression of the gene encoding matrix-degrading metalloproteinases MMP-9 and MMP-13 in the knee joint tissue [70]. In addition, the ethanol extract of Dipsaci radix displayed a protective effect against the destruction of articular cartilage, elevated myeloperoxidase (MPO) and down-regulated dystonin gene expression, modulated WNT/β-catenin signaling pathway, suppressed gene expression of Adamts4, and increased the expression of cartilage collagen genes (e.g., Col2A1, Col9A1, and Col11A1) and SOX5, SOX9, and Frzb genes [30].
Another species of Dipsacus is D. fullonum L., commonly known as teasel or wild teasel [33,34]. Traditionally, D. fullonum has been used to treat Lyme disease [35] and eye infections in cattle [36]. D. fullonum is distributed naturally in Europe to the Caucasus and North-Western Africa [9]. A rich source of valuable ingredients used in the traditional medicine of the Kashmir Himalayas is D. inermis Wall., also known as Wopal haakh/Wopal Hak in the Kashmiri language. It is used in treating cold, fever, cough, sore throat, general fatigue, and body pain and has demonstrated stomachic and carminative properties [37,38,39,71]. The roots of D. japonicus Miq., commonly known as Tuc doan in Vietnam, show tonic, anodyne, and demulcent activities. The recommended daily dose is 10–20 g as a decoction, alcoholic maceration, powder, or pill [41]. Decoction of D. japonicus roots has been used in the traditional medicine of China and Vietnam for rheumatism, sprains, trauma, fractures, relieving joint pain and ostealgia, and hepatic and renal hypofunction [40,41]. D. japonicus is widespread in central and northern China, Korea, Japan, and Vietnam [9,40]. The leaves of D. sativus (L.) Honck. are used as an infusion for the treatment of cardiovascular diseases. This species was originally cultured in Europe and was introduced from Japan to China in 1929 [42].

2.2. Scabiosa spp.

With respect to Scabiosa genera, many studies indicate that species of this genus show antioxidant [42,43,46,50,52,53,56,72,73,74,75,76,77], anti-inflammatory [50], anti-diabetic [43,78], anti-hepatic fibrosis [47,79,80], anti-cancer [56,81], and antibacterial [56,75,77,82,83] properties. Many species of Scabiosa grow naturally in the Mediterranean region [55,56].
In Tunisia, species from Scabiosa were commonly applied for skin treatment [56]. In the Iberian Peninsula, an infusion of S. atropurpurea L. inflorescence is used externally on the skin as an anti-acne treatment and orally for measles, rubeola, and scarlet fever [44,84]. In northern Peru, the aerial parts have been used for menstrual regulation and in Iberia, as a veterinary diuretic [46]. S. atropurpurea is distributed throughout the Mediterranean, Europe, Asia, and southern Africa [46]. Genetic investigations show that S. atropurpurea has 97% similarity with S. tschiliensis and thus is also called Japanese scabiosa [46]. This species is known as Mor uyuzotu or Şeytanotu in Turkey, Ambarina in Northern Peru, and Escabiosa in the Iberian Peninsula [45]. The leaves and flowers of S. stellata L. were used in the traditional medicine of Morocco to treat heel cracks [46,48]. S. stellata is an endemic plant in North Africa and is commonly known as starflower pincushions [75]. Another endemic species in North Africa (Algeria, Egypt, Libya, Morocco, and Tunisia) is S. arenaria Forssk. [9,72]. In traditional Mongolian medicine, the inflorescences of S. comosa Fisch. ex Roem. et Schult. and S. tschilliensis Grunning (known as Lanpenhua in Chinese) are used for liver diseases [74,79]. S. tschiliensis is widespread in China (in Hebei Province) and the Inner Mongolia autonomy district [51,53] and is locally called Meng Gu Shan Luo Bo [51]. Qingganjiuwei powder (composed of nine herbal materials including S. comosa) is commonly used as an anti-fibrosis drug in patients with chronic hepatic disease in Inner Mongolia [85,86]. This drug is accepted by the Inner Mongolia Region Drug Administration [86]. The flowers of S. comosa (Scabiosae flos) are also an ingredient of Gurigumu-7 used in traditional Mongolian and Tibetan medicine to treat liver diseases. This preparation is in the form of a bitter and astringent powder [87].

3. Chemical Constituents of Dipsacus and Scabiosa Species

Dipsacus and Scabiosa have closely related phytochemical profiles [75] and include over 200 specialized metabolites. Some representatives of Dipsacus and Scabiosa biosynthesize chemical compounds of various classes, mainly triterpenoid derivatives (Table 2) [5,6,7,12,21,22,27,40,48,49,51,65,66,81,82,88,89,90,91,92,93,94,95,96], which possess a variety of bioactivities. In addition, iridoids [6,7,13,22,27,33,34,65,66,75,78,84,89,92,93,94,95,96,97,98,99,100] (Table 3), phenolic acids [6,24,27,29,33,34,43,46,53,65,66,73,74,75,76,81,89,92,93,94,95,97,99,101] (Table 4), and flavonoids [7,33,34,42,43,46,48,53,73,74,75,76,81,82,94] (Table 5) have also been reported. Further, few alkaloids have been found in D. asper such as cantleyine, venoterpine, gentianine, dipsaperine ((3S,5S)-5-carboxystrictosidic acid 22-loganin ester), 3β,5α-tetrahydrodesoxycordifoline lactam, and (3R,5S)-5-carboxyvincosidic acid 22-loganin ester [6,92,96,98,100], and lignans have been detected [6,13,46,96,97,99] (Table 6). Scabiosa species are also rich sources of flavonoids, mainly in the aerial parts and flowers [7,43,46,48,49,53,73,74,75,81,82,94]. Essential oils were also isolated from S. arenaria, S. atropurpurea, D. fullonum, and D. japonicus (Table 7) [35,46,83,102]. Fatty acids have also been detected in Scabiosa spp. and D. asper (Table 8) [6,49,94]. The specialized metabolites identified in Dipsacus and Scabiosa are listed in Table 2, Table 3, Table 4, Table 5, Table 6, Table 7 and Table 8. The structures of selected compounds that have displayed some biological activities in a number of studies are shown in Figure 1.

3.1. Terpenoid Derivatives

The most diverse group of specialized metabolites in Dipsacus and Scabiosa are the triterpenoid derivatives, which can be divided into oleanane-type, hederagenin-type, or ursane-type (Table 2). Some compounds are derived from pomoic acid (scabiosaponin H-I) [7,51]. In Dipsacus, the main group of triterpenoids identified is hederagenin and its related saponins, while the rarest is ursane-type terpenoids [5] (Table 2). The oleanane-type triterpenoids were also common in Scabiosa genus (for example, oleanolic acid, scabiosaponins A-G, scabiostellatosides A-F, and hookeroside A and B) (Table 2) [7,48,49,51,82,94]. Some of these specialized metabolites such as oleanolic acid and ursolic acid were detected in the species of both genera [6,7,12,48,49,82,89,94]. Some of them were genus specific, such as hookeroside A and B and scabiosaponin A-K identified in S. tschiliensis whole plants [7,51] and palustroside III and scabiostellatosides A-H detected in S. stellata whole plants [7,48]. The new, tentatively detected triterpenoid derivatives in S. atropurpurea subsp. maritima leaves were oleanolic acid-pentosyl-rhamnosyl-pentosyl-glucosyl-di-glucoside, oleanolic acid-pentosyl-rhamnosyl-glucosyl-glucosyl-di-glucoside, and maslinic acid-pentosyl-rhamnosyl-glucosyl-glucosyl [81]. Yu et al. [12,22,96] reported some new arborinane-type triterpenoid (25-acetoxy-28-dehydroxyrubiarbonone E), ursane-type triterpenoids (2α,3β,24-trihydroxy-23-norurs-12-en-28-oic acid and 2α,3β-dihydroxy-23-norurs-4(24),11,13(18)-trien-28-oic acid), and oleanane-type triterpenoids (2′,3′-O-diacetyl-3-O-α-l-arabinopyranosyl-23-hydroxyolea-12-en-28-oic acid, 2′,4′-O-diacetyl-3-O-α-l-arabinopyranosyl-23-hydroxyolea-12-en-28-oic acid, and 3-O-β-d-glucopyranosyl-(1→3)-α-l-rhamnopyranosyl-(1→2)-α-l-arabinopyranosyl-23-hydroxyolean-18-en-28-oic acid 28-O-β-d-glucopyranosyl-(1→6)-β-d-glucopyranosyl ester) in D. asper roots. A new compound elucidated in D. japonicus roots was saponin XII [40]. In addition, this species has presented the only example to date of japondipsaponin E1 [5,88].
The indicator of the quality of Dipsaci radix is akebia saponin D (asperosaponin VI) [65]. It is known that environmental conditions, geographic location, the growth stage of the plants, or the year of harvest affect the content of the specialized metabolites in plants. Jin et al. [60] demonstrated that the content of akebia saponin D varied in D. asperoides roots collected from different geographical regions of China (i.e., Guizhou, Hubei, Sichuan, and Yunnan Provinces). The highest akebia saponin D content (about 6% of dry weight) was noted in roots collected from Hubei. A similar observation was noted by Du et al. [95]. The level of akebia saponin D ranged from 1.61% to 15.19% in samples of different origin.
Table 2. Terpenoid derivatives identified in Dipsacus and Scabiosa.
Table 2. Terpenoid derivatives identified in Dipsacus and Scabiosa.
No.Compound NameIdentificationSpecies (Part of Plant)
1.(6α,11α)-6-[(2′-O-acetyl-
α-l-arabinopyranosyl)oxy]-3-oxotaraxast-20-ene-11,28-diyl diacetate
NMRD. asper (roots) [12]
2.2′-O-acetyl-akebia saponin D;
2′-O-acetyl-
2′-O-acetyl-3-O-α-l-arabinopyranosyl-23-hydroxyolea-
12-en-28-oic acid;
3-O-(2-O-acetyl)-α-l-arabinopyranosyl-hederagenin 28-O-β-d-glucopyranosyl-(1-6)-β-d-glucopyranoside
HPLC-ESI-QTOF-MS/MS [93], R-ESI-MS, 1H NMR, 13C NMR [12,22,96]D. asper (roots) [12,22,93,96]
3.2α-hydroxyursolic acidNMRD. asper (roots) [12]
4.2α-hydroxy-
3β-O-trans-feruloyloxy-olean-12-en-28-oic acid
NMRD. asper (roots) [12]
5.2α,3β-dihydroxy-
23-norolea-4(24),12-dien-28-oic acid
NMRD. asper (roots) [12]
6.2α,3β-dihydroxy-23-norurs-4(24),11,13(18)-trien-28-oic acidHR-ESI-MS, 1H NMR, 13C NMR, 1H-1H COSY, ROESYD. asper (roots) [12]
7.2α,3β-dihydroxy-
24-norurs-4(23),12-dien-28-oic acid
NMRD. asper (roots) [12]
8.2α,3β,24-trihydroxy-23-norurs-12-en-28-oic acidHR-ESI-MS, 1H NMR, 13C NMR, DEPT, 1H-1H COSY, HMBC, NOESYD. asper (roots) [12]
9.2α,23α-dihydroxy-3β-O-trans-feruloyloxy-
olean-12-en-28-oic acid
NMRD. asper (roots) [12]
10.2′,3′-O-diacetyl-3-O-α-l-arabinopyranosyl-23-hydroxyolea-12-en-28-oic acidHR-ESI-MS, 1H NMR, 13C NMR, HMBCD. asper (roots) [12]
11.2′,4′-O-diacetyl-3-O-α-l-arabinopyranosyl-23-hydroxyolea-12-en-28-oic acidHR-ESI-MS, 1H NMR, 13C NMR, HMBCD. asper (roots) [12]
12.3β-O-trans-feruloyl-2α-hydroxy-urs-12-en-28-oic acidNMRD. asper (roots) [12]
13.3β-
O-trans-feruloyl-2α,23α-dihydroxy-urs-12-en-28-oic acid
NMRD. asper (roots) [12]
14.3β-hydroxy-24-norurs-
4(23),12-dien-28-oic acid
NMRD. asper (roots) [12]
15.3-O-α-l-arabinopyranosylhederagenin
28-O-β-d-glucopyranoside
NMRD. asper (roots) [96]
16.3-O-α-l-arabinopyranosyl-28-O-β-d-glucopyranosyl-(16)-β-d-glucopyranosyloleanolic acidNMR [96], HPLC-ESI-QTOF-MS/MS [93]D. asper (roots) [92,93,96]
17.3-O-β-d-Glucopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-α-l-arabinopyranosyl-
23-hydroxyolean-18-en-28-oic acid 28-O-β-Dglucopyranosyl-(16)-β-d-glucopyranosyl ester
HR-ESI-MS, 1D-TOCSY, 2D-HSQC, TOCSY-HSQC, COSY, HMBCD. asper (roots) [96]
18.3-O-[β-d-xylopyranosyl-
(14)-β-d-glucopyranosyl-(14)][α-l-rhamnopyranosyl-(13)]-β-
d-glucopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-α-l-arabinopyranosylhederagenin
NMRD. asper (roots) [96]
19.3′-O-acetyl-akebia saponin D;
3′-O-acetyl-3-O-α-l-arabinopyranosylhederagenin
28-O-β-d-glucopyranosyl-(16)-β-d-glucopyranoside;
3′-O-acetyl-3-
O-α-l-arabinopyranosyl-23-hydroxyolea-12-en-28-oic acid;
3-O-(3-O-acetyl)-α-l-arabinopyranosyl-hederagenin 28-O-β-d-glucopyranosyl-(1-6)-β-d-glucopyranoside
HR-ESI-MS, 1H NMR, 13C NMR [12,22,96], HPLC-ESI-QTOF-MS/MS [93]D. asper (roots) [12,22,93,96]
20.4′-O-acetyl-akebia saponin D
(asperosaponin Ⅳ) a;
4′-O-acetyl-3-O-α-l-arabinopyranosylhederagenin 28-O-
β-d-glucopyranosyl-(16)-β-d-glucopyranoside;
4′-O-acetyl-3-O-α-l-arabinopyranosyl-23-hydroxyolea-
12-en-28-oic acid;
3-O-(4-O-acetyl)-
α-l-arabinopyranosyl-hederagenin 28-O-β-d-glucopyranosyl-(1-6)-β-d-glucopyranoside
HPLC-ESI-QTOF-MS/MS [93],
HR-ESI-MS, 1H NMR, 13C NMR [12,22,96]
D. asper (roots) [12,22,92,93,96]
21.11α,12α-epoxy-3,6β-dihydroxy-24-norurs-3-en-2-on-
(2813)-olide
NMRD. asper roots) [12]
22.23α-hydroxy-olean-12-en-3-
one
NMRD. asper (roots) [12]
23.25-acetoxy-28-dehydroxyrubiarbonone EHR-ESI-MS, 1D NMR, 2D NMR, 1H-1H COSY, HMBC, NOESYD. asper (roots) [12]
24.Akebia saponin PA
(cauloside A; leontoside A; 3-O-α-l-arabinopyranosyl hederagenin) a
HPLC-ESI-QTOF-MS/MS [93], FAB-MS, 1D NMR, 2D NMR [21,22], HR-ESI-MS [22]D. asper/D. asperoides (roots) [27,93]
D. asper (roots) [6,21,22]
D. asper (roots) [12]
25.Akebia saponins X-YHPLC-ESI-QTOF-MS/MS [93]D. asper (roots) [93]
26.Asperosaponin B D. asper (roots) [93]
27.Asperosaponin E
(3-O-β-d-glucopyranosyl-(14)-[a-l-rhamnopyranosyl-(16)]-β-d-glucopyranosyl-(13)-a-l-rhamnopyranosyl-(12)-a-l-arabinopyranosyl oleanolic acid)
HPLC-ESI-QTOF-MS/MS [93]D. asper (roots) [93]
28.Asperosaponin F
(3-O-α-l-rhamnopyranosyl-(›6)-β-d-glucopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-α-l-arabinopyranosyl oleanolic acid)
HPLC-ESI-QTOF-MS/MS [93]D. asper (roots) [93]
29.Asperosaponin G
(3-O-β-glucopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-α-l-arabinopyranosyl oleanolic acid)
HPLC-ESI-QTOF-MS/MS [93]D. asper (roots) [93]
30.Asperosaponin VUPLC-Q-TOF-MSD. asper (roots) [66]
31.Asperosaponin VI (akebia saponin D) a
(3-O-α-l-arabinopyranosyl hederagenin-28-β-d-glucopyranoside-(16)-
β-d-glucopyranoside)
UHPLC-MS/MS [65], UHPLC-Q-TOF-MS [66], LC-ESI-MS [90], HPLC-ESI-QTOF-MS/MS [93], HPLC-DAD [95]D. asper/D. asperoides (roots) [65,66,89,90,92,93,95]
32.Colchiside
(3-O-β-d-xylopyranosyl-23-O-β-d-glucopyranosyl-28-O-β-d-(6-O-acetyl)-
glucopyranosyl hederagenin)
1D NMR, 2D NMR, DEPT, TOCSY, HMQC, HMBCD. asper (roots) [21]
33.Dipsacoside AUPLC-Q-TOF-MSD. asper Wall. (roots) [66]
34.Dipsacoside BUHPLC-MS/MS [65]D. asperoides (roots) [27,65]
35.Dipsacus saponin AHR-ESI-MS, 1H NMR, 13C NMR [22]D. asper (roots) [6,22,92]
36.Dipsacus saponin BUPLC-Q-TOF-MS [66], HPLC-ESI-QTOF-MS/MS [93]D. asper/D. asperoides (roots) [6,27,66,93]
37.Dipsacus saponin CESI-QTOF-MS/MS [93]D. asper/D. asperoides (roots) [6,27,91,93]
38.Dipsacus saponins J-KESI-QTOF-MS/MS [93]D. asper (roots) [6,93]
39.Dipsacus saponin L
(3-O-α-l-rhamnopyranosyl-(12)-α-l-arabinopyranosyl hederagenin)
ESI-QTOF-MS/MSD. asper (roots) [93]
40.Dipsacus saponin M
(3-O-α-l-rhamnopyranosyl-(12)-α-l-arabinopyranosyl hed-eragenin 28-O-β-d-glucopyranosyl-(16)-β-d-glucopyranosyl)
ESI-QTOF-MS/MSD. asper (roots) [93]
41.Dipsacus saponin N
(3-O-β-d-xylopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-α-l-arabinopyranosyl hederagenin)
ESI-QTOF-MS/MSD. asper (roots) [93]
42.Dipsacus saponin OESI-QTOF-MS/MSD. asper (roots) [93]
43.Dipsacus saponin P
(3-O-β-d-xylopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-α-l-arabinopyranosyl hederagenin 28-O-β-d-glucopyranosyl-(16)-β-d-glucopyranosyl)
ESI-QTOF-MS/MSD. asper (roots) [93]
44.Dipsacus saponin R
(3-O-α-l-rhamnopyranosyl(13)-β-d-glucopyranosyl (13)-α-l-rhamnopyranosyl-(12)-α-l-arabinopyranosyl-hederagenin)
ESI-QTOF-MS/MSD. asper (roots) [93]
45.Dipsacus saponin V-D. asper [6]
46.Dipsacus saponin VIHR-ESI-MS, 1H NMR, 13C NMR [22]D. asper (roots) [6,22]
47.Dipsacus saponin VII-D. asper [6]
48.Dipsacus saponins IX-XI-D. asper [6]
49.Dipsacus saponin XIIESI-QTOF-MS/MS [93]D. asper (roots) [6,93]
50.Dipsacus saponin XIII-D. asper [6]
51.Elmalienoside BNMRD. asper (roots) [96]
52.α-hederin-D. asper [6]
53.Hederagenin-D. asper [6]
54.Hederagenin 28-
O-β-d-glucopyranosyl-(16)-β-d-glucopyranoside
NMRD. asper (roots) [96]
55.Hederagonic acidNMRD. asper (roots) [12]
56.HN saponin FESI-QTOF-MS/MS [93]D. asper (roots) [6,93]
57.Hookeroside A-BNMRS. tschilliensis (whole plants) [7,51]
58.Japondipsaponin E1-D. japonicus (roots) [5,88]
59.Kalopanax saponin A-D. asper (roots) [5]
60.Oleanolic acid1H NMR, 13C NMR [82], GC-MS [49]D. asper (roots) [6,89]
S. arenaria (aerial parts) [82]
S. stellata (aerial parts) [49]
S. tschilliensis (flowers) [94]
61.Macranthoidin AESI-QTOF-MS/MS [93], NMR [12,96]D. asper (roots) [6,12,93,96]
62.Macranthoside B-D. asper [6]
63.Maslinic acidNMRD. asper (roots) [12]
64.Maslinic acid-pentosyl-rhamnosyl-glucosideLC-MS/MSS. atropurpurea subsp. maritima (leaves) [81]
65.Oleanolic acid-pentosyl-rhamnosyl-glucosyl-glucosyl-di-glucosideLC-MS/MSS. atropurpurea subsp. maritima (leaves) [81]
66.Oleanolic acid-pentosyl-rhamnosyl-pentosyl-glucosyl-di-glucosideLC-MS/MSS. atropurpurea subsp. maritima (leaves) [81]
67.Palustroside III
(3-O-[β-d-glucopyranosyl-(12)-β-d-glucuronopyranosyl]-28-O-[β-d-glucopyranosyl]-hederagenin)
-S. stellata (whole plants) [7,48]
68.Saponin XII
(3-O-[β-d-glucopyranosyl(14)][α-l-rhamnopyranosyl(13)]-
emph-β-d-glucopyranosyl(13)-α-l-rhamnopyranosyl(12)-α-arabinopyranosyl hederagenin 28-O-β-d-glucopyranosyl(16)-β-d-glucopyranoside)
1D NMR, 2D NMR, DEPT, HR-ESI-MSD. japonicus (roots) [40]
69.Scabiosaponin A
(3-O-β-d-glucopyranosyl-(14)-β-d-xylopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-α-l-arabinopyranosyloleanolic acid 28-O-β-d-glucopyranosyl-(16)-β-d-glucopyranoside)
2D NMR, DEPT, DQF-COSY, TOCSY, HMQC, HMQC-TOCSY, HMBC, NOESY [51]S. tschilliensis (whole plants) [7,51]
70.Scabiosaponin B
(3-O-β-d-xylopyranosyl-(14)-β-d-glucopyranosyl-(14)-β-d-xylopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-α-l-arabinopyranosyloleanolic
acid 28-O-β-d-glucopyranosyl-(16)-β-d-glucopyranoside)
2D NMR, DEPT, DQF-COSY, TOCSY, HMQC, HMQC-TOCSY, HMBC, NOESY [51]S. tschilliensis (whole plants) [7,51]
71.Scabiosaponin C
(3-O-[β-d-glucopyranosyl-(14)-β-d-xylopyranosyl-(13)-α-l-rhamnopyranosy-(12)][β-d-glucopyranosyl-(14)]-α-l-arabinopyranosyloleanolic
acid 28-O-β-d-glucopyranosyl-(16)-β-d-glucopyranoside)
2D NMR, DEPT, DQF-COSY, TOCSY, HMQC, HMQC-TOCSY, HMBC, NOESY [51]S. tschilliensis (whole plants) [7,51]
72.Scabiosaponin D
(3-O-[α-l-rhamnopyranosyl-(12)][β-d-glucopyranosyl-(14)]-α-l-arabinopyranosyloleanolic acid 28-O-β-d-glucopyranosyl-(16)-β-d-glucopyranoside)
2D NMR, DEPT, DQF-COSY, TOCSY, HMQC, HMQC-TOCSY, HMBC, NOESY [51]S. tschilliensis (whole plants) [7,51]
73.Scabiosaponin E
(3-O-β-d-xylopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-β-d-xylopyranosyloleanolic acid 28-O-β-d-glucopyranosyl-(16)-
β-d-glucopyranoside)
2D NMR, DEPT, DQF-COSY, TOCSY, HMQC, HMQC-TOCSY, HMBC, NOESY [51]S. tschilliensis (whole plants) [7,51]
74.Scabiosaponin F
(3-O-β-d-glucopyranosyl-
(13)-α-l-rhamnopyranosyl-(12)-β-d-xylopyranosyloleanolic acid 28-O-β-d-glucopyranosyl-(16)-β-d-glucopyranoside)
2D NMR, DEPT, DQF-COSY, TOCSY, HMQC, HMQC-TOCSY, HMBC, NOESY [51]S. tschilliensis (whole plants) [7,51]
75.Scabiosaponin G
(3-O-β-d-glucopyranosyl-(14)-β-d-glucopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-β-d-xylopyranosyloleanolic acid 28-O-β-d-glucopyranosyl-(16)-β-d-glucopyranoside)
2D NMR, DEPT, DQF-COSY, TOCSY, HMQC, HMQC-TOCSY, HMBC, NOESY [51]S. tschilliensis (whole plants) [7,51]
76.Scabiosaponin H
(3-O-β-d-glucopyranosyl-(14)-β-d-glucopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-α-L-arabinopyranosylpomolic acid 28-O-β-d-glucopyranosyl-(16)-β-d-glucopyranoside)
2D NMR, DEPT, DQF-COSY, TOCSY, HMQC, HMQC-TOCSY, HMBC, NOESY [51]S. tschilliensis (whole plants) [7,51]
77.Scabiosaponin I
(3-O-β-d-glucopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-α-l-arabinopyranosylpomolic acid 28-O-β-d-glucopyranosyl-(16)-β-d-glucopyranoside)
2D NMR, DEPT, DQF-COSY, TOCSY, HMQC, HMQC-TOCSY, HMBC, NOESY [51]S. tschilliensis (whole plants) [7,51]
78.Scabiosaponin J
(3-O-β-d-glucopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-α-l-arabinopyranosylsiaresinolic acid 28-O-β-d-glucopyranosyl-(16)-β-d-glucopyranoside)
2D NMR, DEPT, DQF-COSY, TOCSY, HMQC, HMQC-TOCSY, HMBC, NOESY [51]S. tschilliensis (whole plants) [7,51]
79.Scabiosaponin K
(3-O-β-d-glucopyranosyl-(14)-β-d-xylopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-α-l-arabinopyranosylsiaresinolic acid 28-O-β-d-glucopyranosyl-(16)-β-d-glucopyranoside)
2D NMR, DEPT, DQF-COSY, TOCSY, HMQC, HMQC-TOCSY, HMBC, NOESY [51]S. tschilliensis (whole plants) [7,51]
80.Scabiostellatoside A
(3-O-[β-d-glucopyranosyl-(14)-β-d-xylopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-α-l-arabinopyranonosyl]-28-O-[β-d-glucopyranosyl-(16)-β-d-glucopyranosyl]-oleanolic acid)
HR-ESI-MS, 1D NMR, 2D NMR, HMBC, COSY, TOCSY, HSQC, ROESY [48]S. stellata (whole plants) [7,48]
81.Scabiostellatoside B
(3-O-α-l-rhamnopyranosyl-(13)-β-d-xylopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-β-d-xylopyranosyl]-28-O-[β-d-glucopyranosyl-(16)-β-d-glucopyranosyl]-oleanolic acid)
HR-ESI-MS, 1D NMR, 2D NMR, HMBC, COSY, TOCSY, HSQC, HSQC-TOCSY, ROESY [48]S. stellata (whole plants) [7,48]
82.Scabiostellatoside C
(3-O-[β-d-glucopyranosyl-(14)-α-l-rhamnopyranosyl-(13)-β-d-xylopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-a-l-arabinopyranosyl]-28-O-[β-d-glucopyranosyl-(16)-β-d-glucopyranosyl]-oleanolic acid)
HR-ESI-MS, 1D NMR, 2D NMR, HMBC, COSY, TOCSY, HSQC, HSQC-TOCSY, ROESY [48]S. stellata (whole plants) [7,48]
83.Scabiostellatoside D
(3-O-[β-d-glucopyranosyl-(14)-α-l-rhamnopyranosyl-(13)-β-d-xylopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-β-d-xylopyranonosyl]-28-O-[β-d-glucopyranosyl-(16)-β-d-glucopyranosyl]-oleanolic acid)
HR-ESI-MS, 1D NMR, 2D NMR, HMBC, COSY, TOCSY, HSQC, HSQC-TOCSY, ROESY [48]S. stellata (whole plants) [7,48]
84.Scabiostellatoside E
(3-O-[α-l-rhamnopyranosyl-
(13)-β-d-xylopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-{β-d-glucopyranosyl-(14)-}α-l-arabinopyranosyl]-28-O-[β-Dglucopyranosyl-(16)-β-d-glucopyranosyl]-oleanolic acid)
HR-ESI-MS, 1D NMR, 2D NMR, HMBC, COSY, TOCSY, HSQC, HSQC-TOCSY, ROESY [48]S. stellata (whole plants) [7,48]
85.Scabiostellatoside F
(3-O-[β-d-glucopyranosyl-(14)-β-d-glucopyranosyl-(14)-β-d-glucopyranosyl-(14)-α-l-rhamnopyranosyl-(13)-β-d-xylopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-β-d-xylopyranosyl]-
oleanolic acid)
HR-ESI-MS, 1D NMR, 2D NMR, HMBC, COSY, TOCSY, HSQC, HSQC-TOCSY, ROESY [48]S. stellata (whole plants) [7,48]
86.Scabiostellatoside G
(3-O-[β-d-glucopyranosyl-(12)-β-d-glucuronopyranosyl]-28-O-[β-d-glucopyranosyl-(16)-β-d-glucopyranosyl]-hederagenin)
HR-ESI-MS, 1D NMR, 2D NMR, HMBC, COSY, TOCSY, HSQC, HSQC-TOCSY, ROESY [48]S. stellata (whole plants) [7,48]
87.Scabiostellatoside H
(3-O-[α-l-rhamnopyranosyl-(12)-β-d-glucuronopyranosyl]-28-O-[β-d-glucopyranosyl]-asiatic acid)
HR-ESI-MS, 1D NMR, 2D NMR, HMBC, COSY, TOCSY, HSQC, HSQC-TOCSY, ROESY [48]S. stellata (whole plants) [7,48]
88.Urceolide-S. tschilliensis (flowers) [94]
89.Ursolic acidNMR [12], GC-MS [49]D. asper (roots) [12]
S. stellata (whole plants) [7,48,49]
a synonymous names of compounds are given in accordance with PubChem database [109].

Iridoids 

Iridoids were found in both genera (Table 3). The first iridoids isolated from D. asper were loganin and sweroside [6]. The presence of iridoid glycosides including loganin and loganic acid was widespread in the roots or leaves of Dipsacus spp. [6,22,27,33,34,65,66,89,92,93,95,96,97,99] and flowers of S. atropurpurea subsp. maritima [7,84] (Table 3). Dipsanosides A-N, dipsasperoside A and B, lisianthioside, triplostoside A, and cocculoside were characteristic only for roots of D. asper [6,13,22,89,92,93,96,98,99,100]. Sweroside and sylvestrosides III-IV were found in two Dipsacus species: D. asper roots and D. fullonum leaves and roots [6,22,27,33,34,65,66,92,93,96,99]. Sweroside and sylvestrosides I and II were also detected in some Scabiosa species, such as S. stellata whole plants, S. tschilliensis, and S. atropurpurea subsp. maritima flowers (Table 3) [7,75,84,94]. 7-O-caffeoyl-sylvestroside I and 7-O-(p-coumaroyl)-sylvestroside I were isolated as new compounds from the whole plants of S. stellata [7,75]. Interestingly, three iridoid-like compounds, viz., eustomoside, eustomoruside, and septemfidoside, were identified only in S. stellata [7,75]. It should be also noted that these iridoids have not been previously described in Caprifoliaceae [75]. Atropurpurin A-B and secologanin-methyl-hemiacetal were reported for the first time in Scabiosa spp. in S. atropurpurea subsp. maritima [78]. Lehbili et al. [75] suggested that Dipsacus and Scabiosa are closely related to one another; septemfidoside, sylvestroside I, and its derivatives (e.g., 7-O-caffeoyl-sylvestroside I and 7-O-[p-coumaroyl]-sylvestroside I) isolated from S. stellata whole plants are closely related to the bis-iridoids, cantleyoside identified in S. atropurpurea subsp. maritima [7,84], Dipsaci radix [6,89,92,93,97,99], and D. fullonum [33] as well as dipsanosides C-G detected in D. asper [6,89]. The bis-iridoids identified in Dipsacus spp. possess a secoiridoid/iridoid subtype skeleton consisting of secologanic acid condensed to the 7-OH of loganin or loganin-like iridoids [89].
The levels of the main iridoids in Dipsaci radix, e.g., loganic acid, loganin, sweroside, and dipsanosides A and B, varied according to location and collection time. The highest level of loganic acid was 31.223 mg/g; loganin, 4.411 mg/g; sweroside, 8.364 mg/g; dipsanoside A, 4.513 mg/g; and dipsanoside B, 7.426 mg/g [99]. Du et al. [95] also demonstrated that the content of loganic acid (0.71–1.10%) and loganin (0.29–0.61%) varied between samples from different origins in China. In addition, the level of some iridoids in the leaves and roots including loganic acid, loganin, sweroside, sylvestroside III, and cantleyoside detected in D. fullonum varied according to the time of year of harvesting [33]. The leaves of the plants collected in the first year of vegetation contained a higher total iridoid content than the roots and leaves harvested in the second year. The most abundant iridoids were sylvestroside III in the leaves and cantleyoside in the roots. Moreover, sweroside (0.67 mg/g dry weight) and sylvestroside III (34.8 mg/g d.w.) were present in higher amounts in leaves, while the levels of loganic acid (5.27 mg/g d.w.), loganin (3.02 mg/g d.w.), and cantleyoside (21.41 mg/g d.w.) were higher in the roots [33].
Table 3. Iridoids identified in Dipsacus and Scabiosa species.
Table 3. Iridoids identified in Dipsacus and Scabiosa species.
No.Compound NameSpecies (Part of Plant)
1.3′-O-β-d-glucopyranosyl swerosideD. asper (roots) [89]
2.6′-O-β-d-apiofuranosyl swerosideD. asper (roots) [89,99]
3.7-O-caffeoyl-sylvestroside IS. stellata (whole plants) [7,75]
4.7-O-(p-coumaroyl)-sylvestroside IS. stellata (whole plants) [7,75]
5.Atropurpurin AS. atropurpurea subsp. maritima (whole plants) [78]
6.Atropurpurin BS. atropurpurea subsp. maritima (whole plants) [78]
7.CantleyosideD. asper (roots) [6,89,92,93,97,99]
D. fullonum (leaves and roots) [33]
S. atropurpurea subsp. maritima (roots) [7,84]
8.CocculosideD. asper (roots) [96]
9.Dipsanoside A and dipsanoside BD. asper (roots) [6,22,92,93,99]
10.Dipsanosides C-GD. asper (roots) [6,89]
11.Dipsanoside HD. asper [6]
12.Dipsanosides M-ND. asper (roots) [6,13]
13.Dipasperoside AD. asper (roots) [100]
14.Dipasperoside BD. asper (roots) [98]
15.EustomorusideS. stellata (whole plants) [7,75]
16.EustomosideS. stellata (whole plants) [7,75]
17.Rhamnopyranosyl-cantleyosideD. asper (roots) [99]
18.LisianthiosideD. asper (roots) [6,89]
19.Loganic acidD. asper (roots) [6,22,65,66,92,93,95,99]
D. fullonum (leaves, roots) [33,34]
S. atropurpurea subsp. maritima (flowers) [7,84]
20.Loganic acid ethyl esterD. asper (roots) [97]
21.LoganinD. asper/D. asperoides (roots) [6,22,27,65,66,89,92,93,95,96,97,99]
D. fullonum (leaves, roots) [33,34]
S. atropurpurea subsp. maritima (flowers) [7,84]
22.Secologanin-methyl-hemiacetalS. atropurpurea subsp. maritima (whole plants) [78]
23.SeptemfidosideS. stellata (whole plants) [7,75]
24.SwerosideD. asper/D. asperoides (roots) [6,22,27,65,66,92,93,96,99]
D. fullonum leaves, roots) [33]
S. stellata (whole plants) [7,75]
S. atropurpurea subsp. maritima (flowers) [7,84]
S. tschilliensis (flowers) [94]
25.Sylvestroside ID. asper (roots) [22,92,99]
S. stellata (whole plants) [7,75]
S. tschilliensis (flowers) [94]
26.Sylvestroside IIS. tschilliensis (flowers) [94]
27.Sylvestroside IIID. asper [6]
D. fullonum (leaves, roots) [33,34]
28.Sylvestroside IVD. asper (roots) [6]
D. fullonum (leaves) [34]
29.Triplostoside AD. asper (roots) [6,89,92,93,99]

3.2. Phenolic Acids

Many phenolic acids have been noted in Dipsacus and Scabiosa species (Table 4) [6,24,27,29,33,34,43,46,53,65,66,73,74,75,76,81,89,92,93,94,95,97,99,101]. Mainly derivatives of hydroxycinnamic acid were identified, with chlorogenic acid as the most abundant component. This phenolic acid was detected in Dipsaci radix [6,27,29,65,93,95,97,99], D. fullonum leaves and roots [33,34,101], S. comosa and S. tschiliensis inflorescences [53,74], S. atropurpurea stems [46], and S. atropurpurea subsp. maritima leaves [81]. In addition, other mono-caffeoylquinic acid derivatives (1-O-caffeoylquinic acid, 3-O-caffeoylquinic acid methyl ester, and 4-O-caffeoylquinic acid) and dicaffeoylquinic acid derivatives (3,4-di-O-caffeoylquinic acid, 3,5-di-O-caffeoylquinic acid, 4,5-di-O-caffeoylquinic acid, and their methyl derivative) have been identified (Table 4) [6,24,27,33,43,65,66,74,75,76,81,92,93,95,99]. The hydroxycinnamic acid derivatives included caffeic acid, caffeic acid methyl ester, p-coumaric acid, p-coumaric acid 3-glucoside, 2,6-dihydroxycinnamic acid, 5-O-ferruloylqunic acid, p-hydroxycinnamic acid, and sinapic acid [6,29,46,53,73,74,76,81,89,92,93,94,95,97,99]. The benzoic acid derivatives were 3,4-dihydroxybenzoic acid, protocatechuic acid, protocatechuic acid 3-glucoside, and vanillic acid [6,74,81,89,92] (Table 4).
The phenolic acid content in Dipsaci radix varied according to the geographical region of China [95,99]. The total level of phenolic acids ranged from 0.98 mg/g to 49.55 mg/g, with the highest level observed in plant material collected from Guizhou Province [99]. The predominant phenolic acid was chlorogenic acid (0.186–19.174 mg/g; 2.02–8.28%) [95,99], which was also an abundant phenolic in D. fullonum [33]. However, qualitative differences were reported between the leaves and the roots, with the highest level being found in the leaves obtained from plants in the second year of vegetation (28.44 mg/g d.w.). Wang et al. [53] also found differences in the content of chlorogenic acid in plants of S. tschiliensis between the pre-flowering, flowering, and fruiting stages. The level was also dependent on the extraction solvent; the richest source of chlorogenic acid, 45.35 mg/g d.w., was found in the ethyl acetate fraction from plants at the pre-flowering stage.
Table 4. Phenolic acids identified in Dipsacus and Scabiosa species.
Table 4. Phenolic acids identified in Dipsacus and Scabiosa species.
No.Compound NameSpecies (Part of Plant)
1.1-O-caffeoylquinic acidD. asper (roots) [6]
S. stellata (whole plants) [76]
2.2′-O-caffeoyl-d-glucopyranoside esterD. asper (roots) [89]
3.2,6-dihydroxycinnamic acidD. asper (roots) [6,89]
4.3-O-caffeoylquinic acid methyl esterS. atropurpurea subsp. maritima (leaves) [81]
S. atropurpurea L. (aerial parts) [43]
5.3,4-di-O-caffeoylquinic acid (isochlorogenic acid B) aD. asper (roots) [6,24,66,92,93,95,99]
S. atropurpurea sbsp. maritima (leaves) [81]
S. comosa (inflorescences) [74]
S. stellata (whole plants) [76]
S. tschilliensis (inflorescences) [74]
6.3,4-dihydroxybenzoic acidD. asper (roots) [92]
7.3,5-di-O-caffeoylquinic acid (isochlorogenic A) aD. asper/D. asperoides (roots) [6,24,27,65,66,92,93,95,99]
D. fullonum (leaves, roots) [33]
S. atropurpurea subsp. maritima (leaves) [6]
S. comosa (inflorescences) [74]
S. stellata (whole plants) [75,76]
S. tschilliensis (inflorescences) [74]
8.4-O-caffeoylquinic acid (cryptochlorogenic acid) aD. asper (roots) [6,65,66]
D. fullonum (leaves, roots) [33]
S. atropurpurea subsp. maritima (leaves) [81]
S. stellata (whole plants) [76]
9.4,5-di-O-caffeoylquinic acid (isochlorogenic acid C) aD. asper (roots) [6,24,92,93,95,99]
S. comosa (inflorescences) [74]
S. stellata (whole plants) [75,76]
S. tschilliensis (inflorescences) [74]
10.5-O-feruloylquinic acidS. stellata (whole plants) [76]
11.5-O-p-coumaroylquinic acidS. stellata (whole plants) [76]
12.Caffeic acidD. asper/D. asperoides (roots) [6,29,89,92,93,95,97,99]
S. comosa (inflorescences) [74]
S. tschilliensis (inflorescences) [53,74]
S. atropurpurea (stems) [46]
13.Caffeic acid methyl esterS. tschilliensis (flowers) [94]
14.Chlorogenic acidD. asper/D. asperoides (roots) [6,27,29,65,93,95,97,99]
D. fullonum (leaves, roots) [33,34,101]
S. atropurpurea subsp. maritima (leaves) [81]
S. atropurpurea (stems) [46]
S. comosa (inflorescences) [74]
S. tschilliensis
(inflorescences) [53,74]
15.Methyl 3,4-di-O-caffeoylquinateD. asper (roots) [6,24]
16.Methyl 3,5-di-O-caffeoylquinateD. asper (roots) [6,24]
17.Methyl 4,5-di-O-caffeoylquinateD. asper (roots) [6,24]
18.Neochlorogenic acidD. fullonum (leaves, roots) [33]
S. atropurpurea subsp. maritima (leaves) [81]
19.p-coumaric acidS. comosa (inflorescences) [74]
S. tschilliensis (inflorescences) [74]
S. atropurpurea (stems) [46]
S. arenaria (roots) [73]
20.p-coumaric acid 3-glucosideS. atropurpurea. subsp. maritima (leaves) [81]
21.p-coumaroylquinic acidS. atropurpurea subsp. maritima (leaves) [81]
22.p-hydroxycinnamic acidS. atropurpurea (stems) [46]
23.Protocatechuic acidS. comosa (inflorescences) [74]
S. tschilliensis (inflorescences) [74]
24.Protocatechuic acid 3-glucosideS. atropurpurea subsp. maritima (leaves) [81]
25.Quinic acidS. comosa inflorescences) [74]
S. tschilliensis inflorescences [74]
S. atropurpurea subsp. maritima (leaves) [81]
26.Sinapic acidS. stellata (whole plants) [76]
27.Vanillic acidD. asper (roots) [6,89,92]
a synonymous names of compounds are given in accordance with PubChem database [109].

3.3. Flavonoids

The Scabiosa genus is a rich source of flavonoids, which are identified mainly in the flowers but also in the leaves, stems, and roots (Table 5) [7,43,46,48,53,73,74,75,76,81,82,94]. Many studies note that apigenin, luteolin, and their derivatives are particularly common [7,43,46,74,75,76,81,82,94]. The new flavonoids identified for the first time in Scabiosa spp. were diosmetin-7-O-glucoside, luteolin-7,3′-diglucoside, luteolin 3′-glucoside, and quercetin 3,4′-diglucoside in S. atropurpurea subsp. maritima leaves [81]; quercimeritrin in S. atropurpurea stems; quercitrin, rutin, kaempferol-3-O-β-d-6-O-(p-hydroxycinnamoyl)-glucopyranoside, and kaempferol-3-O-β-d-(3,6-di-p-(hydroxycinnamoyl)-glucopyranoside in S. tschilliensis flowers [46,94]; and tamarixetin derivative 3-β-l-rhamnosyl-(1→2)[β-l-rhamnosyl-(1→6)]β-d-glucoside] and tiliroside in S. stellata whole plants [76]. Isoorientin, isovitexin, orientin, saponarin, and saponaretin were detected for the first time in Dipsacus spp., in D. fullonum leaves [33,34].
Table 5. Flavonoids identified in Dipsacus and Scabiosa species.
Table 5. Flavonoids identified in Dipsacus and Scabiosa species.
No.Compound NameSpecies (Part of Plant)
1.3-O-[3-O-acetyl-6-O-(p-coumaroyl)-β-d-glucopyranosyl]-kaempferolS. stellata (whole plants) [48]
2.ApigeninS. tschilliensis (flowers) [94]
S. comosa (inflorescences) [74]
S. tschilliensis (inflorescences) [74]
3.Apigenin-2″-O-pentosyl-8-C-glucosideS. stellata (whole plants) [76]
4.Apigenin-4′-glucoside (apigenin-4′-O-β-d-glucopyranoside) aS. comosa (inflorescences) [74]
S. tschilliensis (inflorescences/flowers) [74,94]
5.Apigenin-7-arabino(1~6)-glucosideS. comosa (inflorescences) [74]
S. tschilliensis (inflorescences) [74]
6.Apigenin-7-glucoside (apigenin-7-O-β-d-glucopyranoside) aS. atropurpurea subsp. maritima (leaves) [81]
S. comosa (inflorescences) [74]
S. tschilliensis (inflorescences/flowers) [74,94]
7. Apigenin-7-O-rutinoside S. tschilliensis (flowers) [94]
8.Diosmetin-6(or 8)-C-glucosideS. stellata (whole plants) [76]
9.Diosmetin-7-O-glucosideS. atropurpurea subsp. maritima (leaves) [81]
10.Hyperin (hyperoside; quercetin 3-O-galactoside) aS. atropurpurea (stems) [46]
S. stellata (whole plants) [7,75,76]
11.IcariinS. tschiliensis (inflorescences) [53]
12.Isoorientin (luteolin-6-C-glucoside) aD. fullonum (leaves) [34]
D. sativus L. (leaves) [42]
S. comosa (inflorescences) [74]
S. stellata (whole plants) [7,75,76]
S. tschilliensis (inflorescences) [74]
13.Isoquercitrin (quercetin 3-glucoside) aS. atropurpurea (stems) [46]
S. comosa (inflorescences) [74]
S. tschilliensis (inflorescences) [74]
14.Isovitexin (saponaretin; apigenin-6-C-glucoside) aD. fullonum (leaves) [33,34]
D. sativus (leaves) [42]
15.Kaempferol-3-O-β-d-(3,6-di-p-(hydroxycinnamoyl)-glucopyranosideS. tschilliensis (flowers) [94]
16.Kaempferol-3-O-β-d-6-O-(p-hydroxycinnamoyl)-glucopyranosideS. tschilliensis (flowers) [94]
17.Kaempferol-3-O-rutinoside derivativeS. stellata (whole plants) [76]
18.Lucenin-2 (luteolin-6,8-di-C-glucoside) aS. stellata (whole plants) [76]
19.LuteolinS. atropurpurea subsp. maritima (leaves) [81]
S. atropurpurea (aerial parts, stems) [7,43,46]
S. comosa (inflorescences) [74]
S. tschilliensis (inflorescences/flowers) [74,94]
20.Luteolin-2″-O-pentosyl-6-C-hexosideS. stellata (whole plants) [76]
21.Luteolin-4′-O-β-d-glucopyranoside (luteolin-4′-glucoside) aS. comosa (inflorescences) [74]
S. tschilliensis (flowers) [74,94]
22.Luteolin 3′-glucosideS. atropurpurea subsp. maritima (leaves) [81]
23.Luteolin hexosideS. atropurpurea (stems) [46]
24.Luteolin-6-C-glucoside-7-O-glucosideS. stellata (whole plants) [76]
25.Luteolin 7-rutinoside (luteolin-7-O-β-d-rutinoside) aS. atropurpurea (aerial parts) [43]
S. atropurpurea subsp. maritima (leaves) [81]
26.Luteolin-7,3′-diglucosideS. atropurpurea subsp. maritima (leaves) [81]
27.Luteolin 7-O-β-d-glucoside (cynaroside; luteolin-7-O-β-d-glucopyranoside) aS. arenaria (aerial parts) [82]
S. atropurpurea (stems) [46]
S. atropurpurea subsp. maritima (leaves/aerial parts) [43,81]
S. tschilliensis (flowers) [81]
28.MyricetinS. arenaria (roots) [73]
29.Orientin (luteolin 8-C-β-d-glucopyranoside, luteolin 8-C-glucoside) aD. fullonum (leaves) [33]
30.Quercetin-3-rutinosideS. comosa (inflorescences) [74]
S. tschilliensis (inflorescences) [74]
31.Quercetin 3,4′-diglucosideS. atropurpurea subsp. maritima (leaves) [81]
32.Quercimeritrin (quercetin 7-glucoside) aS. atropurpurea (stems) [46]
33.Quercitrin (quercitin-3-O-rhamnoside) aS. tschiliensis (inflorescences) [53]
34.Rutin (quercetin 3-rutinoside) aS. tschiliensis (inflorescences) [53]
35.Saponarin
(apigenin-6-C-glucoside-7-O-glucoside) a
D. fullonum (leaves, roots) [33,34]
D. sativus (leaves) [42]
36.Swertiajaponin (isoorientin 7-methyl ether) aS. stellata (whole plants) [7,75]
37.Tamarixetin derivative (3-β-l-rhamnosyl-(12)[β-l-rhamnosyl-(16)]β-d-glucosideS. stellata (whole plants) [7,76]
38.TilirosideS. stellata (whole plants) [7,76]
a synonymous names of compounds are given in accordance with PubChem database [109].

3.4. Lignans

Lignans were identified only in Dipsaci radix and include prinsepiol, fraxiresinol, and their derivatives such as dipsalignans A-D [6,13,99]. Dipsaci radix also demonstrated derivatives of pinoresinol and syringaresinol (Table 6) [13,46,96,97,99]. Syringaresinol hexoside was detected as a new compound in S. atropurpurea stems [46].
Table 6. Lignans identified in Dipsacus and Scabiosa species.
Table 6. Lignans identified in Dipsacus and Scabiosa species.
No.Compound NameSpecies (Part of Plant)
1.(7R, 8S, 7′R, 8′S)-fraxiresinol-4′-O-β-d-glucopyranosideD. asper (roots) [99]
2.(7R, 8S, 7′R, 8′S)-prinsepiol-
4-O-β-d-glucopyranoside
D. asper (roots) [99]
3.(7R, 8S, 7′R, 8′S)-8-hydroxypinoresinol-
4′-O-β-d-glucopyranoside
8′-hydroxypinoresinol-4′-O-β-d-glucopyranoside
D. asper (roots) [96,99]
4.(+)-1-hydroxy-2,6-bis-epi-pinoresinolD. asper (roots) [6]
5.(+)-8-hydroxy-7,7′-bis-epi-pinoresinolD. asper (roots) [13]
6.Dipsalignan A
((+)-8-hydroxy-7,7′-bis-epi-fraxiresinol)
D. asper (roots) [6,13]
7.Dipsalignan B
((+)-(7S, 8S, 7′R, 8′S)-prinsepiol)
D. asper (roots) [6,13]
8.Dipsalignan C
((+)-(7S, 8S, 7′R, 8′S)-5-methoxyprinsepiol)
D. asper (roots) [6,13]
9.Dipsalignan D
((+)-(7S, 8R, 7′S, 8′R)-5-methoxyprinsepiol)
D. asper (roots) [6,13]
10.Syringaresinol-4′,4″-O-bis-β-d-glucosideD. asper (roots) [97]
11.Syringaresinol hexosideS. atropurpurea (stems) [46]

3.5. Polysaccharides

Dipsacus spp. also include polysaccharides [11,63,110,111]. The water-soluble polysaccharide from D. asperoides roots (ADAPW) has a molecular weight of 16 kDa and contains glucose, rhamnose, arabinose, and mannose in a molar ratio of 8.54:1.83:1.04:0.42 [11]. The polysaccharide WDRAP-1, with a molecular weight of 61 kDa, was composed of glucose, mannose, galactose, arabinose, and rhamnose in a molar ratio of 3.1:0.9:5.2:1.1:0.3. The predominant monosaccharides were glucose (29.2%) and galactose (49.1%) [63]. Xu et al. [110] found two polysaccharides in D. asperoides roots, DAI-1 and DAI-2, which consisted only of glucose and had respective molecular masses of 17 and 4 kDa. Sun et al. [111] isolated from D. asper roots a homogenous polysaccharide (DAP) with a molecular weight of 26.1 kDa that was composed of galactose and mannose in a molar ratio of 1:1.

3.6. Essential Oils

Essential oils are present in some species of Dipsacus and Scabiosa (Table 7) [35,83,102].
The essential oils isolated by hydrodistillation from dried and fresh roots and leaves of D. fullonum were rich in many components; however, quantitative and qualitative differences were noted. The dominant compound in the essential oils from leaves, regardless of fresh or dried materials, was phytol (branched-chain unsaturated diterpene alcohol; precursor for vitamins E and K1), whose content ranged from 61.08% to 72.31%. It was not detected in root essential oil. In addition, the main components in fresh leaf essential oil, i.e., with a level above 5%, were 9,12,15-octadecatrienoic acid methyl ester and cyclohexane, cyclopropylidene. On the other hand, in the essential oils from the dry and fresh roots, the main component was n-hexadecanoic acid, especially in the dried material (16.00%), which was also rich in 11,14,17-eicosatrienoic acid, methyl ester (15.86%). n-hexadecanoic acid was also identified in the essential oils from dried and fresh leaves but at much lower levels (2.01–2.34%) [35]. In the essential oil from the flowering aerial parts of D. japonicus, linalool (11.78%), trans-geraniol (8.58%), and 1,8-cineole (7.91%) predominated. The other main ingredients present at above 5% were β-caryophyllene (5.58%), α-terpineol (5.32%), β-selinene (5.15%), and spathulenol (5.04%) [102].
Qualitative and quantitative differences were also observed in the essential oils isolated from different parts of S. arenaria [83]. The main compounds detected in the oil from the aerial parts and flower oils were chrysanthenone (23.43–38.52%), camphor (11.75–12.98%), and α-thujone (9.5–10.7%), while α-thujone (34.39%), camphor (17.48%), and β-thujone (15.29%) predominated in the fruit oil. In addition, longifelone (2.41–3.96%) and filifolone (1.99–3.72%) were only identified in oils from the vegetative parts of the plants and the flowers. In S. atropurpurea stems, in volatile fractions VF1 (extracted by hexane) and VF2 (extracted by chloroform), the most abundant ingredients were 1,8 cineole (8.1–33.8%), tetradecene (5.7–24.1%), and (E)-β-ionone (5.9–20.7%). It is worth mentioning that dihydroactinidiolide, which was present in significant amounts (26.1%), was identified only in the chloroform fraction [46].
Table 7. The main chemical constituents of the essential oils isolated from some species of Dipsacus and Scabiosa.
Table 7. The main chemical constituents of the essential oils isolated from some species of Dipsacus and Scabiosa.
Compounds (Content)Species (Part of Plant)
n-hexadecanoic acid (2.92–16.00%); 11,14,17-eicosatrienoic acid, methyl ester (15.86% only in dried material); 9,12,15-octadecatrienoic acid, methyl ester (0.65–3.2%)D. fullonum (roots; fresh or dried) [35]
Phytol (61.08–72.31%); 9,12,15-octadecatrienoic acid, methyl ester (6.06% only in fresh material); cyclohexane, cyclopropylidene (2.90–5.20%); n-hexadecanoic acid (2.01–2.34%); 3-buten-2-one, 4-(2,6,6-trimethyl-1-cyclohexen-1-yl) (1.5–3.2%)D. fullonum (leaves; fresh or dried) [35]
Linalool (11.78%); trans-geraniol (8.58%); 1,8-cineole (7.91%); β-caryophyllene (5.58%); α-terpineol (5.32%); β-selinene (5.15%); and spathulenol (5.04%); geranyl acetone (3.88%); α-pinene (3.57%)D. japonicus (flowering aerial parts) [102]
1,8 cineole (8.1–33.8%); tetradecene (5.7–24.1%); (E)-β-ionone (5.9–20.7%); dihydroactinidiolide (26.1% in chloroform fraction, not detected in hexane fraction); (Z)-jasmone (5.6% in hexane fraction, not detected in chloroform fraction); eugenol (3.6% in hexane fraction, not detected in chloroform fraction); (E)-β-damascenone (3.0–6.4%); (E)-geranylacetone (3.0–9.2%); linalool (3.3–4.9%); 2-hydroxy-5-methylacetophenone (2.9–4.4%); cis-linalool oxide (1.6–3.0%)S. atropurpurea (stems; volatile fraction extracted by hexane or chloroform) [46]
Chrysanthenone (23.43%), camphor (12.98%) and α-thujone (10.7%); α-fenchol (4.08%); sabinene (3.11%); trans-alloocimene (3.03%);S. arenaria (vegetative parts; stems and leaves) [83]
Chrysanthenone (38.52%), camphor (11.7%) and α-thujone (9.5%); α-fenchol (5.86%); filifolone (3.72%); longifolene (3.96%)S. arenaria (flowers) [83]
α-thujone (34.39%), camphor (17.48%), and β-thujone (15.29%); camphene (3.62%); 1,8-cineole (3.48%); sabinene (3.46%)S. arenaria (fruits) [83]

3.7. Fatty Acids

The presence of fatty acids has only been investigated in S. stellata aerial parts and D. asper roots [6,49] (Table 8). Thirteen fatty acids including saturated and unsaturated acids have been identified. The fatty acids present in the hexane extract of S. stellata aerial parts accounted for 87% of the total content with linolenic acid, palmitic acids, and linoleic acids predominating [49].
Table 8. Fatty acids identified in Dipsacus and Scabiosa species.
Table 8. Fatty acids identified in Dipsacus and Scabiosa species.
No.Compound NameSpecies (Part of Plant)
1.Behenic acidS. stellata (aerial parts) [49]
2.Dodecanoic acidS. stellata (aerial parts) [49]
3.Dotriacontanic acidD. asper [6]
4.Eicosanoic acidS. stellata (aerial parts) [49]
5.Hexadecatrienoic acidS. stellata (aerial parts) [49]
6.Lignoceric acidS. stellata (aerial parts) [49]
7.Linoleic acidS. stellata (aerial parts) [49]
8.Linolenic acidS. stellata (aerial parts) [49]
9.Myristic acidS. stellata (aerial parts) [49]
10.Palmitic acidS. stellata (aerial parts) [49]
11.Pentacosanoic acidD. asper (roots) [6]
12.Stearic acidS. stellata (aerial parts) [49]
S. tschilliensis (flowers) [94]
13.Triacontanoic acidS. stellata (aerial parts) [49]

4. Safety of Use

The use of traditional medicines, herbs, and supplements of plant origin is becoming more popular. However, it is necessary to determine the side effects that they may cause or their toxicity to use safely.
An in vitro study showed that the extract of D. asperoides was not toxic for normal cells at a concentration up to 500 µg/mL [26,112]. The viability of RAW 264.7 macrophages and periodontal ligament stem cells after treatment with water or ethanol extracts for 24 h was about 90–100% [26,112]. However, the periodontal ligament stem cells treated for 7–21 days with 500 µg/mL of D. asper ethanol extract showed morphology changes [112]. The cell viability of J774A.1 murine macrophage was also not affected by the methanol extract of D. inermis at a concentration up to 100 µg/mL [39]. The concentration above 300 µg/mL decreased the cell viability below 80%. In addition, Dipsaci radix stimulated the proliferation of MC3T3-E1 and primary osteoblastic cells in the concentration range of 3–300 µg/mL after 24 h and 48 h [113]. It should be also noted that akebia saponin D, a quality indicator of Dipsaci radix, at a concentration of 25–200 µM has no cytotoxic effect in mouse primary chondrocytes after 24 h [25]. In addition, saponin favored the proliferation of rat bone marrow stomal cells on days 4 and 7 in a dose-dependent manner (0.01–10 µM) [64] and enhanced the proliferation of human mesenchymal stem cells at a concentration up to 1 mg/L after 3 and 5 days [114].
The toxicity of Dipsacus or Scabiosa plants has been tested not only in vitro but also in vivo. According to Zhou et al. [115], the clinical safety of D. asper has been evaluated by Zhan et al. [116]. The total saponin extract (0.28 g/tablet) was administered to volunteers for six months, resulting in only mild side effects such as abdominal discomfort, constipation, swollen gums, raised level of blood alanine aminotransferase (ALT), and dysphoria.
A study using F344 rats which were orally treated with the aqueous extract of Dipsaci radix at doses of 0.125, 0.25, 0.5, 1, or 2 g/kg body weight (b.w.)/day for 13 weeks resulted in no deaths or pathophysiological changes [20]. On the other hand, Xiao et al. [117] found that an extract of D. asper roots may have an adverse effect at a concentration of 2–32 g/kg/day. Dipsaci radix enhanced fetal malformation in a dose-dependent manner in pregnant ICR mice. In addition, the extract at a dosage of 32 g/kg/day, i.e., 17-times higher than recommended for an adult human, was toxic to the fetus; it led to abnormalities of fetal skeletal development including malformed limbs (polydactylia) and sternum (hypoplasia and split) as well as inhibited mineralization cartilaginous tissue and osteogenesis. It is worth emphasizing that the extract also inhibited the mouse embryogenic stem cells and 3T3 cells growth in a dose-dependent manner (0.1–125 mg/mL) with IC50 values of 6.83 mg/mL and 5.13 mg/mL, respectively [117].
The ethyl acetate extract of S. stellata whole plants orally administered for 14 days at a concentration of 0.5–2 g/kg in a single dose was not toxic for albino Wistar rats and did not induce animal mortality [50]. Moreover, no changes in respiration and urination or in hematological and serum biochemical parameters such as ALT, aspartate aminotransferase (AST), total bilirubin, urea, creatinine, cholesterol, triglycerides, and glucose levels were observed in comparison to those in the control animals [50]. Mouffouk et al. [77] estimated the cytotoxicity of the hydroethanolic extract, petroleum ether, ethyl acetate, and n-butanolic fractions of S. stellata whole plants in larvae of brine shrimp lethality method. A dose-dependent pattern (at a concentration of 10–100 µg/mL) in the mortality rate of the brine shrimp nauplii was noted. Only the n-butanolic extract caused mortality above 50% at a concentration of above 80 µg/mL. The 70% ethanol extract of aerial parts of S. atropurpurea possessed an LD50 value for adult male albino rats of 5 g/kg b.w. [43].

5. Pharmacokinetic and Bioavailability of Some Specialized Metabolites of Dipsaci radix

The synergistic effect of numerous, various specialized metabolites of plants is responsible for the medicinal properties of plants. Therefore, knowing the chemical composition of herbal materials is the basis for understanding the mechanisms of their action [118]. It is important to know the pharmacokinetics of drugs to understand the toxicity of their preparations or their therapeutic potential. The therapeutic effectiveness of the preparations/compounds is related to their bioavailability. The bioavailability of compounds depends on many different parameters, such as digestion, absorption, or metabolism [119]. Akebia saponin D, the main ingredient of Dipsaci radix, shows low absorption. Therefore, its therapeutic effect is limited [119]. Wang et al. [119] suggested that the microcrystalline form of akebia saponin D obtained by antisolvent precipitation may enhance the bioavailability of akebia saponin D.
A few specialized metabolites identified in the roots of D. asper, e.g., 4-O-caffeoylquinic acid, 3-O-caffeoylquinic acid, 3,5-di-O-caffeoylquinic acid, loganic acid, loganin, sweroside, dipsacoside B, and asperosaponin VI, showed rapid absorption after intragastric administration to Sprague-Dawley rats at a concentration of 75.6 g/kg; these ingredients reached the maximum plasma concentration in an hour [15]. In addition, it was shown that sauteing with rice wine of D. asper roots enhanced the bioavailability of these specialized metabolites, indicated by a significant increase in maximum plasma concentration and area under the curve for the plasma concentration from zero to the last quantifiable time-point as well as an increase in the level of bioactive compounds in rat liver and kidney tissues compared with those in the crude material (aqueous extract) [15,120].

6. Biological Activities of Dipsacus and Scabiosa Species

6.1. Strengthening the Bone Tissue and Antiarthritic Activity

The roots of D. asperoides, known in Chinese as Xu Duan, means “connects broken bones” [26]. Until now, numerous in vitro and in vivo studies have demonstrated that Dipsaci radix or the pure compounds isolated from this plant material can be potential agents for promoting osteoblast formation and may have an anabolic systemic skeletal effect. Dipsaci radix can improve bone density and affect bone histomorphology. Dipsaci radix has also shown osteoprotective properties in ovariectomized animals [6,18,23,25,68,110,111,112,113,115,121,122,123,124,125,126,127,128]. In healthy BALB/c mice, an increase in the bone trabeculae density, bone volume/tissue volume ratio, bone surface/tissue volume, and trabecular number and the depletion in the trabecular separation on the proximal tibia after drinking water extract of Dipsaci radix were observed [61].
Osteoporosis is characterized by an increase in bone fragility. Osteoporosis can usually occur with aging and after menopause due to estrogen deficiency, which contributes to the reduction of bone density and bone mass and degradation of the microstructure [129]. The hormone replacement therapy that is used in osteoporosis treatment increases bone density. However, the use of hormone replacement therapy for a long time should be limited due to the serious side effects of its use [126]. Modern therapies enhance bone metabolism by promoting osteoblast activity and by inhibiting the effects of osteoclasts [126]. The ethanol extract of D. asperoides roots showed concentration-dependent progestogenic activity (40–100 μg/mL) in the T47D progesterone receptor-positive human mammary adenocarcinoma cell line; 100 μg/mL of extract demonstrated the equivalent of 31.45 ng/mL of progesterone treatment. These results indicate that D. asperoides roots can be used as an option for progestins [130]. Moreover, asperosaponin D may promote the osteogenic differentiation of human mesenchymal stem cells through the estrogen signaling pathway [131].
Liu et al. [18,122] reported that Dipsaci radix decoction has anti-osteoporosis properties in ovariectomized Wistar or Sprague-Dawley rats (oral treatment at a dose of 100–500 mg/kg b.w./day) by increasing trabecular bone formation and bone mineral density and preventing bone mass loss and trabecular structure changes; it also decreases the serum alkaline phosphatase (ALP) level and the level of bone turnover markers, e.g., serum osteocalcin and urinary deoxypyridinoline/creatinine ratio, and receptor activator for nuclear factor κB ligand (RANKL) in osteoblasts and bone marrow stromal cells of the tibia. A recent study showed that Dipsaci radix may be able to control osteoblast differentiation, osteoclast proliferation, and mineralization via regulating mitogen-activated protein kinases (MAPK), nuclear-kappa B factor (NF-κb), TNF-α, and Toll-like receptor (TLR4) signaling pathways [127]. Intragastric treatment of bilaterally ovariectomized Wistar rats with wine processed Dipsaci radix (with a dose of 75.6 g/kg/day) resulted in protection from an increase in urine Ca/creatinine and P/creatinine levels and serum ALP and osteocalcin concentrations and increased the femur bone mineral density. These effects were comparable to that observed in rats treated with 17β-estradiol [68].
In traditional Chinese medicine, Xian-Ling-Gu-Bao capsules have been used to prevent and treat osteoporosis, osteoarthritis, aseptic bone necrosis, or climacteric syndrome. Xian-Ling-Gu-Bao was officially approved in 2002 by the China Food and Drug Administration as an over-the-counter drug for the treatment of osteoporosis [132,133]. This product is composed of the raw material of six plant species: Epimedii herba (70%), Dipsaci asperoidis radix (10%), Anemarrhenae rhizoma (5%), Psoraleae fructus (5%), Rehmanniae radix (5%), and Salviae miltiorrhizae radix (5%) [133]. Xian-Ling-Gu-Bao administered to ovariectomized C57/BL6 mice for six weeks displayed anti-osteoporosis effects by enhancing bone mineral density and bone strength and by decreasing the serum level of the bone formation marker procollagen type I N-terminal propeptide (PINP) and the bone resorption marker C-terminal telopeptide of type I collagen (CTX) [125]. Wu et al. [132] also reported that Xian-Ling-Gu-Bao showed the ability to prevent osteoporosis in two osteoporosis models, prednisolone-treated zebrafish (Danio rerio) and ovariectomized Sprague-Dawley rats. Xian-Ling-Gu-Bao altered the protein levels of osteoprotegerin and RANKL. An increase in the OPG/RANKL ratio may suppress bone loss. It is worth mentioning that a dose of 1800 mg/kg (a concentration of six-times the recommended daily dose) did not cause toxicity or adverse effects in the heart, kidney, liver, stomach, or small intestine [132].
Another formulation containing Dipsaci radix used in traditional Chinese medicine for treating kidney disease, osteoporosis, strengthening bones, and bone fractures is Du-Zhong-Wan. This preparation is composed of Eucommiae cortex (salted bark of Eucommia ulmoides Oliv.) and Dipsaci radix in an equal weight ratio (1:1) [23]. It was shown that Du-Zhong-Wan displayed anti-osteoporotic activity in the Sprague-Dawley rat osteopenia model. After treatment with Du-Zhong-Wan at a dose of 2–6 g/kg/day for 12 weeks, an increase in the bone mineral density of the femur was observed, together with an improvement of trabecular bone mass and microarchitecture, reduction of the bone resorption and tartrate-resistant acid phosphatase 5b (TRACP-5b) level, and higher serum level of osteocalein, and serum and endometrium level of estrogen [23]. Tian et al. [134] reported that Du-Zhong-Wan favored fractured callus formation and improved osteoblastogenesis and angiogenesis by increasing the level of the H-type vessel endothelium markers (CD31 and endomucin) and proangiogenic factor SLIT3 in C57BL/6 mice after ovariectomy with the open transverse femoral fracture.
Niu et al. [113,123] showed that the 60% ethanolic extract and total saponins of Dipsaci radix at a concentration of 50–500 mg/kg/day may have anti-osteoporotic properties in Sprague-Dawley female rats after bilateral ovariectomy or hindlimb unloading Sprague-Dawley rat model by preventing a decrease in bone mass and by improving bone mineral density, biomechanical strength, and trabecular bone architecture. In addition, Dipsaci radix suppressed osteoclastogenesis through a reduction of the serum level of bone turnover marker (osteocalcin) and urine concentration of phosphorus, calcium, and deoxypyridinoline/creatinine ratio.
In vitro studies demonstrated that Dipsaci radix stimulated osteoblastic proliferation, maturation, and differentiation via bone morphogenetic protein-2 (BMP-2)/MAPK/Smad1/5/8-dependent Runt-related transcription factor 2 (Runx2) signaling pathway. Dipsaci radix inhibited osteoclastogenesis by an increase in OPG/RANKL ratio in MC3T3-E1 murine preosteoblasts and primary osteoblastic cells [113,124]. The ethanol extract of D. asper enhanced osteogenic differentiation of periodontal ligament stem cells through activation of the vascular endothelial growth factor (VEGF)/PI3K/Akt pathway. In addition, periodontal ligament stem cells displayed greater mineralization and mRNA expression of osteogenesis-related genes such as Col-1, ALP, Runx2, and osteocalcin [112].
Treatment with the triterpenoid akebia saponin D also inhibited the osteoclastic gene RANKL in bone marrow mesenchymal stem cells, suppressing osteoclastogenesis and promoting osteogenesis. In addition, the combination of akebia saponin D and BMP-2 immobilized in 2-N, 6-O-sulfated chitosan alleviated osteoclastic formation, enhanced osteogenesis, and promoted angiogenesis by stimulating SMADs, TGF-β1, VEGFA, and OPG/RANKL signaling pathways [135]. Moreover, loganic acid enhanced osteoblastic differentiation in preosteoblast MC3T3-E1 cells and suppressed osteoclast differentiation of primary-cultured monocytes derived from mouse bone marrow [136].
Akebia saponin D was also found to have a potential application in osteoarthritis therapy. Gu et al. [25] demonstrated that it showed anti-inflammatory activity in mouse primary chondrocytes and alleviated osteoarthritis following the surgical destabilization of a medial meniscus model of osteoarthritis in C57BL/6 male wild-type mice. Akebia saponin D was able to inhibit the production of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), nitric oxide (NO), prostaglandin E2 (PGE2), IL-6, TNF-α, and NF-κB; it also activated the nuclear factor erythroid-2-related factor 2 (Nrf2)/Heme oxygenase 1 (HO-1) pathway and suppressed the expression of matrix synthesis degradation-related proteins such as disintegrin, metalloproteinase with thrombospondin motifs 5 (ADAMTS-5), as well as MMP13 in IL-1β treated chondrocytes. It was also found to enhance the expression of Aggrecan and Collagen II [25]. A recent study found it to also enhance the proliferation and differentiation of human mesenchymal stem cells into nucleus pulposus-like cells through p-ERK1/2 and p-Smad2/3 activation, which may prevent intervertebral disc degeneration [114]. Akebia saponin D also favored the proliferation and osteogenic differentiation of rat bone marrow stromal cells through the phosphatidylinositol-3 kinase/AKT signaling pathway, thus inhibiting osteoporosis. This active compound elevated osteogenic differentiation markers such as ALP activity and calcium deposit formation and mRNA level of osteogenic-related genes (ALP, osteocalcin, type 1 collagen (COL 1), and RUNX2) [64].
Another terpenoid compound, hederagenin, also alleviated the progression of osteoarthritis and inhibited inflammation and cartilage degradation. In an in vitro study, hederagenin exerted chondroprotective and anti-inflammatory effects by suppressing the JAK2/STAT3/MAPK pathway, inhibited extracellular matrix degradation, elevated Aggrecan and Collagen II levels, and reduced levels of MMPs and ADAMTS5. In addition, this pentacyclic triterpenoid saponin inhibited cartilage destruction in rats induced by monosodium iodoacetate [137]. Hederagenin 3-O-(2-O-acetyl)-α-l-arabinopyranoside and the dichloromethane fraction of Dipsaci radix also improved osteoblastic differentiation of human alveolar bone marrow-derived mesenchymal stem cells. This specialized metabolite caused the formation of calcified nodules and enhanced the level of bone differentiation protein expression such as sialoprotein and osteocalcin similar to dexamethasone [121].
Moreover, the main iridoid glycoside isolated from Dipsaci radix, sweroside (at a concentration of 1 µM), enhanced rat osteoblast-like UMR 106 cell proliferation, while loganic acid, loganin, and sweroside favored mineralization. Loganin and sweroside suppressed the formation of adipocytes in 3T3-L1 cells [125]. The anti-osteoporotic property of sweroside was also confirmed by Wu et al. [128]. This compound enhanced mineralization of MC3T3-E1 cells by increasing the protein expression of the membrane estrogen receptor-α and G protein-coupled receptor 30 (GPR30), which activate the p38 signaling pathway.
MC3T3-E1 mouse embryonic osteoblast proliferation and differentiation were also promoted by various concentrations (25, 50, or 100 μg/mL) of a 17 kDa polysaccharide (DAI-1) isolated from D. asperoides in high glucose concentrations; this appeared to act via the stimulation of the bone morphogenetic protein 2 (BMP-2)/Smad/runt-related transcription factor 2 (Runx2)/Osterix signaling pathway. The polysaccharide also enhanced osteocalcin level and the mRNA and protein levels of BMP-2 and Runx2 [110]. A homogenous polysaccharide with a molecular weight of 26.1 kDa isolated from the roots of D. asper (at a dose of 50 or 200 mg/kg b.w.) was also able to enhance mRNA and protein levels of VEGF and osteoprotegin, suppress mRNA and protein expression levels of RANKL, and activate the PI3K/Akt/eNOS signaling pathway in ovariectomized rats [111]. The water-soluble polysaccharide (ADAPW), with an average molecular weight of 16 kDa, inhibited the viability of the human osteosarcoma HOS cells by induction of apoptosis cells and inhibition of the PI3K/Akt signaling pathway [11].
An increase in bone resorption may also lead to osteo- or rheumatoid arthritis. Rheumatoid arthritis is a chronic autoimmune and inflammatory disease of the connective tissue [17]. Dipsaci radix has been also applied for many years in traditional Chinese medicine to treat other bone diseases such as rheumatic arthritis [6]. The aqueous extract of D. asperoides roots at a concentration of 50 mg/kg and 100 mg/kg, administered orally once a day for 21 days, displayed antiarthritic effects in collagen-induced rheumatoid arthritis in male DBA/1 mice by enhancement of the ankle joint architecture and suppression of arthritis score (synovitis, pannus, and bone erosion scores) and serum levels of anti-CII IgG2a antibody and the inflammatory mediators (TNF-α, IL-1β, and IL-6). These effects were comparable or stronger to those after treatment with 1 mg/kg of the anti-rheumatoid drug indomethacin [138].
Akebia saponin D at a concentration of 10 or 20 mg/kg/day also showed anti-osteoclastogenic activity in the arthritic joints of male BALB/c and DBA/1 with collagen-induced arthritis. The saponin reduced mRNA expression of osteoclastogenesis markers such as TRAP, CtsK, MMP-9, and β3-integrin. In addition, akebia saponin D also inhibited phosphorylation of Akt, p38, and JNK and mRNA and protein levels of osteoclastogenesis markers in RANKL-induced osteoclastogenesis bone marrow-derived monocytes [17]. Dipsaus saponins also inhibited chondrocyte apoptosis in a rat model of osteoarthritis in a dose-dependent pattern by decreasing expression of Bax, caspase-3, and caspase-9 and by increasing expression of Bcl-2 [139]. Cantleyoside, an iridoid identified in D. asper roots, inhibited proliferation of human rheumatoid arthritis fibroblast synovial cells (HFLS-RA) and induced cell apoptosis through AMPK/Sirt 1/NF-κB pathway activation [140]. Moreover, the protective effect of sweroside was observed in IL-1β-induced inflammation in rat articular chondrocytes. The anti-inflammatory effect of this iridoid was mediated by the inhibition of NF-κB and mTORC1 signaling pathways [141].
These above findings suggest that Dipsaci radix may have a beneficial therapeutic effect in the treatment of postmenopausal osteoporosis and may protect against arthritis.

6.2. Anti-Neurodegenerative Activity

Alzheimer’s disease is a neurodegenerative disease that destroys memory and deteriorates cognitive function [142].
Five-month administration of 4 g/kg ethanol extract of D. asper roots improved neurocognitive dysfunction in the passive avoidance task and diminished expression of hippocampal β-amyloid protein (Aβ) positive cells in aluminum chloride-treated male Sprague-Dawley rats. This effect increased with the time of treatment (1–5 months). It may be important in the treatment of Alzheimer’s disease and memory system dysfunction [31]. Akebia saponin D, at a dose of 30–270 mg/kg administered for four weeks, also showed a preventive effect against memory cognitive impairment in ibotenic acid-exposed male Sprague-Dawley rats [143]. Moreover, it was shown that the saponin can protect against learning and memory dysfunction in rats induced by bilateral intracerebroventricular injections of Aβ1–42 in the Y-maze and Morris water-maze tests. Akebia saponin D attenuated the activation of microglia and astroglia and protein expression level of IL-1, COX-2, TNF-α, NF-κB, and Akt phosphorylation in the rat brain [143]. The total saponin of D. asperoides also possessed neuroprotective properties in rat cortical and hippocampal neurons against damage induced with β-amyloid protein, ameliorated cell viability, and decreased lactate dehydrogenase (LDH) release and lipid peroxidation at a concentration of 150–300 mg/L [144]. Other triterpene compounds, such as oleanolic acid, ursolic acid, and hederagenin, also showed neuroprotective activity in multiple brain disorders [145,146]. For example, hederagenin diminished Aβ deposition in the head area of Caenorhabditis elegans, ameliorated cognitive impairment or pathological changes in APP/PS1 mice, and induced PPARα/TFEB-dependent autophagy of BV2 cells [145]. Moreover, treatment with sweroside, a secoiridoid glycoside, alleviated memory deficits in scopolamine-induced Zebrafish (Danio rerio) in behavioral tests such as the tank diving test, the Y-maze, and the novel object recognition test [147]. The flavonoid apigenin also suppressed neurotoxicity and cognitive function in LPS-induced mice. The compound protected against neuronal degenerative changes in mice hippocampi [148].
It was also found that 50% methanol extracts from leaves and roots of D. fullonum demonstrated anti-acetylcholinesterase activity [33]. The ethyl acetate and n-butanolic fractions of 70% methanol extract from S. stellata whole plants, at a concentration of 200 μg/mL, exhibited moderate (30.8%) and low (10.9%) acetylcholinesterase (AChE) inhibitory activity, respectively [77]. On the other hand, the ethyl acetate and n-butanolic fractions of stems and leaves from S. arenaria (at a dose of 1 mg/mL) were able to inhibit AChE in 97.61% and 90.47%, respectively. The value of IC50 ranged from 0.016 mg/mL to 0.029 mg/mL. In comparison to eserine (positive control), the IC50 value was 0.0029 µg/mL [72]. The root extract also displayed potent activity. The strongest effect was demonstrated by the n-butanolic fraction (87.61% AChE inhibition and IC50 value = 0.02 mg/mL) [73]. Yu et al. [96] showed the effectiveness of some compounds isolated from the roots of D. asper; for example, oleanane triterpenoid saponin (3-O-β-d-glucopyranosyl-(1→3)-α-l-rhamnopyranosyl-(1→2)-α-l-arabinopyranosyl-23-hydroxyolean-18-en-28-oic acid 28-O-β-d-glucopyranosyl-(1→6)-β-d-glucopyranosyl ester) was found to inhibit acetylcholinesterase with an IC50 value of 15.8 μM. Other terpenoid compounds isolated from Dipsaci radix, such as dipsacus saponin IV, dipsacus saponin XI, and dipsacus saponin X, displayed strong AChE inhibitory activity, while cauloside A, dipsacus saponin C, and dipsacus saponin XI were more effective against butyrylcholinesterase. These activities were higher than that of the positive control, berberine. Moreover, the saponins were found to be more effective than the iridoids, loganic acid and sweroside. The terpenoids also inhibited β-site amyloid precursor protein cleaving enzyme 1 (BACE1) and advanced glycation end-product (AGE) formation [149].

6.3. Hepatoprotective Activity

Liver fibrosis is a chronic liver disease caused by many agents such as hepatitis B virus (HBV), hepatitis C virus (HCV), non-alcoholic steatohepatitis, or alcoholic fatty liver disease [150].
The flavonoids and phenolic acids from Scabiosa spp. have demonstrated anti-hepatic fibrosis potential in male Wistar or male Sprague-Dawley rats treated intraperitoneally with carbon tetrachloride (CCl4) (a selective hepatotoxic drug); similar effects were also noted for drugs used in Mongolian medicine, such as Qingganjiuwei and Gurigumu-7 (composed of various herbs including S. comosa flowers) [43,47,86,87].
Qingganjiuwei, a drug commonly used in Inner Mongolia in patients with chronic hepatic disease, improved liver morphology and structure in CCl4-treated SD rats when administered at 1.575–4.725 g/kg/day for eight weeks. The drug reduced hepatocyte necrosis, lymphocytic infiltration, and pseudolobuli and lowered COL1, tissue inhibitor of metalloproteinase1 (TIMP1), and α-smooth muscle actin (α-SMA) expression. The drug also activated the MAPK pathway in the liver through the suppression of extracellular signal-regulated kinase (ERK), C-Jun amino-terminal kinases (JNKs), and stress-activated protein kinase-2 (p38 proteins) [86]. Qingganjiuwei also increased mRNA and protein expressions of MMP2 and MMP9 and inhibited the levels of the serum aminotransferases (ALT and AST) [86].
The methanol-eluted fraction of Gurigumu-7 extract (0.264 g/kg) displayed a more potent hepatoprotective effect in mice with CCl4-induced liver damage compared to crude Gurigumu-7 extract, even when applied at a four-times higher concentration (1.084 mg/kg). The methanol fraction alleviated histopathological changes in the liver and serum ALT, ASP, and liver malonyldialdehyde (MDA) levels and enhanced the liver superoxide dismutase (SOD) level in a dose-dependent manner (66, 132, and 264 mg/kg) [87].
Anti-hepatic fibrosis was also reported for S. comosa and S. tschilliensis [79,80]. Some specialized metabolites inhibited the viability of hepatic stellate LX-2 cells at concentrations of 12.5–200 μM; this included those belonging to flavonoids, which accounted for about 60% of the total identified compounds in the inflorescences of Scabiosa plants. The flavonoids enhanced the expression of Stat1, Pparg, Hsp90aa1 genes, signal transduction and transcriptional activator 1 (STAT1), and peroxisome proliferator-activated receptor G (PPARG) proteins, which play key roles in the pathogenesis of liver fibrosis [79]. Apigenin exhibited the strongest ability to inhibit cell proliferation [79]. The flavonoid-rich extract of S. comosa inflorescences at concentrations of 100 and 200 mg/kg also suppressed hepatic fibrosis in Wistar rats pre-treated with CCl4; the extract inhibited the level of biochemical parameters in blood serum (ALT, AST, ALP, and hyaluronic acid), the markers of liver fibrosis (laminin, amino-terminal propeptide of type III procollagen (PIIINP), collagen IV, collagen deposition in the liver tissues), and expression of α-SMA, collagen I, and fibronectin [47]. Moreover, the extract attenuated phosphorylation of Smad3 in liver tissue and TGF-β1-pre-treatment primary mouse hepatic stellate cells. In the latter, it was observed that the expression of the fibrotic genes (α-SMA, collagen I, and fibronectin) was suppressed in a dose-dependent manner [47]. Ethanol extract of aerial parts of S. atropurpurea and hexane, ethyl acetate, n-butanolic, and chloroform fractions were able to decrease the levels of serum ALT, AST, and ALP in CCl4-induced liver damage in albino rats after treatment with a dose of 100 mg/kg [43].
Sweroside demonstrated a protective effect against liver fibrosis in mouse models treated with CCl4 and methionine-choline-deficient diet-induced non-alcoholic steatohepatitis [151,152]. Sweroside treatment yielded an anti-fibrotic effect through the FXR-miR29a pathway [151]. Sweroside also improved NASH symptoms by inhibiting the activation of the hepatic NLRP3 inflammasome [152]. Apigenin was found to inhibit palmitic acid-induced pyroptosis by regulating the pyrin domain containing 3 (NLRP3) inflammasome in HepG2 cells and primary mouse hepatic cells [153]. Other polyphenols, derivatives of caffeoylquinic acid such as 1,5-di-O-caffeoylquinic acid, isochlorogenic acid C, isochlorogenic acid B, chlorogenic acid, isochlorogenic acid A, neochlorogenic acid, and caffeic acid also suppressed LX-2 hepatic stellate cell growth [79]. The two predominant caffeoylquinic acid derivatives in inflorescences of S. comosa and S. tschilliensis, viz., 3,5-di-O-caffeoylquinic acid and chlorogenic acid, demonstrated anti-hepatitis C virus (HCV) activity in the Huh-7.5 cell line infected with HCV [74].
In addition, some triterpenoid derivatives isolated from the whole plants of S. tschiliensis, such as scabiosaponins E, F, G, I, and J; hookerosides A and B; and prosapogenin 1b, showed strong inhibition of pancreatic lipase. Moreover, 0.12 mg/mL prosapogenin 1b exerted similar inhibitory properties as the lipase inhibitor orlistat at a concentration of 0.005 mg/mL [51]. Akebia saponin D reduced lipid droplet accumulation in BRL cells, attenuated hepatic steatosis, and elevated the expression of Bcl-2/adenovirus E1B 19-kDa interacting protein 3 (BNip3) and phospho-AMPP; it also improved mitochondrial function and autophagy modulation, inhibited rotenone-induced BRL cell apoptosis, elevated Bcl-2/Bax ratio, and suppressed the level of intracellular reactive oxygen species (ROS) and mitochondrial membrane potential loss in rotenone-treated BRL cells and rat liver mitochondria [154,155].

6.4. Cardioprotective Activity

Cardiovascular diseases are one of the leading causes of death worldwide [156].
The anti-atherosclerotic effect of akebia saponin D was studied in vitro in H2O2-treated human umbilical vein endothelial cells (HUVECs) and in vivo in ApoE−/− mice [157]. It was shown that this saponin protected against H2O2-induced cytotoxicity in HUVEC, inhibited ROS level, the mitochondrial membrane potential disruption, and apoptosis in oxidative stress-induced endothelial cells in a dose-dependent manner (50–200 µM); the mechanism was believed to involve increasing Bcl-2 family protein levels and decreasing caspase-3 and Bax activation. Moreover, doses of 150 mg/kg/day and 450 mg/kg/day reduced aortic plaque formation in mice as well as aortic and liver apoptosis, serum triglyceride (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), and lipid deposition in the liver, as well as atherosclerotic lesion size; it also enhanced the expression of antioxidant enzymes (SOD, catalase (CAT), and glutathione (GSH)) in vascular tissue and liver [157]. Li et al. [158] demonstrated that sweroside has a protective effect on ischemia-reperfusion-induced myocardial injury by inhibiting oxidative stress and pyroptosis partially via modulation of the Kelch-like ECH-associated protein 1 (Keap1)/Nrf2 pathway. The iridoid also suppressed aconitine-induced cardiac toxicity in the H9c2 cardiomyoblast cell line [159]. Long-term gavage (for six weeks) of akebia saponin D protected against fibrosis myocardial ischemia injury, inhibited cardiac dysfunction, and reduced infarct size in a Sprague-Dawley rat model with chronic myocardial infarction induced by permanent ligation of the left coronary artery. Treatment with this saponin decreased hydroxyproline level and changed the activity of the oxidative stress enzymes by elevating SOD and DSH-peroxidase (GSH-Px) levels and by reducing MDA content. Moreover, akebia saponin D regulated inflammatory mediators by diminishing the levels of TNF-a and IL-6 and by elevating the level of IL-10 [160]. Several in vitro and in vivo studies found flavonoids detected in Scabiosa spp., such as apigenin and luteolin, to have cardioprotective properties [161,162,163].
However, Song et al. [164] demonstrated that D. asper roots, drunk widely as a tea for beneficial health effects, or dipsacus saponin D may have an undesirable effect on platelets and may increase the risk of thrombosis. D. asper roots were found to be an herb with procoagulant activities on platelets and prothrombotic properties. Dipsacus saponin C elevated procoagulant activity in a dose- and time-dependent manner, elevated intracellular calcium level, and decreased ATP. In addition, it caused translocation of Bax and Bak, cytochrome c release, caspase-3 activation, and the disruption of mitochondrial membrane potential. The oral administration of 10 mg/kg and 25 mg/kg dipsacus saponin C also resulted in an increase in thrombus formation in a rat venous thrombosis model [164].

6.5. Renal and Gastritis Protection

A 61 kDa polysaccharide (WDRAP-1) isolated from D. asperoides roots showed protective activity against oxidative stress generated in renal ischemia-reperfusion injury in male Wistar rats and displayed strong superoxide and hydroxyl radical scavenging activities in vitro. It was shown that oral pre-treatment of rats with the polysaccharide at a concentration of 50–200 mg/kg b.w. for 14 days before ischemia-reperfusion may improve renal injury (especially at the highest dose); treatment suppressed the level of renal injury indicators including creatinine, blood urea nitrogen, lactate dehydrogenase (LDH), and serum MDA and enhanced serum SOD and some renal tissue antioxidant enzyme activities (SOD, GSH-Px, and CAT) [63]. Hederagenin was also found to protect against renal fibrosis. This terpenoid compound attenuated the proliferation and fibrosis of TGF-β-treated NRK-49 F cells by targeting the muscarinic acetylcholine receptor [165].
Dipsacus saponin C, a saponin isolated from the roots of D. asper, was found to have a protective effect against HCl·ethanol-induced gastritis and indomethacin-induced gastric ulcers in male Sprague-Dawley rats. It was shown that treatment with dipsacus saponin C caused a decrease in gastric secretion volume and gastric acid production in pylorus-ligated rats. It was found that this compound had a moderate effect on colonization and growth inhibition of Helicobacter pylori at a concentration of 50–100 µM. In addition, dipsacus saponin C also showed a cytotoxic effect for SNU638 and AGS human gastric cancer cells with an IC50 at 54.6 mM and 37.3 mM, respectively [91].
Moreover, akebia saponin D may exert a therapeutic role by regulating the intestinal microbiome and protecting intestinal epithelial cells from external damage. It was found to achieve this by inhibiting oxidative damage to the intestinal barrier by downregulating PPAR-γ/FABP4 in the human intestinal cell line FHs74 Int [166].
Luteolin was also found to have therapeutic effects against interstitial fibrosis-induced renal anemia in vitro and in vivo. This activity was mediated via the SIRT1/forkhead box O3 (FOXO3) pathway [167].

6.6. Anti-Asthmatic Effect

Dipsaci radix alleviated the asthmatic response in BALB/c mice with allergic asthma induced by an ovalbumin. Dipsaci radix treatment at a dose of 20 m/kg or 40 mg/kg resulted in attenuation of the methacholine response, inflammatory cell infiltration, and mucus secretion in the bronchial airway and a decrease in the levels of pro-inflammatory cytokines (IL-5 and IL-13) in bronchoalveolar lavage fluid, eotaxin, serum total IgE, expression of iNOS, and NF-κB phosphorylation in lungs [27]. Similarly, apigenin, one of the flavonoids identified in Scabiosa spp., was found to inhibit inflammatory mediators and eosinophilia in lung and airway tissues in in vivo acute lung injury and asthma models [163].

6.7. Anti-Diabetic Activity

A polysaccharide (DAP) isolated from D. asper roots demonstrated beneficial effects on renal function and renal pathological changes as well as antihyperglycemic, hypolipidemic, and antioxidant activities in streptozotocin-induced diabetic Wistar rats [16]. Four weeks of intragastric administration (100 mg/kg and 300 mg/kg per day) in type 2 diabetic rat model resulted in a reduction of glycosylated, fasting blood glucose, serum creatinine, blood urea nitrogen, urine protein, and urinary albumin excretion. Moreover, oral administration of the polysaccharide (300 mg/kg) suppressed the serum level of TC, TG, LDL, and renal AGE-RAGE formation (advanced glycation end products-receptor for advanced glycation end products) and enhanced SOD, CAT, and GSH activities in the kidney of rats with diabetic nephropathy [16]. Similar antilipemic effects were also observed for hederagenin. This pentacyclic triterpene exerts its potential through the p38MAPK pathway in oleic acid-induced HepG2 cells and in hyperlipidemic Sprague-Dawley rats [168].
The ethanol extract and the hexane, ethyl acetate, n-butanolic, and chloroform fractions of the aerial parts of S. atropurpurea demonstrated anti-hyperglycemic activity by decreasing the blood glucose level in albino rats with alloxan-induced hyperglycemia [43]. The methanol extract of S. atropurpurea subsp. maritima whole plants demonstrated α-glucosidase inhibitory activity with an IC50 value = 100 μg/mL. This effect was higher than that found for the positive control, the anti-diabetic drug acarbose (IC50 = 196 μg/mL) [78]. Methanolic extracts from the fresh leaves and roots of D. fullonum also inhibited porcine pancreatic α-amylase activity [35]. These extracts demonstrated low effectiveness in this study, with the strongest activity being found to be IC50 = 86.01 µg/mL for the dried leaf extract. This activity was more than 100-times lower compared to that of acarbose (IC50 = 0.69 μg/mL).

6.8. Anti-Inflammatory Activity

Several plant species of Dipsacus and Scabiosa or some specialized metabolites isolated from them can be valuable, new anti-inflammatory agents [26,32,39,50,92,169].
The water extract of D. asper roots demonstrated anti-inflammatory properties in the lipopolysaccharide (LPS)-activated murine macrophage cell line RAW 264.7 by suppressing NO production with an IC50 = 45.1 µg/mL [92]. In a later study [26], an aqueous extract of D. asperoides roots at a dose of 50–500 µg/mL showed inhibitory potential on inflammation and oxidative stress in RAW 264.7 macrophages exposed to LPS; it was found to act by lowering NF-κB and ERK1/2 phosphorylation, nuclear translocation of NF-κB, and activation of Nrf2/HO-1. The extract reduced the levels of inflammatory mediators (iNOS, COX-2, and cytokines IL-6 and IL-1β) as well as ROS levels [26]. The methanol extract of D. inermis leaves also showed the ability to inhibit the production of NO, COX-2, PGE2, pro-inflammatory mediators (IL-1β and IL-6, and TNF-α), intracellular ROS level, and phosphorylation of NF-κBp65 and IκBα in a dose-dependent manner (25–100 µg/mL) in the LPS-induced murine macrophage cell line J774A.1 [39]. The anti-inflammatory potential of D. inermis leaf extract was also confirmed in vivo in Wistar albino rats; it protected against vascular permeability (caused by acetic acid) and paw oedema (induced by carrageenan) in a concentration- and time-dependent manner. In addition, the serum levels of TNF-α, IL-1β, and IL-6 were significantly reduced while IL-10 level was enhanced after oral administration of the extract at a concentration of 50–100 mg/kg b.w. [39]. Anti-inflammatory properties in a Wistar rat model of carrageenan-induced paw oedema also demonstrated the ethyl acetate extract of S. stellata whole plants. The highest activity was observed in the first hour after treatment with the extract at a concentration of 50 mg/kg (72.73% of inhibition). This effect was stronger than that of diclofenac (about 45% of inhibition). In addition, the anti-inflammatory effect of the plant extract lasted up to 24 h [50].
Some compounds belonging to iridoids, saponins, or phenolic acids (for example, dipsasperoside A, dipsanoside A and B, dipsacus saponin A, akebia saponin D, or caffeic acid) isolated from the roots of D. asper also were able to reduce the production of NO in RAW 264.7 cells. The potent activity was demonstrated by akebia saponin D and dipasperoside A, with IC50 values of 12.7 µM and 15.2 µM, respectively; these values were higher than those for the positive control, a nonselective NOS inhibitor, NG-monomethyl-L-arginine (IC50 = 22.6 µM) [92]. Reduced NO levels and iNOS expression have also been observed in LPS-induced RAW 264.7 cells after treatment with akebia saponin D (at a concentration of 25–100 µM) [32]. Akebia saponin D also suppressed the expression of DNA methyltransferase (DNMT) 3b, the levels of PGE2 and p-STAT3, as well as the protein and mRNA levels of IL-6 and TNF-α [169]. The levels of the inflammatory indicators, prostaglandin E2, i-NOS, COX-2, TNF-α, IL-1β, and IL-6, were also decreased after treatment with 40 and 80 µM sweroside in LPS-induced RAW264.7 cells. In addition, sweroside suppressed inflammation through the sirtulin 1 (SIRT1)/NF-κB and SIRT1/Forkhead transcription factor O1 signaling pathways [170].
Akebia saponin D also showed anti-inflammatory activity in vivo by reducing paw oedema in carrageenan-induced Sprague Dawley rats and by inhibiting xylene-induced ear swelling in mice. It also lowered the level of NO in rat plasma in a carrageenan-induced rat paw oedema model [32].
Anti-inflammatory potential was also demonstrated by apigenin, which was believed to act by the modulation of the p38/MAPK, PI3K/Akt and NF-κB pathways [163].

6.9. Antioxidant Activity

The most frequently used methods to determine the antioxidant properties of Dipsacus and Scabiosa plant extracts were the DPPH (2,2-diphenyl-1-picrylhydrazyl radical scavenging assay), ABTS (2,2′-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) radical cation assay), ORAC (oxygen radical absorbance capacity), FRAP (ferric ion reducing antioxidant assay), and CUPRAC assays (cupric-reducing antioxidant capacity). The antioxidant activity varied depending on the plant material (whole plants, leaves, and roots), the solvent, extraction time, and the extraction method (Table 9) [14,24,33,34,42,46,50,52,53,56,72,73,74,76,77,171,172].
The antioxidant activity of the methanolic extract from D. asper roots was confirmed in DPPH and Cu2+-mediated LDL oxidation with IC50 values of 90.2 and 134.4 µg/mL, respectively. This activity may be attributed to caffeoylquinic acid derivatives identified in root extract such as 3,4-di-O-caffeoylquinic acid, 4,5-di-O-caffeoylquinic acid, 3,5-di-O-caffeoylquinic acid, and their methyl derivatives. These specialized metabolites showed potent antioxidant activity against DPPH radical formation and Cu2+-mediated LDL oxidation with IC50 values of 10.4–18.2 µM and 1.8–2.3 µM, respectively [24]. The content of the total polyphenols in the acetone/water extract (7:3) of D. fullonum whole plant was 19.52 mg GAE (gallic acid equivalents)/g d.w. of plant material; it displayed an antioxidant capacity lower than 5 mmol TEAC (Trolox equivalent antioxidant capacity)/100 g d.w. plant material in the DPPH and ABTS assays [171]. The antioxidant properties were also demonstrated in 50% methanolic extract of leaves and roots (ultrasound assisted extraction) in the ORAC assay with values of 14.78 mmol TEAC/100 g d.w. and 10.87 mmol TEAC/100 g d.w. for the leaf and root extracts, respectively [33]. A similar observation was found by Saar-Reismaa et al. [34]. The crude 70% ethanol extract of D. fullonum leaves showed antioxidant activity in the ORAC assay (10.8 mmol TEAC/100 mL). The fraction NP7 of crude leaf extract that was rich in two chlorogenic acid derivatives, saponarin and isoorientin, also displayed antioxidant activity (12.5 mmol TEAC/100 mL), while the fraction NP2 containing bis-iridoids was ineffective (0.78 mmol TEAC/100 mL) [34].
On the other hand, the aqueous extract of D. fullonum leaves obtained by ultrasound assisted extraction (extraction time, four hours) showed high radical scavenging activity (RSA) (73.81%) in DPPH [172]. The procedure extraction with the aqueous solution of amino acid ionic liquids, viz., triethanolammonium salts of two amino acids methionine ([TEAH]+[Met]) and threonine ([TEAH]+[Thr]), resulted in a beneficial effect on the antioxidant activity of leaf extracts in FRAP and CUPRAC assays compared to the extract after extraction with the pure water. In addition, extraction with the aqueous solution of [TEAH]+[Thr] (extraction time, two hours) increased the total polyphenol content to 8.16 mg GAE/g raw material. The use of [TEAH]+[Met] in aqueous solution and reducing the extraction time to one hour resulted in a similar level of polyphenol (7.38 mg GAE per g of raw material) [172]. Antioxidant effect was also observed for the extract of D. sativus leaf. The extract at a dose of 300 mg/kg/day in ICR mice pre-treated with D-galactose to induce oxidative stress resulted in an increase in the level of SOD and a decrease in the level of MDA in the peripheral blood plasma [42].
S. stellata whole plants are a rich source of polyphenolic compounds. The ethyl acetate and n-butanolic fractions have moderate antioxidant activity in simple, chemical, antioxidant tests such as DPPH, ABTS assays, and FRAP test. The n-butanolic fraction displayed stronger potential in the DPPH assay, i.e., FRS50 (free radical scavenge) = 64.46 µg/mL, compared to ascorbic acid FRS50 = 8.21 µg/mL [76]. The dichloromethane fraction did not possess significant activity in antioxidant assays, which may be related to the low content (below 1 mg of gallic acid/g dry extract) of phenolic compounds [76]. A 70% ethanol extract of S. stellata whole plants in DPPH also displayed antioxidant activity, with an IC50 value of 86 μg/mL [75]. The petroleum ether, ethyl acetate, and n-butanolic fractions from S. stellata whole plants showed various antioxidant potential in different chemical models such as DPPH and ABTS, FRAP, CUPRAC, β-carotene, phosphomolybdate assay, ferrous ions, and metal chelating assays [50]. The strongest effect was demonstrated by the n-butanolic fraction in DPPH (IC50 = 21.22 μg/mL) and chelation in ferrous iron assay (EC50 = 1.65 mg/mL), while the ethyl acetate fraction was most active in ABTS (IC50 at 14 μg/mL), CUPRAC (A0.50 = 28.5 µg/mL), and β-carotene assays (IC50 = 10.34 μg/mL). In addition, the n-butanolic fraction in DPPH had a higher reducing power than butylated hydroxyl toluene (BHT) (22.32 µg/mL) but lower than α-tocopherol (13.02 µg/mL), butylated hydroxy anisole (BHA) (6.82 µg/mL), and ascorbic acid (3.1 µg/mL) [50]. The highest protein denaturation inhibition was found for the ethyl acetate extract, which showed 78.86% inhibition at the maximal tested concentration (1 mg/mL). Ibuprofen (standard drug) at the same concentration caused 100% inhibition [77]. Antioxidant activity was also reported for 70% ethanol extracts of S. comosa and S. tschilliensis inflorescences. S. tschilliensis had a stronger antioxidant activity than S. comosa in DPPH, ABTS, and FRAP assays. For example, the IC50 values in DPPH were 272.8 µg/mL and 331.1 µg/mL, respectively [74]. Wang et al. [52] reported that the 90% ethanol extract of S. tschilliensis at a concentration of 26.5 µg/mL scavenged 50% of DPPH free radicals (IC50 value for ascorbic acid was 5.41 µg/mL). The crude extract (95% ethanol) and four solvent partitioned fractions (water, n-butanol, ethyl acetate, and petroleum ether) from S. tschiliensis whole plants at various growing stages (pre-flowering, flowering, and fruiting stage) showed different antioxidant activities in DPPH, ABTS, inhibition of lipid peroxidation, or OH scavenging activity [53]. The IC50 values for the crude extract were in the range of 25.65–86.79 µg/mL while the ethyl acetate fraction from the pre-flowering stage of plants had the highest antioxidant capacity (IC50 8.47 µg/mL) in DPPH. This value was comparable to that of vitamin C (7.6 µg/mL). The ethyl acetate fraction from the pre-flowering stage of plants also possessed the highest ABTS (58.76 µg/mL), hydroxyl radical scavenging ability (67.64 µg/mL), and lipid-peroxidation-inhibition activity [53]. The n-butanolic and ethyl acetate fractions of S. arenaria roots also displayed strong antioxidant activity in four assays: DPPH, ABTS, reducing power, and β-carotene bleaching inhibition activity. The n-butanolic fraction demonstrated excellent ability, mainly in the β-carotene bleaching inhibition assay (IC50 = 0.018 mg/mL). This effect was stronger than that obtained for BHT (IC50 = 0.04 mg/mL). In the DPPH and ABTS assays, the IC50 values for both fractions were comparable to BHT [73]. The ethyl acetate fractions of the roots, flowers, fruits, and aerial parts (stems and leaves) of S. arenaria showed the beneficial antioxidant ability in DPPH, with IC50 values of 0.017–0.019 mg/mL; the best properties were observed for the flowers [72,73]. Other species of Scabiosa, S. artropurpurea and S. atropurpurea subsp. maritima, also demonstrated antioxidant properties. In comparison to ascorbic acid (IC50 = 0.084 mg/mL), among four tested extracts, the ethanol extract of S. artropurpurea stems exhibited the best antioxidant capacity, with IC50 = 0.1383 mg/mL in DPPH assay. In addition, the hexanoic volatile fraction (VF1) and ethyl acetate extract displayed similar effects with IC50 values of 0.4798 mg/mL and 0.4806 mg/mL, respectively [46]. The antioxidant activity of 70% ethanol extract of aerial parts of S. atropurpurea and hexane, ethyl acetate, n-butanol, and chloroform fractions were also demonstrated by increasing blood glutathione in diabetic albino rats [43]. Silver nanoparticles with S. atropurpurea subsp. maritima water fruit extract also was found as a promising antioxidant agent in DPPH and FRAP assays. The IC50 values were 0.112 mg/mL and 0.036 mg ascorbic acid equivalent antioxidant capacity/g d.w., respectively, and were comparable to that for ascorbic acid [56].
The pure compounds isolated from D. asper roots, i.e., six dicaffeoylquinic acid derivatives (3,4-di-O-caffeoylquinic acid, methyl 3,4-di-O-caffeoyl quinate, 3,5-di-O-caffeoylquinic acid, methyl 3,5-di-O-caffeoyl quinate, 4,5-di-O-caffeoylquinic acid, and methyl 4,5-di-O-caffeoyl quinate) exhibited strong antioxidant capacity in the DPPH assay (10.4–18.2 µM) and displayed inhibitory activity against Cu2+-mediated LDL oxidation (1.8–2.3 µM), stronger than those obtained for the positive controls, BHT and caffeic acid [24]. In another study, 3,5-di-O-caffeoylquinic acid also demonstrated significant antioxidant properties in the DPPH assay, with an IC50 value of 3.63 µg/mL [74]. Two secoiridoid glucosides, eustomoruside and eustomoside, and one flavonoid, isoorientin, isolated from the whole plant S. stellata Cav. also displayed strong radical scavenging activities in DPPH with an IC50 value of 7.1–8.5 μg/mL compared to that for ascorbic acid (IC50 = 6.3 μg/mL) [75]. The polysaccharide fraction from the roots of D. asperoides also demonstrated antioxidant effects in DPPH and ABTS assays but with little potency; the EC50 value was 0.355 mg/mL in DPPH free radical scavenging activity and 5.867 mg/mL in ABT [14].

6.10. Anticancer Activity

The synthetic drugs used in chemotherapy not only have strong toxic effects on cancer cells, but they also have strong adverse side effects in chemotherapy. Many plant specialized metabolites are used in cancer therapies and new substances, and new plant species with potential anti-cancer activity are being sought.
Considerable attention has been noted regarding the cytotoxicity of Dipsacus and Scabiosa against various cancer cell lines including lung carcinoma A549, hepatoma Bel7402 and Hep3B, gastric carcinoma BGC-823, AGS, KATO III, MKN-45, and SNU-638, liver H157 and HepG2, colon cancer HCT-8, ovary cancer A2780, breast MCF-7, human breast cancer MCF-7 and MDB-MB-231, acute myeloid leukemia OCI-AML3, osteosarcoma HOS, or fibrosarcoma HT1080 cell lines. Some pure specialized metabolites of different classes identified in the plant extracts have also demonstrated potent or promising cytotoxic effects in vitro (Table 10) [11,12,40,48,56,75,81,89,90,96,173,174,175].
An aqueous extract of D. asperoides roots inhibited the viability of human mammary carcinoma-derived triple negative MDA-MB-231 cells (with IC50 = 15 μg/mL) and arrested the cell cycle in the G2/M phase; it also induced apoptosis by increasing pro-apoptotic caspase 3/7 activity and by suppressing the expression of BRAF, p-ERK, MEK, pPI3K, pAKT, and cyclin-dependent kinase 4/6 in a dose-dependent manner [175]. The cytotoxic activity of bis-iridoid glycosides fraction of D. fullonum leaf methanol extract (with sylvestroside III and IV as the main compounds) was evaluated against human breast cancer cell lines MCF7 and MDB-MB-231 and human cervical cancer HeLa cell line [174]. The two breast cancer cell lines were most sensitive to the fraction, resulting in a viability of 64.0% for MCF7 cells and 69.5% for MDB-MD-231 cells [174]. In addition, the ethanolic extracts of the aerial parts and flowers of D. fullonum have antiproliferative activity on the human hepatocellular carcinoma Hep3B cell line with an IC50 value above 100 µg/mL [176]. The methanol extract of S. atropurpurea subsp. maritima leaves at a concentration of IC10, IC20, or IC30 enhanced the toxicity of doxorubicin in human epithelial colorectal adenocarcinoma Caco-2 cells with IC50 = 1.04 µg/mL (vs. 2.41 µg/mL when the cells were treated only with doxorubicin) [81]. In addition, the combination of doxorubicin with S. atropurpurea extracts at a concentration of IC50 and IC10, respectively, increased the percentage of apoptotic cells, the percentage of caspase-activated cells, mRNA levels of the apoptosis related-genes (Bax, caspase-3, p21), and decreased the expression level of anti-apoptotic genes (Bcl-2). It was a stronger effect than that obtained for doxorubicin or S. atropurpurea extract alone. The plant methanol extract also reversed P-glycoprotein or multidrug resistance-associated protein in Caco-2 cells [81]. The use of silver nanoparticles with S. atropurpurea subsp. maritima water fruit extract was found to be promising anticancer agents with cytotoxic activity against the human multiple myeloma U266 cell line and the human breast cancer cell line MDA-MB-231. The silver nanoparticles inhibited the growth of cells in a concentration-dependent manner with IC50 values of 10 and 12 µg/mL, respectively [56].
Some specialized metabolites such as phenolic acids, triterpenoid derivatives, or iridoids isolated from Dipsacus or Scabiosa revealed cytotoxic effects in various cancer cell lines [11,12,40,48,75,89,90,96,173]. Phenolic acids, such as caffeic acid, 2,6-dihydroxycinnamic acid, vanillic acid, 2′-O-caffeoyl-d-glucopyranoside ester, and caffeoylquinic acid, demonstrated cytotoxic activity against five cancer cell lines (A549, Bel7402, BGC-823, HCT-8, and A2780) with IC50 values ranging from 3.883 µg/mL to 7.395 µg/mL. The positive control, fluorouracil (a known cytostatic compound), had an IC50 value of 0.177–0.695 µg/mL [89]. Akebia saponin PA from D. asperoides caused the death of various human gastric cancer cell lines (AGS, MKN-45, SNU-638, and KATO III) via both apoptosis and autophagy. The IC50 values were 24.1 µM (MKN-45 cells), 27.6 µM (SNU-638), 30.3 µM (AGS), and 36.5 µM (KATO III). In addition, akebia saponin PA increased the AGS cell number in the sub-G1 phase and activated caspase-3, cleavage of PARP-1, MAPK, and p38/c-Jun N-terminal kinase. Autophagy was induced through the PI3K/AKT/mTOR and AMPK/mTOR pathways [173]. Another saponin, akebia saponin D, was also found to induce cytotoxicity of the human monocyte-like histiocytic U937 cells in a concentration-dependent manner (0.1–1000 µM); it also enhanced the percentage of sub-G1 cells and increased Bax and p53 gene expression [90]. Saponin XII isolated from the roots of D. japonicus (1–2 µg/mL) suppressed the growth of acute myeloid leukemia OCI-AML3 cells; it stimulated apoptosis, increased the number of cells in the G0/G1 phase of the cell cycle, decreased the number of cells in the S and G2/M phases, and activated caspase-3 [40]. Some triterpenoid saponins and iridoids isolated from S. stellata whole plants were found to have cytotoxic effects against the fibrosarcoma HT1080 cell line [48,75]. Scabiostellatoside F, at a concentration of 12.0 mM, was able to inhibit HT1080 cell growth by 50% [48]. Other triterpenoid saponins, scabiostellatoside B, D, E, and H, were found to have an IC50 of 38–49 µM. In addition, scabiostellatoside A, C, and G were not cytotoxic at a concentration of 50 mM [48]. Yu et al. [12] found that some compounds isolated from D. asper roots such as ursane and oleanane type triterpenoids with a feruloyloxy group or an arabinosyl moiety at C-3 were more cytotoxic than arboinane-type triterpenoids against four tumor cell lines: lung A549, liver H157 and HepG2, and breast MCF-7. Moreover, the highest activity was shown by an ursane-type triterpenoid (3β-O-trans-feruloyl-2α-hydroxy-urs-12-en-28-oic acid) with IC50 values of 5.66 μM (H157), 9.36 μM (MCF-7), 9.5 μM (HepG2), and 12.8 μM (A549). The oleanane-type triterpenoid arabinoglycosides with a diacetylated sugar unit displayed cytotoxicity against A549 and H157 cell lines with IC50 values below 10 μM. The compounds with a free or monoacetylated sugar moiety demonstrated cytotoxic activity with IC50 values above 20 μM [12]. Another oleanane-type triterpenoid saponin isolated from D. asper roots (3-O-[β-d-xylopyranosyl-(1→4)-β-d-glucopyranosyl-(1→4)][α-l-rhamnopyranosyl-(1→3)]-β-d-glucopyranosyl-(1→3)-α-l-rhamnopyranosyl-(1→2)-α-l-arabinopyranosylhederagenin) displayed cytotoxicity against two lung cancer cells lines, A549 and H157, with IC50 values of 6.94 and 9.06 μM, respectively [96].
The 16 kDa water-soluble polysaccharide (ADAPW) isolated from D. asperoides roots had the ability to inhibit the growth of human osteosarcoma cell line HOS and induce apoptosis in a concentration-dependent manner (100, 200, and 400 µg/mL) after 24 h. It was also found to down-regulate PI3K and pAkt protein levels, reduce mitochondrial membrane potential, and increase intracellular ROS level [11].
However, a number of iridoid glycosides (dipsanosides C-G, 3′-O-β-d-glucopyranosyl sweroside, loganin, cantleyoside, triplostoside A, lisianthioside, and 6′-O-β-d-apiofuranosyl sweroside) had no cytotoxic effect on a set of tested cell lines, including lung carcinoma A549, hepatoma Bel7402, gastric carcinoma BGC-823, colon cancer HCT-8, and ovary cancer A2780 [89]. Similarly, 7-O-(E-p-coumaroyl)-sylvestroside I isolated from the whole plants of S. stellata also was not cytotoxic (IC50 > 100 μg/mL) to fibrosarcoma HT1080 cells. However, 7-O-(E-caffeoyl)-sylvestroside I showed moderate activity, with an IC50 value of 35.9 μg/mL [75].
Taken together, these above results indicated that some Dipsacus and Scabiosa plants or some specialized metabolites may display anticancer activity and may be useful as chemopreventive agents.

6.11. Antimicrobial and Anti-Insecticidal Activity

An increase in bacterial resistance to antibiotics has caused researchers to look for alternative solutions, which may be natural antibiotics [177].
Recent studies confirmed that extracts or essential oils from Dipsacus or Scabiosa spp. such as D. asper, D. fullonum, D. japonicus, S. stellata, S. arenaria, or S. atropurpurea subsp. maritima have antimicrobial activity [21,33,34,56,75,77,82,83,102]. Traditionally, D. fullonum is known as the remedy for Lyme disease caused by Borrelia burgdorferi whose vectors are ticks. The anti-Borrelia activity of D. fullonum/D. sylvestris extracts were evaluated in only a few studies in the recent ten years [34,178,179]. A 70% ethanol extract of D. fullonum leaves and its fractions showed significant anti-Borrelia activity against the stationary phase of B. burgdorferi strain B31 [34]. The strongest growth inhibition was found for a crude ethanol extract, which suppressed the cell viability by about 80% at a concentration of 305.5 mg/L. The NP5 fraction, containing loganic acid, and NP7, rich in saponarin, isoorientin, and two chlorogenic acid derivatives, were also effective, with a residual viability of 23.4–29.8% at a concentration of 332.8 mg/L and 340.2 mg/L, respectively; these values were comparable to that of the positive control, the triple antibiotic combination (doxycylin, cefoperazone, and daptomycin at a dose of 22.2 mg/L, 33.4 mg/L, and 80.1 mg/L, respectively) [34]. In contrast, Feng et al. [179] found that the 45% ethanolic extract of D. fullonum (accidentally mixed with a sample of D. asper) at a concentration of 0.25–1% was not active against either the non-growing stationary phase or growing B. burgdorferi, with residual viability of 84–90% and MIC > 2%. Among three tested extracts (70% ethanolic, ethyl acetate, and dichloromethane extracts) from D. sylvestris roots, only the ethanol extract was inactive against B. burgdorferi while the ethyl acetate extract showed the strongest ability [178]. A 50% methanolic extracts of D. fullonum leaves and roots were also tested against other microorganisms, including bacteria (Bacillus subtilis B5, Escherichia coli ATCC 10536, Pseudomonas aeruginosa DSM 939, P. fluorescens W1, and Staphylococcus aureus DSM 799) and yeasts (Candida famata AII4b, C. tropicalis ATCC 60557, C. sphaerica FII7A, Saccharomyces cerevisiae SV30, and Yarrowia lipolytica PII6a). It was found that the cell growth inhibitory activity differed among plant materials and bacteria strains. The greatest effect of growth inhibition zones was observed for the root extract against E. coli ATCC 10536 and S. aureus DSM 799 [33].
The antibacterial potential was also found for S. arenaria [82]. Various degree of antibacterial activity was related to the type of plant material (stems and leaves, roots, flowers, and fruits) and solvent used (crude extract and its fractions such as ethyl acetate, n-butanol, and aqueous). It was found that the highest antibacterial effect was noted for the n-butanolic fraction of fruits. In this case, MIC values for two Escherichia coli strains and two Pseudomonas aeruginosa strains were 0.019 mg/mL and 0.156 mg/mL, respectively. The butanolic fractions of the aerial parts and flowers were active against only E. coli strains with MIC values of 0.078 mg/mL and 0.156 mg/mL, respectively. In addition, Staphylococcus aureus ATCC 25923 and S. saprophyticus were sensitive to the butanolic fraction of fruits (MIC = 0.625 mg/mL). Among four tested strains of Candida spp. (C. albicans ATCC 90028, C. glabrata ATCC 90030, C. parapsilosis ATCC 22019, and C. krusei ATCC 6258), the most sensitive was C. albicans ATCC 90028 with MIC = 0.0195 mg/mL. E. coli ATCC 25922 and C. albicans ATCC 90028 were also sensitive to eleven subfractions from the butanolic fraction of the aerial part (MIC = 0.0195 mg/mL) [82]. A 70% ethanol extract of the whole plant S. stellata showed the highest antibacterial activity against Streptococcus pyogenes with MIC = 1.2 mg/mL (in comparison to gentamicin MIC = 2 µg/mL). For other strains of Gram-positive bacteria (Bacillus subtilis, Enterococcus faecalis ATCC 1034, Staphylococcus aureus 8325-4, S. aureus CIP 53.154, S. epidermidis, Micrococcus luteus, and Listeria innocua), Gram-negative bacteria (Escherichia coli CIP 54.127, Enterobacter cloacae, Salmonella enterica, Serratia marcescens, Proteus vulgaris, Klebsiella pneumoniae, Providencia stuartii, Pseudomonas aeruginosa ATCC 9027, and Shigella sonnei) and five yeasts (Candida albicans, C. glabrata, C. tropicalis, C. kefyr, and Cryptococcus neoformans), MIC ranged from 2.5 mg/mL to above 10 mg/mL [75]. The highest antimicrobial activity was found for fractions B and C obtained after eluting from a Diaion HP-20 column with 25% and 50% methanol. Staphylococcus spp., Candida spp. (C. albicans, C. tropicalis, and C. kefyr), and Cryptococcus neoformans were the most sensitive microorganisms to both fractions with MIC values of 0.6–1.5 mg/mL, while E. faecalis ATCC 1034, M. luteus, and S. pyogenes were also sensitive to fraction B [75]. The ethyl acetate, n-butanol, and the petroleum ether extracts from S. stellata whole plants were also tested for antibacterial activity in the agar disk diffusion assay against ten bacterial strains including four Gram-positive (Staphylococcus aureus ATCC 25923, S. albus, Enterococcus spp., and Streptococcus D) and six Gram-negative bacteria (Escherichia coli ATCC 35218, Pseudomonas aeruginosa ATCC 15442, Acinetobacter baumannii, Proteus mirabilis, Salmonella typhimurium, and Enterobacter sakazaki) [77]. Three bacterial strains, S. albus, P. aregionosa ATCC 15442, and S. typhimurium, were the most resistant strains to all extracts. The highest activity was exhibited by the ethyl acetate extract against the clinical strain of P. mirabilis (16–20 mm of the growth inhibition zones at a concentration of 0.0625–1 mg/mL). In addition, this extract also was active against five other bacterial strains, including S. aureus ATCC 25923, A. baumannii, E. coli ATCC 35218, Enterococcus sp., and Streptococcus D. The petroleum ether extract showed inhibitory activity against S. aureus (ATCC 25923) and E. coli (ATCC 35218) while the n-buthanol extract against A. baumannii and E. sakazaki [77]. In another study, the antibacterial and antifungal activities of the silver nanoparticles with S. atropurpurea subsp. maritima water extract from fruit against bacteria (Escherichia coli, Micrococcus luteus, Staphylococcus aureus, and Klebsiella pneumoniae) and fungal pathogens including Candida clinical strains (C. albicans, C. tropicalis, and C. glabrata), Microsporum canis, Trichophytom rubrum, and Trichophytom interdigitale were also reported. The silver nanoparticles inhibited the cell growth of bacteria and Candida sp., as evidenced by the zone inhibition (19.3–28 mm) and the MIC value (3.9–15.62 µg/mL). The lowest MIC value was found for two dermatophyte species, T. rubrum and T. interdigitale. In addition, the antifungal potential of the silver nanoparticles was associated with the disruption of membrane integrity and attenuation of the biofilm and hyphae formation [56]. D. asper crude extract from the roots also displayed antifungal activity in vivo in a whole-plant assay. This property was evaluated against seven plant pathogenic fungi such as Magnaporthe grisea causing rice blast, Rhizoctonia solani causing rice sheath blight, Botrytis cinerea causing tomato gray mold, Phytophthora infestans causing tomato late blight, Puccinia recondita causing wheat leaf rust, Blumeria graminis f. sp. hordei causing barley powdery mildew, and Colletotrichum coccodes causing red pepper anthracnose. It was shown that the activity was dependent on the fungal pathogens and the solvent used (n-hexane, ethyl acetate, acetone, methylene chloride, and methanol) for extraction. The fungi causing the tomato late blight and the tomato gray mold were the most sensitive to Dipsacus root extract. The greatest anti-fungal effect was demonstrated by the ethyl acetate and acetone extracts at a concentration of 1–2 mg/mL that inhibited tomato diseases by 90% [21].
Antifungal activity was also demonstrated by the pure compounds isolated from the roots of D. asper such as cauloside A (the main compound of the extract). Cauloside A was most effective against fungal pathogens causing the tomato late blight, the rice blast, and the tomato gray mold at a dose of 0.5 mg/mL. Colchiside inhibited the growth of Phytophthora infestans while three sterols (campesterol, β-sitosterol, and stigmasterol) displayed the weakest antifungal activity [21]. Among twelve specialized metabolites isolated from the whole plant S. stellata, two iridoids, viz., 7-O-caffeoyl-sylvestroside I and 7-O-(p-coumaroyl)-sylvestroside I, showed the highest antimicrobial activity with an MIC value of 31.2 µg/mL against Enterococcus faecalis ATCC 1054 and Staphyllococcus epidermis; sylvestroside I was also able to inhibit the growth of E. coli CIP 54.127 (MIC = 62.5 µg/mL). These iridoids also inhibited the growth of S. aureus CIP 53.154 with an MIC value of 62.5 µg/mL [75]. 2′,4′-O-diacetyl-3-O-α-l-arabinopyranosyl-23-hydroxyolea-12-en-28-oic acid and hederagonic acid, isolated from D. asper roots, inhibited the growth of S. aureus ATCC 25923 with IC50 values of 12.3 and 10.3 µM, respectively. Furthermore, 2α,3β,24-trihydroxy-23-norurs-12-en-28-oic acid and 2α,3β-dihydroxy-23-norurs-4(24),11,13(18)-trien-28-oic acid also exhibited antimicrobial activity but the IC50 value was three-times higher [12]. The other triterpenoid derivative, oleanolic acid, found in some Dipsacus and Scabiosa species, showed weaker antibacterial properties against E. coli ATCC 25922, Pseudomonas aeruginosa ATCC 27853, and Candida albicans ATCC 90028 with an IC50 value ranging from 170 µM to 680 µM [82].
It is well known that essential oils and their ingredients have potent antimicrobial properties [177]. The essential oil isolated from flowers of S. arenaria showed a strong ability (stronger than the positive control, thymol; MIC = 0.2 mg/mL) to inhibit the growth of cells of two Staphylococcus aureus strains with an MIC = 0.1562 mg/mL [83]. Notably, the essential oil isolated from fruits was found to be an anticandidal agent against Candida albicans ATCC 90028, C. parapsilosis ATCC 27853, C. kreusei ATCC 6258, and C. glabrata ATCC 90030 (MIC = 0.625 mg/mL) [83]. The essential oil isolated from D. japonicus flowering aerial parts can be used as a promising, natural insecticidal agent against stored-product insects such as adult red flour beetles (Tribolium castaneum) and maize weevils (Sitophilus zeamais), and they displayed contact toxicity with LD50 values of 13.45 μg/adult and 18.32 μg/adult, respectively. This essential oil also had fumigant activity against adult insects with LC50 5.26 mg/l air for T. castaneum and 10.11 mg/l air for S. zeamais. The strongest fumigant toxicity was possessed by one of the abundant ingredients in D. japonicus essential oil, i.e., 1,8-cineole [102].
Regarding antiviral activity, only one study showed that dipsalignan A, (+)-1-hydroxy-2,6-bis-epi-pinoresinol, and dipsanosides M-N displayed inhibitory activities against human immunodeficiency virus-1 (HIV-1) integrase. The IC50 values were 53.26 μM, 61.74 μM, 84.03 μM, and 92.67 μM, respectively. The positive control, baicalein, had a value of 1.37 μM [13].

6.12. Others

Akebia saponin D was also found to be a potential antidepressant agent. Intraperitoneal injection (40 mg/kg/d) alleviated LPS-induced microglia-mediated neuroinflammatory response in mice by inhibiting the TLR4/NF-κB signaling pathway in the hippocampus and prefrontal cortex [180]. It also ameliorated chronic mild stress-induced depressive-like behaviors in C57BL/6 mice by inducing a neuroprotective microglial phenotype in the hippocampus through the PPAR-γ pathway [181]. A similar antidepressant effect was also found for apigenin in a mouse model of chronic mild stress [182].
Gong et al. [32] found akebia saponin D to be effective against pain. It displayed an anti-nociceptive effect in SPF KM mice by shortening the licking time in the formalin test, increasing the reaction time to heat stimuli, and inhibiting acetic acid-induced writhing in mice.
Akebia saponin D activated the expression of the progesterone receptor in primary decidual cells and the Notch signaling pathway. Gao et al. [62] proposed that Dipsaci radix and its main ingredient, akebia saponin D, may promote decidualization in pregnant women. Bushen Antai, a Chinese herbal medicine preparation containing Dipsaci radix, was found to reduce the pregnancy loss caused by mifepristone administration [183]. This preparation may stimulate estrogen and progesterone receptors through Akt and Erk1/2 signaling pathways in the maternal–fetal interface of pregnant rats.

7. Conclusions

The present review broadens the knowledge of the phytochemistry of some species of Dipsacus and Scabiosa genera as well as their biological properties. The phytochemical analyses showed qualitative similarities in some specialized metabolites, especially iridoids, between species of both genera. Some species of Dipsacus and Scabiosa contain above 200 different compounds belonging to iridoids, triterpenoids derivatives, flavonoids, or phenolic acids with caffeoylquinic acid derivatives. Dipsacus spp. were predominated by terpenoid saponins while Scabiosa spp. were rich sources of iridoids and flavonoids. Apigenin, luteolin, and their derivatives were particularly common. Hederagenin and its related saponins are the main group of triterpenoids identified in Dipsacus. The oleanane-type triterpenoids were also common in Scabiosa genus. The wine-processing method has a beneficial effect on the biological activity of Dipsaci radix and the level of some specialized metabolites with its quality indicator, akebia saponin D. Dipsacus, and Scabiosa species and their constituent compounds possess beneficial biological activities. Many in vitro and in vivo studies confirmed their traditional medicinal uses. Dipsaci radix and akebia saponin D demonstrated anti-osteoporosis and antiarthritic properties. Scabiosa spp. showed anti-hepatic fibrosis potential. In addition, akebia saponin D displayed cardioprotective activity. Antioxidant, antimicrobial, and anti-inflammatory activities of both genera were also confirmed. Some newly identified specialized metabolites such as polysaccharides displayed promising biological properties. Thus, in the future, it is worth paying more attention to their pharmacological activities. The varied biological activities of extracts from Dipsacus and Scabiosa as well as the pure compounds isolated from them indicate their potential use in the future as effective, natural herbal drugs in the treatment of various diseases in official medical applications.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules28093754/s1, Table S1. The list of accepted species in the genera Dipsacus and Scabiosa according to Plants of the World Online database [9].

Author Contributions

Conceptualization, data collection, design of the study, interpretation of the data, drafting the manuscript, visualization, reviewing, and editing, E.S.; critical revision of the manuscript, E.S. and A.S.; supervision, E.S. and A.S. The authors declare no conflict of interest. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Medical University of Lodz, grant number 503/3-012-01/503-31-001.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Not applicable.

References

  1. Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative Stress: Harms and Benefits for Human Health. Oxid. Med. Cell. Longev. 2017, 2017, 8416763. [Google Scholar] [CrossRef] [PubMed]
  2. Forman, H.J.; Zhang, H. Targeting oxidative stress in disease: Promise and limitations of antioxidant therapy. Nat. Rev. Drug Discov. 2021, 20, 689–709. [Google Scholar] [CrossRef] [PubMed]
  3. Vona, R.; Pallotta, L.; Cappelletti, M.; Severi, C.; Matarrese, P. The Impact of Oxidative Stress in Human Pathology: Focus on Gastrointestinal Disorders. Antioxidants 2021, 10, 201. [Google Scholar] [CrossRef] [PubMed]
  4. The Global Biodiversity Information Facility. Available online: https://www.gbif.org/ (accessed on 21 January 2023).
  5. Zhao, Y.-M.; Shi, Y.-P. Phytochemicals and biological activities of Dipsacus species. Chem. Biodivers. 2011, 8, 414–430. [Google Scholar] [CrossRef]
  6. Tao, Y.; Lin, C.; Yan, J. Traditional uses, processing methods, phytochemistry, pharmacology and quality control of Dipsacus asper Wall. ex C.B. Clarke: A review. J. Ethnopharmacol. 2020, 258, 112912. [Google Scholar] [CrossRef] [PubMed]
  7. Pinto, D.C.G.A.; Rahmouni, N.; Beghidja, N.; Silva, A.M.S. Scabiosa genus: A rich source of bioactive metabolites. Medicines 2018, 5, 110. [Google Scholar] [CrossRef] [PubMed]
  8. The World Flora Online Database. Available online: http://www.worldfloraonline.org/ (accessed on 4 February 2023).
  9. Plants of the World Online Database. Available online: https://powo.science.kew.org/ (accessed on 27 October 2022).
  10. Flora of China Database. Available online: http://www.efloras.org/ (accessed on 4 February 2023).
  11. Chen, J.; Yao, D.; Yuan, H.; Zhang, S.; Tian, J.; Guo, W.; Liang, W.; Li, H.; Zhang, Y. Dipsacus asperoides polysaccharide induces apoptosis in osteosarcoma cells by modulating the PI3K/Akt pathway. Carbohydr. Polym. 2013, 95, 780–784. [Google Scholar] [CrossRef]
  12. Yu, J.-H.; Yu, Z.-P.; Wang, Y.-Y.; Bao, J.; Zhu, K.-K.; Yuan, T.; Zhang, H. Triterpenoids and triterpenoid saponins from Dipsacus asper and their cytotoxic and antibacterial activities. Phytochemistry 2019, 162, 241–249. [Google Scholar] [CrossRef] [PubMed]
  13. Sun, X.; Ma, G.; Zhang, D.; Huang, W.; Ding, G.; Hu, H.; Tu, G.; Guo, B. New Lignans and Iridoid Glycosides from Dipsacus asper Wall. Molecules 2015, 20, 2165–2175. [Google Scholar] [CrossRef] [PubMed]
  14. Tan, L.-H.; Zhang, D.; Yu, B.; Zhao, S.-P.; Wang, J.-W.; Yao, L.; Cao, W.-G. Antioxidant activity and optimization of extraction of polysaccharide from the roots of Dipsacus asperoides. Int. J. Biol. Macromol. 2015, 81, 332–339. [Google Scholar] [CrossRef]
  15. Tao, Y.; Ren, Y.; Li, W.; Cai, B.; Di, L.; Shi, L.; Hu, L. Comparative pharmacokinetic analysis of extracts of crude and wine-processed Dipsacus asper in rats by a sensitive ultra-performance liquid chromatography-tandem mass spectrometry approach. J. Chromatogr. B. 2016, 1036, 33–41. [Google Scholar] [CrossRef]
  16. Li, T.; Hua, Q.; Li, N.; Cui, Y.; Zhao, M. Protective effect of a polysaccharide from Dipsacus asper Wall on streptozotocin (STZ)-induced diabetic nephropathy in rat. Int. J. Biol. Macromol. 2019, 133, 1194–1200. [Google Scholar] [CrossRef]
  17. Liu, K.; Liu, Y.; Xu, Y.; Nandakumar, K.S.; Tan, H.; He, C.; Dang, W.; Lin, J.; Zhou, C. Asperosaponin VI protects against bone destructions in collagen induced arthritis by inhibiting osteoclastogenesis. Phytomedicine 2019, 63, 153006. [Google Scholar] [CrossRef] [PubMed]
  18. Liu, Z.-G.; Zhang, R.; Li, C.; Ma, X.; Liu, L.; Wang, J.-P.; Mei, Q.-B. The osteoprotective effect of Radix Dipsaci extract in ovariectomized rats. J. Ethnopharmacol. 2009, 123, 74–81. [Google Scholar] [CrossRef]
  19. Chen, D.X.; Li, L.Y.; Zhang, X.; Wang, Y.; Zhang, Z. Genetic diversity and population structure of wild Dipsacus asperoides in China as indicated by ISSR markers. Genet. Mol. Res. 2014, 13, 6340–6349. [Google Scholar] [CrossRef]
  20. Han, J.-S.; Lee, B.-S.; Han, S.-R.; Han, H.-Y.; Chung, M.-K.; Min, B.-S.; Seok, J.H.; Kim, Y.-B. A subchronic toxicity study of Radix Dipsaci water extract by oral administration in F344 rats. Regul. Toxicol. Pharmacol. 2016, 81, 136–145. [Google Scholar] [CrossRef] [PubMed]
  21. Choi, N.H.; Jang, J.Y.; Choi, G.J.; Choi, Y.H.; Jang, K.S.; Nguyen, V.T.; Min, B.-S.; Dang, Q.L.; Kim, J.-C. Antifungal activity of sterols and dipsacus saponins isolated from Dipsacus asper roots against phytopathogenic fungi. Pestic. Biochem. Phys. 2017, 141, 103–108. [Google Scholar] [CrossRef] [PubMed]
  22. Yu, J.; Zhang, J.; Sun, X.; Xiao, C.; Wang, X.; Zhou, T. Comprehensive separation of iridoid glycosides and triterpenoid saponins from Dipsacus asper with salt-containing solvent by high-speed countercurrent chromatography coupled with recycling mode. J. Sep. Sci. 2020, 43, 1265–1274. [Google Scholar] [CrossRef]
  23. Li, F.; Yang, X.; Bi, J.; Yang, Z.; Zhang, C. Antiosteoporotic activity of Du-Zhong-Wan water extract in ovariectomized rats. Pharm. Biol. 2016, 54, 1857–1864. [Google Scholar] [CrossRef]
  24. Hung, T.M.; Na, M.K.; Thuong, P.T.; Su, N.D.; Sok, D.E.; Song, K.S.; Seong, Y.H.; Bae, K.H. Antioxidant activity of caffeoyl quinic acid derivatives from the roots of Dipsacus asper Wall. J. Ethnopharmacol. 2006, 108, 188–192. [Google Scholar] [CrossRef]
  25. Gu, M.; Jin, J.; Ren, C.; Chen, X.; Gao, W.; Wang, X.; Wu, Y.; Tian, N.; Pan, Z.; Wu, A.; et al. Akebia Saponin D suppresses inflammation in chondrocytes via NRF2/HO-1/NF-κB axis and ameliorates osteoarthritis in Mice. Food Funct. 2020, 11, 10852–10863. [Google Scholar] [CrossRef]
  26. Park, J.-Y.; Park, S.-D.; Koh, Y.J.; Kim, D.-I.; Lee, J.-H. Aqueous extract of Dipsacus asperoides suppresses lipopolysaccharide-stimulated inflammatory responses by inhibiting the ERK1/2 signaling pathway in RAW 264.7 macrophages. J. Ethnopharmacol. 2019, 231, 253–261. [Google Scholar] [CrossRef]
  27. Shin, N.-R.; Lee, A.Y.; Park, G.; Ko, J.-W.; Kim, J.-C.; Shin, I.-S.; Kim, J.-S. Therapeutic Effect of Dipsacus asperoides C. Y. Cheng et T. M. Ai in Ovalbumin-Induced Murine Model of Asthma. Int. J. Mol. Sci. 2019, 20, 1855. [Google Scholar] [CrossRef] [PubMed]
  28. Wagner, H.; Bauer, R.; Melchart, D.; Xiao, P.-G.; Staudinger, A. (Eds.) Chromatographic Fingerprint Analysis of Herbal Medicines, Thin-layer and High Performance Liquid Chromatography of Chinese Drugs, Second, Revised and Enlarged Edition; Springer: Wien, Austria, 2011; Volume 1, pp. 677–689. [Google Scholar]
  29. Pearson, J.L.; Lee, S.; Suresh, H.; Low, M.; Nang, M.; Singh, S.; Lamin, F.; Kazzem, M.; Sullivan, S.; Khoo, C.S. The Liquid Chromatographic Determination of Chlorogenic and Caffeic Acids in Xu Duan (Dipsacus asperoides) Raw Herb. ISRN Anal. Chem. 2014, 2014, 968314. [Google Scholar] [CrossRef]
  30. Chun, J.M.; Lee, A.Y.; Nam, J.Y.; Lim, K.S.; Choe, M.S.; Lee, M.Y.; Kim, C.; Kim, J.-S. Effects of Dipsacus asperoides Extract on Monosodium Iodoacetate-Induced Osteoarthritis in Rats Based on Gene Expression Profiling. Front. Pharmacol. 2021, 12, 615157. [Google Scholar] [CrossRef]
  31. Zhang, Z.-J.; Qian, Y.-H.; Hu, H.-T.; Yang, J.; Yang, G.-D. The herbal medicine Dipsacus asper Wall extract reduces the cognitive deficits and overexpression of β-amyloid protein induced by aluminum exposure. Life Sci. 2003, 73, 2443–2454. [Google Scholar] [CrossRef]
  32. Gong, L.-L.; Yang, S.; Liu, H.; Zhang, W.; Ren, L.-L.; Han, F.-F.; Lv, Y.-L.; Wan, Z.-R.; Liu, L.-H. Anti-nociceptive and anti-inflammatory potentials of Akebia saponin D. Eur. J. Pharmacol. 2018, 845, 85–90. [Google Scholar] [CrossRef] [PubMed]
  33. Oszmiański, J.; Wojdyło, A.; Juszczyk, P.; Nowicka, P. Roots and leaf extracts of Dipsacus fullonum L. and their biological activities. Plants 2020, 9, 78. [Google Scholar] [CrossRef] [PubMed]
  34. Saar-Reismaa, P.; Bragina, O.; Kuhtinskaja, M.; Reile, I.; Laanet, P.-R.; Kulp, M.; Vaher, M. Extraction and fractionation of bioactives from Dipsacus fullonum L. leaves and evaluation of their anti-Borrelia activity. Pharmaceuticals 2022, 15, 87. [Google Scholar] [CrossRef]
  35. Witkowska-Banaszczak, E. Dipsacus fullonum L. leaves and roots-identification of the components of the essential oils and alpha-amylase inhibitory activities of methanolic extracts. Acta Pol. Pharm. 2018, 75, 951–957. [Google Scholar] [CrossRef]
  36. Bullitta, S.; Piluzza, G.; Viegi, L. Plant resources used for traditional ethnoveterinary phytotherapy in Sardinia (Italy). Genet. Resour. Crop Evol. 2007, 54, 1447–1464. [Google Scholar] [CrossRef]
  37. Jeelani, S.M.; Rather, G.A.; Sharma, A.; Lattoo, S.K. In perspective: Potential medicinal plant resources of Kashmir Himalayas, their domestication and cultivation for commercial exploitation. J. Appl. Res. Med. Aromat. Plants. 2018, 8, 10–25. [Google Scholar] [CrossRef]
  38. Kanta, C.; Sharma, I.P.; Shiekh, M.A. Ethnobotanical studies on medicinal plants of Langate area, Kupwara, Jammu and Kashmir, India. J. Med. Plants Stud. 2018, 6, 94–97. [Google Scholar]
  39. Hassan, S.; Sajjad, N.; Khan, S.U.; Gupta, S.; Bhat, M.A.; Ali, R.; Ahmad, Z.; Ganie, S.A.; Hamid, R. Dipsacus inermis Wall. modulates inflammation by inhibiting NF-κB pathway: An in vitro and in vivo study. J. Ethnopharmacol. 2020, 254, 112710. [Google Scholar] [CrossRef]
  40. Cham, B.T.; Linh, N.T.T.; Thao, D.T.; Anh, N.T.H.; Tam, N.T.; Anh, B.K.; Muscari, I.; Adorisio, S.; Sung, T.V.; Thuy, T.T.; et al. Cell growth inhibition of saponin XII from Dipsacus japonicus Miq. on acute myeloid leukemia cells. Molecules 2020, 25, 3325. [Google Scholar] [CrossRef]
  41. World Health Organization; Regional Office for the Western Pacific. Medicinal Plants in Viet Nam; Series No. 3; Dan, N.V., Ed.; WHO Regional Office for the Western Pacific: Manila, Philippines, 1990; pp. 151–152. ISBN 9290611014. Available online: https://apps.who.int/iris/handle/10665/207579 (accessed on 21 January 2023).
  42. Yang, B.; Feng, X.; Xu, J.; Lei, H.; Zhang, L. Multi-Component HPLC Analysis and Antioxidant Activity Characterization of Extracts from Dipsacus sativus (Linn.) Honck. Int. J. Food Prop. 2016, 19, 1000–1006. [Google Scholar] [CrossRef]
  43. Elhawary, S.S.; Eltantawy, M.E.; Sleem, A.A.; Abdallah, H.M.; Mohamed, N.M. Investigation of Phenolic Content and Biological Activities of Scabiosa atropurpurea L. World Appl. Sci. J. 2011, 15, 311–317. [Google Scholar]
  44. Bonet, M.À.; Parada, M.; Selga, A.; Vallés, J. Studies on pharmaceutical ethnobotany in the regions of L’Alt Empordà and Les Guilleries (Catalonia, Iberian Peninsula). J. Ethnopharmacol. 1999, 68, 145–168. [Google Scholar] [CrossRef]
  45. Erarslan, Z.B.; Yeşil, Y. The anatomical properties of Scabiosa atropurpurea L. (Caprifoliaceae). Istanbul J. Pharm. 2018, 48, 1–5. [Google Scholar] [CrossRef]
  46. Hrichi, S.; Chaabane-Banaoues, R.; Bayar, S.; Flamini, G.; Majdoub, Y.O.E.; Mangraviti, D.; Mondello, L.; Mzoughi, R.E.; Babba, H.; Mighri, Z.; et al. Botanical and Genetic Identification Followed by Investigation of Chemical Composition and Biological Activities on the Scabiosa atropurpurea L. Stem from Tunisian Flora. Molecules 2020, 25, 5032. [Google Scholar] [CrossRef]
  47. Ma, Y.; Yuan, H.; Jin, R.; Bao, X.; Wang, H.; Su, X.; Mu, M.G.S.L.; Liang, J.; Zhang, J.; Wu, X. Flavonoid-rich Scabiosa comosa inflorescence extract attenuates CCl4-induced hepatic fibrosis by modulating TGF-β-induced Smad3 phosphorylation. Biomed. Pharmacother. 2018, 106, 426–433. [Google Scholar] [CrossRef]
  48. Lehbili, M.; Magid, A.A.; Kabouche, A.; Voutquenne-Nazabadioko, L.; Morjani, H.; Harakat, D.; Kabouche, Z. Triterpenoid saponins from Scabiosa stellata collected in North-eastern Algeria. Phytochemistry 2018, 150, 40–49. [Google Scholar] [CrossRef]
  49. Rahmouni, N.; Pinto, D.C.G.A.; Santos, S.A.O.; Beghidja, N.; Silva, A.M.S. Lipophilic composition of Scabiosa stellata L.: An underexploited plant from Batna (Algeria). Chem. Pap. 2018, 72, 753–762. [Google Scholar] [CrossRef]
  50. Mouffouk, C.; Hambaba, L.; Haba, H.; Mouffouk, S.; Bensouici, C.; Mouffouk, S.; Hachemi, M.; Khadraoui, H. Acute toxicity and in vivo anti-inflammatory effects and in vitro antioxidant and anti-arthritic potential of Scabiosa stellata. Orient. Pharm. Exp. Med. 2018, 18, 335–348. [Google Scholar] [CrossRef]
  51. Zheng, Q.; Koike, K.; Han, L.-K.; Okuda, H.; Nikaido, T. New Biologically Active Triterpenoid Saponins from Scabiosa tschiliensis. J. Nat. Prod. 2004, 67, 604–613. [Google Scholar] [CrossRef] [PubMed]
  52. Wang, J.; Liu, K.; Xu, D.; Wang, Q.; Bi, K.; Song, Y.; Li, J.; Zhang, L. Rapid micropropagation system in vitro and antioxidant activity of Scabiosa tschiliensis Grunning. Plant Growth Regul. 2013, 69, 305–310. [Google Scholar] [CrossRef]
  53. Wang, J.; Liu, K.; Li, X.; Bi, K.; Zhang, Y.; Huang, J.; Zhang, R. Variation of active constituents and antioxidant activity in Scabiosa tschiliensis Grunning from different stages. J. Food Sci. Technol. 2017, 54, 2288–2295. [Google Scholar] [CrossRef] [PubMed]
  54. Park, S.; Jun, M.; Park, S.; Park, S. Lineage-Specific Variation in IR Boundary Shift Events, Inversions, and Substitution Rates among Caprifoliaceae s.l. (Dipsacales) Plastomes. Int. J. Mol. Sci. 2021, 22, 10485. [Google Scholar] [CrossRef] [PubMed]
  55. Carlson, S.E.; Linder, H.P.; Donoghue, M.J. The historical biogeography of Scabiosa (Dipsacaceae): Implications for Old World plant disjunctions. J. Biogeogr. 2012, 39, 1086–1100. [Google Scholar] [CrossRef]
  56. Essghaier, B.; Toukabri, N.; Dridi, R.; Hannachi, H.; Limam, I.; Mottola, F.; Mokni, M.; Zid, M.F.; Rocco, L.; Abdelkarim, M. First Report of the Biosynthesis and Characterization of Silver Nanoparticles Using Scabiosa atropurpurea subsp. maritima Fruit Extracts and Their Antioxidant, Antimicrobial and Cytotoxic Properties. Nanomaterials 2022, 12, 1585. [Google Scholar] [CrossRef]
  57. Ryder, M.L. Teasel Growing for Cloth Raising. Folk Life 1969, 7, 117–119. [Google Scholar] [CrossRef]
  58. Rector, B.G.; Harizanova, V.; Sforza, V.; Widmer, T.; Wiedenmann, R.N. Prospects for biological control of teasels, Dipsacus spp., a new target in the United States. Biol. Control 2006, 36, 1–14. [Google Scholar] [CrossRef]
  59. Akar, Z. Chemical compositions by using LC-MS/MS and GC-MS and antioxidant activities of methanolic extracts from leaf and flower parts of Scabiosa columbaria subsp. columbaria var. columbaria L. Saudi. J. Biol. Sci. 2021, 28, 6639–6644. [Google Scholar] [CrossRef]
  60. Jin, H.; Yu, H.; Wang, H.; Zhang, J. Comparative Proteomic Analysis of Dipsacus asperoides Roots from Different Habitats in China. Molecules 2020, 25, 3605. [Google Scholar] [CrossRef]
  61. Wong, R.W.K.; Rabie, A.B.M.; Hägg, E.U.O. The Effect of Crude Extract from Radix Dipsaci on Bone in Mice. Phytother. Res. 2007, 21, 596–598. [Google Scholar] [CrossRef] [PubMed]
  62. Gao, J.; Zhou, C.; Li, Y.; Gao, F.; Wu, H.; Yang, L.; Qiu, W.; Zhu, L.; Du, X.; Lin, W.; et al. Asperosaponin VI promotes progesterone receptor expression in decidual cells via the notch signaling pathway. Fitoterapia 2016, 113, 58–63. [Google Scholar] [CrossRef]
  63. Cong, G.; Cui, L.; Zang, M.; Hao, L. Attenuation of renal ischemia/reperfusion injury by a polysaccharide from the roots of Dipsacus asperoides. Int. J. Biol. Macromol. 2013, 56, 14–19. [Google Scholar] [CrossRef] [PubMed]
  64. Ke, K.; Li, Q.; Yang, X.; Xie, Z.; Wang, Y.; Shi, J.; Chi, L.; Xu, W.; Hu, L.; Shi, H. Asperosaponin VI promotes bone marrow stromal cell osteogenic differentiation through the PI3K/AKT signaling pathway in an osteoporosis model. Sci. Rep. 2016, 6, 35233. [Google Scholar] [CrossRef] [PubMed]
  65. Tao, Y.; Du, Y.; Su, D.; Li, W.; Cai, B. UHPLC–MS/MS quantification combined with chemometrics for the comparative analysis of different batches of raw and wine-processed Dipsacus asper. J. Sep. Sci. 2017, 40, 1686–1693. [Google Scholar] [CrossRef]
  66. Tao, Y.; Huang, S.; Li, W.; Cai, B. Simultaneous Determination of Ten Bioactive Components in Raw and Processed Radix Dipsaci by UPLC-Q-TOF-MS. J. Chromatogr. Sci. 2019, 57, 122–129. [Google Scholar] [CrossRef]
  67. Tao, Y.; Chen, X.; Ren, Y.C.; Cai, B.C. Study on regulation of wine-processed dipsacus asper on OPG/RANK/RANKL axis system in osteoporosis rats. J. Mod. Med. Health 2016, 32, 1127–1129. [Google Scholar]
  68. Tao, Y.; Chen, X.; Li, W.; Cai, B.; Di, L.; Shi, L.; Hu, L. Global and Untargeted Metabolomics Evidence of the Protective Effect of Different Extracts of Dipsacus asper Wall. ex C.B. Clarke on Estrogen Deficiency after Ovariectomia in Rats. J. Ethnopharmacol. 2017, 199, 20–29. [Google Scholar] [CrossRef] [PubMed]
  69. Zhao, B.T.; Jeong, S.Y.; Moon, D.C.; Son, K.H.; Son, J.K.; Woo, M.H. Quantitative determination and pattern recognition analyses of bioactive marker compounds from Dipsaci Radix by HPLC. Arch. Pharm. Res. 2013, 36, 1345–1353. [Google Scholar] [CrossRef]
  70. Chun, J.M.; Lee, A.Y.; Moon, B.C.; Choi, G.; Kim, J.-S. Effects of Dipsacus asperoides and Phlomis umbrosa Extracts in a Rat Model of Osteoarthritis. Plants 2021, 10, 2030. [Google Scholar] [CrossRef] [PubMed]
  71. Kaul, M.K. High altitude botanicals in integrative medicine-Case studies from Northwest Himalaya. Indian J. Trad. Knowl. 2010, 9, 18–25. [Google Scholar]
  72. Besbes Hlila, M.; Omri, A.; Ben Jannet, H.; Lamari, A.; Aouni, M.; Selmi, B. Phenolic composition, antioxidant and anti-acetylcholinesterase activities of the Tunisian Scabiosa arenaria. Pharm. Biol. 2013, 51, 525–532. [Google Scholar] [CrossRef] [PubMed]
  73. Besbes Hlila, M.; Mosbah, H.; Mssada, K.; Benn Jannet, H.; Aouni, M.; Selmi, B. Acetylcholinesterase inhibitory and antioxidant properties of roots extracts from the Tunisian Scabiosa arenaria Forssk. Ind. Crops Prod. 2015, 67, 62–69. [Google Scholar] [CrossRef]
  74. Ma, J.N.; Bolraa, S.; Ji, M.; He, Q.Q.; Ma, C.M. Quantification and antioxidant and anti-HCV activities of the constituents from the inflorescences of Scabiosa comosa and S. tschilliensis. Nat. Prod. Res. 2016, 30, 590–594. [Google Scholar] [CrossRef]
  75. Lehbili, M.; Magid, A.A.; Hubert, J.; Kabouche, A.; Voutquenne-Nazabadioko, L.; Renault, J.-H.; Nuzillard, J.-M.; Morjani, H.; Abedini, A.; Gangloff, S.C.; et al. Two new bis-iridoids isolated from Scabiosa stellata and their antibacterial, antioxidant, anti-tyrosinase and cytotoxic activities. Fitoterapia 2018, 125, 41–48. [Google Scholar] [CrossRef]
  76. Rahmouni, N.; Pinto, D.C.G.A.; Beghidja, N.; Benayache, S.; Silva, A.M.S. Scabiosa stellata L. phenolic content clarifies its antioxidant activity. Molecules 2018, 23, 1285. [Google Scholar] [CrossRef]
  77. Mouffouk, C.; Mouffouk, S.; Hambaba, L.; Haba, H.; Mouffouk, S. Evaluation of cytotoxic effect, anticholinesterase, antioxidant, antiarthritic and antibacterial activities of the Algerian species Scabiosa stellata L. In Proceedings of the International Conference on Veterinary, Agriculture and Life Science, The Eurasia Proceedings of Science, Technology, Engineering & Mathematics, Antalya, ICVALS 2019, Antalya, Turkey, 26–29 October 2019; Volume 8, pp. 1–11, ISSN 2602-3199. [Google Scholar]
  78. Kılınc, H.; Masullo, M.; Lauro, G.; D’Urso, G.; Alankus, O.; Bifulco, G.; Piacente, S. Scabiosa atropurpurea: A rich source of iridoids with α-glucosidase inhibitory activity evaluated by in vitro and in silico studies. Phytochemistry 2023, 205, 113471. [Google Scholar] [CrossRef] [PubMed]
  79. Chen, Q.; Wang, Y.; Ma, F.; Han, M.; Wang, Z.; Xue, P.; Lu, J. Systematic profiling of the effective ingredients and mechanism of Scabiosa comosa and S. tschilliensis against hepatic fibrosis combined with network pharmacology. Sci. Rep. 2021, 11, 2600. [Google Scholar] [CrossRef]
  80. Menggensilimu, Y.H.; Zhao, C.; Bao, X.; Wang, H.; Liang, J.; Yan, Y.; Zhang, C.; Jin, R.; Ma, L.; Zhang, J.; et al. Anti-liver fibrosis effect of total flavonoids from Scabiosa comosa Fisch. ex Roem. et Schult. on liver fibrosis in rat models and its proteomics analysis. Ann. Palliat. Med. 2020, 9, 272–285. [Google Scholar] [CrossRef]
  81. Toumia, I.B.; Sobeh, M.; Ponassi, M.; Banelli, B.; Dameriha, A.; Wink, M.; Ghedira, L.C.; Rosano, C. A Methanol Extract of Scabiosa atropurpurea Enhances Doxorubicin Cytotoxicity against Resistant Colorectal Cancer Cells In Vitro. Molecules 2020, 25, 5265. [Google Scholar] [CrossRef]
  82. Besbes Hlila, M.; Mosbah, H.; Majouli, K.; Ben Nejma, A.; Ben Jannet, H.; Mastouri, M.; Aouni, M.; Selmi, B. Antimicrobial Activity of Scabiosa arenaria Forssk. Extracts and Pure Compounds Using Bioguided Fractionation. Chem. Biodivers. 2016, 13, 1262–1272. [Google Scholar] [CrossRef]
  83. Besbes, M.; Amel Omri, A.; Cheraif, I.; Daami, M.; Ben Jannet, H.; Mastouri, M.; Aouni, M.; Selmi, B. Chemical Composition and Antimicrobial Activity of Essential Oils from Scabiosa arenaria Forssk. Growing Wild in Tunisia. Chem. Biodivers. 2012, 9, 829–839. [Google Scholar] [CrossRef] [PubMed]
  84. Polat, E.; Alankus-Caliskan, Ö.; Karayildirim, T.; Bedir, E. Iridoids from Scabiosa atropurpurea L. subsp. maritima Arc. (L.). Biochem. Syst. Ecol. 2010, 38, 253–255. [Google Scholar] [CrossRef]
  85. Ge, H.Y.; Zhang, S.H.; Zhao, B.S.; Yu, C.K. Clinical effect of Mongolian medicine Qinggan Jiuwei powder in treatment of alcoholic liver fibrosis. J. Clin. Hepatol. 2017, 33, 2316–2320. [Google Scholar] [CrossRef]
  86. Ge, H.; Wang, A.; Su, Y.; Yu, C.L.; Gao, L.; Li, Y. Ameliorative effects of Qingganjiuwei powder, a traditional Mongolian medicine, against CCl4-induced liver fibrosis in rats. J. Ethnopharmacol. 2021, 264, 113226. [Google Scholar] [CrossRef]
  87. Xu, H.; Ma, Q.; Ma, J.; Wu, Z.; Wang, Y.; Ma, C. Hepato-protective effects and chemical constituents of a bioactive fraction of the traditional compound medicine-Gurigumu-7. BMC Complement. Altern. Med. 2016, 16, 179. [Google Scholar] [CrossRef] [PubMed]
  88. Wei, F.; Lou, Z.C.; Gao, M.; Miao, Z.C. Application of new techniques of NMR in the structure elucidation of Japondipsaponin E-1 isolated from Dipsacus japonicus Miq. Yaoxue Xuebao 1995, 30, 831–837. [Google Scholar]
  89. Tian, X.-Y.; Wang, Y.-H.; Liu, H.-Y.; Yu, S.-S.; Fang, W.-S. On the Chemical Constituents of Dipsacus asper. Chem. Pharm. Bull. 2007, 55, 1677–1681. [Google Scholar] [CrossRef]
  90. Jeong, S.-I.; Zhou, B.; Bae, J.-B.; Kim, N.-S.; Kim, S.-G.; Kwon, J.; Kim, D.-K.; Shin, T.-Y.; Jeon, H.; Lim, J.-P.; et al. Apoptosis-inducing Effect of Akebia Saponin D from the Roots of Dipsacus asper Wall in U937 Cells. Arch. Pharm. Res. 2008, 31, 1399–1404. [Google Scholar] [CrossRef]
  91. Hwang, S.A.; Hwang, I.Y.; Jung, J.; Kang, H.K.; Son, K.H.; Jeong, C.S. Dipsacus saponin C from Dipsacus asper Reduces the Risk of Gastritis and Gastric Ulcer in Rats. Food Nutr. Sci. 2012, 3, 931–941. [Google Scholar] [CrossRef]
  92. Li, F.; Tanaka, K.; Watanabe, S.; Tezuka, Y.; Saiki, I. Dipasperoside A, a Novel Pyridine Alkaloid-Coupled Iridoid Glucoside from the Roots of Dipsacus asper. Chem. Pharm. Bull. 2013, 61, 1318–1322. [Google Scholar] [CrossRef]
  93. Ling, Y.; Liu, K.; Zhang, Q.; Liao, L.; Lu, Y. High performance liquid chromatography coupled to electrospray ionization and quadrupole time-of-flight–mass spectrometry as a powerful analytical strategy for systematic analysis and improved characterization of the major bioactive constituents from Radix Dipsaci. J. Pharm. Biomed. Anal. 2014, 98, 120–129. [Google Scholar] [CrossRef]
  94. Wang, G.-V.; Zhao, Z.-L.; Xue, P.-F.; Ma, F.-X.; Zhang, D.-Y.; Wang, N.-N.; Li, M.-H. Chemical constituents from flowers of Scabiosa tschilliensis. Zhongguo Zhong Yao Za Zhi 2015, 40, 807–813. [Google Scholar]
  95. Du, W.; Li, X.; Yang, Y.; Yue, X.; Jiang, D.; Ge, W.; Cai, B. Quantitative determination, principal component analysis and discriminant analysis of eight marker compounds in crude and sweated Dipsaci Radix by HPLC-DAD. Pharm. Biol. 2017, 55, 2129–2135. [Google Scholar] [CrossRef]
  96. Yu, Z.-P.; Wang, Y.-Y.; Yu, S.-J.; Bao, J.; Yu, J.-H.; Zhang, H. Absolute structure assignment of an iridoid-monoterpenoid indole alkaloid hybrid from Dipsacus asper. Fitoterapia 2019, 135, 99–106. [Google Scholar] [CrossRef]
  97. Ji, D.; Zhang, C.; Li, J.; Yang, H.; Shen, J.; Yang, Z. A New Iridoid Glycoside from the Roots of Dipsacus asper. Molecules 2012, 17, 1419–1424. [Google Scholar] [CrossRef]
  98. Li, F.; Tanaka, K.; Watanabe, S.; Tezuka, Y.; Dipasperoside, B. A New Trisiridoid Glucoside from Dipsacus asper. Nat. Prod. Commun. 2016, 11, 891–894. [Google Scholar] [CrossRef] [PubMed]
  99. Sun, X.; Zhang, Y.; Yang, Y.; Liu, J.; Zheng, W.; Ma, B.; Guo, B. Qualitative and quantitative analysis of furofuran lignans, iridoid glycosides, and phenolic acids in Radix Dipsaci by UHPLC-Q-TOF/MS and UHPLC-PDA. J. Pharm. Biomed. Anal. 2018, 154, 40–47. [Google Scholar] [CrossRef]
  100. Li, F.; Nishidono, Y.; Tanaka, K.; Watanabe, S.; Tezuka, Y. A New Monoterpenoid Glucoindole Alkaloid from Dipsacus asper. Nat. Prod. Commun. 2020, 15, 1934578X20917292. [Google Scholar] [CrossRef]
  101. Kuhtinskaja, M.; Vaher, M. Extraction and analysis of bioactive compounds from Dipsacus Fullonum and Galium Verum for Lyme borreliosis treatment. Biomed. J. Sci. Tech. Res. 2018, 11, 8614–8616. [Google Scholar] [CrossRef]
  102. Liu, Z.L.; Jiang, G.H.; Zhou, L.; Liu, Q.Z. Analysis of the essential oil of Dipsacus japonicus flowering aerial parts and its insecticidal activity against Sitophilus zeamais and Tribolium castaneum. Z. Naturforsch. C J. Biosci. 2013, 68, 13–18. [Google Scholar] [CrossRef]
  103. Zeng, J.; Huang, T.; Xue, M.; Chen, J.; Feng, L.; Du, R.; Feng, Y. Current knowledge and development of hederagenin as a promising medicinal agent: A comprehensive review. RSC Adv. 2018, 8, 24188. [Google Scholar] [CrossRef]
  104. Xu, J.; Hu, Z.; He, H.; Ou, X.; Yang, Y.; Xiao, C.; Yang, C.; Li, L.; Jiang, W.; Zhou, T. Transcriptome analysis reveals that jasmonic acid biosynthesis and signaling is associated with the biosynthesis of asperosaponin VI in Dipsacus asperoides. Front. Plant Sci. 2022, 13, 1022075. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, C.; Wang, X.; Zhao, S.; Sun, W.; Tong, S. Preparative separation of structural isomeric pentacyclic triterpene oleanolic acid and ursolic acid from natural products by pH-zone-refining countercurrent chromatography. RSC Adv. 2019, 9, 38860. [Google Scholar] [CrossRef]
  106. Deng, S.; West, B.J.; Jensen, C.J. UPLC-TOF-MS Characterization and Identification of Bioactive Iridoids in Cornus mas Fruit. J. Anal. Methods Chem. 2013, 2013, 710972. [Google Scholar] [CrossRef]
  107. Chiu, C.-H.; Lin, K.-H.; Lin, H.-H.; Chu, W.-X.; Lai, Y.-C.; Chao, P.-Y. Analysis of Chlorogenic Acid in Sweet Potato Leaf Extracts. Plants 2022, 11, 2063. [Google Scholar] [CrossRef]
  108. Bligh, S.W.A.; Ogegbo, O.; Wang, Z.T. Flavonoids by HPLC. In Natural Products; Ramawat, K., Mérillon, J.M., Eds.; Springer: Berlin/Heidelberg, Germany, 2013; pp. 2107–2144. [Google Scholar] [CrossRef]
  109. PubChem Database. Available online: https://pubchem.ncbi.nlm.nih.gov/ (accessed on 21 January 2023).
  110. Xu, D.; Liu, J.; Zheng, W.; Gao, Q.; Gao, Y.; Leng, X. Identification of Polysaccharides from Dipsacus asperoides and Their Effects on Osteoblast Proliferation and Differentiation in a High-Glucose Environment. Front. Pharmacol. 2022, 13, 851956. [Google Scholar] [CrossRef]
  111. Sun, X.; Wei, B.; Peng, Z.; Fu, Q.; Wang, C.; Zhen, J.; Sun, J. Protective effects of Dipsacus asper polysaccharide on osteoporosis in vivo by regulating RANKL/RANK/OPG/VEGF and PI3K/Akt/eNOS pathway. Int. J. Biol. Macromol. 2019, 129, 579–587. [Google Scholar] [CrossRef] [PubMed]
  112. Huang, T.-Y.; Shahrousvand, M.; Hsu, Y.-T.; Su, W.-T. Polycaprolactone/Polyethylene Glycol Blended with Dipsacus asper Wall Extract Nanofibers Promote Osteogenic Differentiation of Periodontal Ligament Stem Cells. Polymers 2021, 13, 2245. [Google Scholar] [CrossRef]
  113. Niu, Y.B.; Li, Y.H.; Kong, X.H.; Zhang, R.; Sun, Y.; Li, Q.; Li, C.; Liu, L.; Wang, J.; Mei, Q.B. The beneficial effect of Radix Dipsaci total saponins on bone metabolism in vitro and in vivo and the possible mechanisms of action. Osteoporos. Int. 2012, 23, 2649–2660. [Google Scholar] [CrossRef] [PubMed]
  114. Niu, Y.-T.; Xie, L.; Deng, R.-R.; Zhang, X.-Y. In the presence of TGF-β1, Asperosaponin VI promotes human mesenchymal stem cell differentiation into nucleus pulposus like-cells. BMC Complement. Med. Ther. 2021, 21, 32. [Google Scholar] [CrossRef] [PubMed]
  115. Zhou, D.; Zhang, H.; Xue, X.; Tao, Y.; Wang, S.; Ren, X.; Su, J. Safety Evaluation of Natural Drugs in Chronic Skeletal Disorders: A Literature Review of Clinical Trials in the Past 20 years. Front. Pharmacol. 2022, 12, 801287. [Google Scholar] [CrossRef]
  116. Zhan, H.S.; Shi, Y.Y.; Zhao, Y.F.; Hu, X.Y. Phase III Clinical Investigation of Xu Duan Zhuang Gu Capsules to the Primary Osteoporotic Patient. Chin. J. Osteoporo. 2009, 15, 197–200. [Google Scholar]
  117. Xiao, T.T.; Xu, M.; Yang, X.H.; Kok, L.; Chow, Y.L.; Zhao, Z.Z.; Leung, K.S.-Y.; Yang, Z.J.; Tian, X.Y.; Li, Z.; et al. The evaluation on embryo toxicity of Dipsaci Radix with mice and embryonic stem cells. J. Ethnopharmacol. 2014, 151, 114–122. [Google Scholar] [CrossRef]
  118. Huang, Y.-X.; Liu, E.-W.; Wang, L.; Huo, Y.; Wang, Q.; Olaleye, O.; Wang, T.; Gao, X.-M. LC/MS/MS determination and pharmacokinetic studies of six compounds in rat plasma following oral administration of the single and combined extracts of Eucommia ulmoides and Dipsacus asperoides. Chin. J. Nat. Med. 2014, 12, 469–476. [Google Scholar] [CrossRef]
  119. Wang, Q.-H.; Yang, X.-L.; Xiao, W.; Wang, Z.-Z.; Ding, G.; Huang, W.-Z.; Yang, Z.-L.; Zhang, C.-F. Microcrystalline Preparation of Akebia Saponin D for its Bioavailability Enhancement in Rats. Am. J. Chin. Med. 2015, 43, 513–528. [Google Scholar] [CrossRef]
  120. Tao, Y.; Du, Y.; Li, W.; Cai, B.; Di, L.; Shi, L.; Hu, L. Integrating UHPLC–MS/MS quantification and DAS analysis to investigate the effects of wine-processing on the tissue distributions of bioactive constituents of herbs in rats: Exemplarily shown for Dipsacus asper. J. Chromatogr. B. Anal. Technol. Biomed. Life Sci. 2017, 1055–1056, 135–143. [Google Scholar] [CrossRef] [PubMed]
  121. Kim, B.-S.; Kim, Y.-C.; Zadeh, H.; Park, Y.-J.; Pi, S.-H.; Shin, H.-S.; You, H.-K. Effects of the dichloromethane fraction of Dipsaci Radix on the osteoblastic differentiation of human alveolar bone marrow-derived mesenchymal stem cells. Biosci. Biotechnol. Biochem. 2011, 75, 13–19. [Google Scholar] [CrossRef]
  122. Liu, M.; Xiao, G.G.; Rong, P.; Zhang, Z.; Dong, J.; Zhao, H.; Li, H.; Li, Y.; Pan, J.; Liu, H.; et al. Therapeutic effects of radix dipsaci, pyrola herb, and cynomorium songaricum on bone metabolism of ovariectomized rat. BMC Complement. Altern. Med. 2012, 12, 67. Available online: http://www.biomedcentral.com/1472-6882/12/67 (accessed on 21 January 2023). [CrossRef]
  123. Niu, Y.; Li, C.; Pan, Y.; Li, Y.; Kong, X.; Wang, S.; Zhai, Y.K.; Wu, X.; Fan, W.; Mei, Q. Treatment of Radix Dipsaci extract prevents long bone loss induced by modeled microgravity in hindlimb unloading rats. Pharm. Biol. 2015, 53, 110–116. [Google Scholar] [CrossRef]
  124. Niu, Y.-B.; Kong, X.-H.; Li, Y.-H.; Fan, L.; Pan, Y.-L.; Li, C.-R.; Wu, X.-L.; Lu, T.-L.; Mei, Q.-B. Radix Dipsaci total saponins stimulate MC3T3-E1 cell differentiation via the bone morphogenetic protein-2/MAPK/Smad-dependent Runx2 pathway. Mol. Med. Rep. 2015, 11, 4468–4472. [Google Scholar]
  125. Wang, X.; He, Y.; Guo, B.; Tsang, M.-C.; Tu, F.; Dai, Y.; Yao, Z.; Zheng, L.; Xie, X.; Wang, N.; et al. In Vivo Screening for Anti-osteoporotic Fraction from Extract of Herbal Formula Xianlinggubao in Ovariectomized Mice. PLoS ONE 2015, 10, e0118184. [Google Scholar] [CrossRef]
  126. He, J.; Li, X.; Wang, Z.; Bennett, S.; Chen, K.; Xiao, Z.; Zhan, J.; Chen, S.; Hou, Y.; Chen, J.; et al. Therapeutic Anabolic and Anticatabolic Benefits of Natural Chinese Medicines for the Treatment of Osteoporosis. Front. Pharmacol. 2019, 10, 1344. [Google Scholar] [CrossRef] [PubMed]
  127. Zhang, W.; Xue, K.; Gao, Y.; Huai, Y.; Wang, W.; Miao, Z.; Dang, K.; Jiang, S.; Qian, A. Systems pharmacology dissection of action mechanisms of Dipsaci Radix for osteoporosis. Life Sci. 2019, 235, 116820. [Google Scholar] [CrossRef]
  128. Wu, Q.-C.; Tang, X.-Y.; Dai, Z.-Q.; Dai, Y.; Xiao, H.-H.; Yao, X.-S. Sweroside promotes osteoblastic differentiation and mineralization via interaction of membrane estrogen receptor-α and GPR30 mediated p38 signalling pathway on MC3T3-E1 cells. Phytomedicine 2020, 68, 153146. [Google Scholar] [CrossRef] [PubMed]
  129. Aibar-Almazán, A.; Voltes-Martínez, A.; Castellote-Caballero, Y.; Afanador-Restrepo, D.F.; Carcelén-Fraile, M.d.C.; López-Ruiz, E. Current Status of the Diagnosis and Management of Osteoporosis. Int. J. Mol. Sci. 2022, 23, 9465. [Google Scholar] [CrossRef] [PubMed]
  130. Ahmed, H.M.M.; Yeh, J.-Y.; Tang, Y.-C.; Cheng, W.T.-K.; Ou, B.-R. Molecular screening of Chinese medicinal plants for progestogenic and antiprogestogenic activity. J. Biosci. 2014, 39, 453–461. [Google Scholar] [CrossRef] [PubMed]
  131. Niu, J.; Wang, Y.; Meng, Y.; Qi, W.; Wen, J. Asperosaponin VI induces osteogenic differentiation of human umbilical cord mesenchymal stem cells via the estrogen signaling pathway. Medicine 2022, 101, e32344. [Google Scholar] [CrossRef] [PubMed]
  132. Wu, H.; Zhong, Q.; Wang, J.; Wang, M.; Fang, F.; Xia, Z.; Zhong, R.; Huang, H.; Ke, Z.; Wei, Y.; et al. Beneficial Effects and Toxicity Studies of Xian-ling-gu-bao on Bone Metabolism in Ovariectomized Rats. Front. Pharmacol. 2017, 8, 273. [Google Scholar] [CrossRef]
  133. Tang, X.-Y.; Dai, Z.-Q.; Wu, Q.-C.; Zeng, J.-X.; Gao, M.-X.; Xiao, H.-H.; Yao, Z.-H.; Dai, Y.; Yao, X.-S. Simultaneous determination of multiple components in rat plasma and pharmacokinetic studies at a pharmacodynamic dose of Xian-Ling-Gu-Bao capsule by UPLC-MS/MS. J. Pharm. Biomed. Anal. 2020, 177, 112836. [Google Scholar] [CrossRef]
  134. Tian, S.; Zou, Y.; Wang, J.; Li, Y.; An, B.-Z.; Liu, Y.-Q. Protective effect of Du-Zhong-Wan against osteoporotic fracture by targeting the osteoblastogenesis and angiogenesis couple factor SLIT3. J. Ethnopharmacol. 2022, 295, 115399. [Google Scholar] [CrossRef] [PubMed]
  135. Chen, F.; Liang, W.; Mao, L.; Yin, Y.; Zhang, L.; Li, C.; Liu, C. Synergy effects of Asperosaponin VI and bioactive factor BMP-2 on osteogenesis and anti-osteoclastogenesis. Bioact. Mater. 2022, 10, 335–344. [Google Scholar] [CrossRef] [PubMed]
  136. Park, E.; Lee, C.G.; Lim, E.; Hwang, S.; Yun, S.H.; Kim, J.; Jeong, H.; Yong, Y.; Yun, S.-H.; Choi, C.W.; et al. Osteoprotective Effects of Loganic Acid on Osteoblastic and Osteoclastic Cells and Osteoporosis-Induced Mice. Int. J. Mol. Sci. 2021, 22, 233. [Google Scholar] [CrossRef]
  137. Shen, Y.; Teng, L.; Qu, Y.; Huang, Y.; Peng, Y.; Tang, M.; Fu, Q. Hederagenin Suppresses Inflammation and Cartilage Degradation to Ameliorate the Progression of Osteoarthritis: An In vivo and In vitro Study. Inflammation 2023, 46, 655–678. [Google Scholar] [CrossRef]
  138. Jung, H.W.; Jung, J.K.; Son, K.H.; Lee, D.H.; Kang, T.M.; Kim, Y.S.; Park, Y.-K. Inhibitory effects of the root extract of Dipsacus asperoides C.Y. Cheng et al. T.M.Ai on collagen-induced arthritis in mice. J. Ethnopharmacol. 2012, 139, 98–103. [Google Scholar] [CrossRef]
  139. Li, X.-R.; Li, J.; Ren, Q.; Sun, S. The molecular mechanism of treating osteoarthritis with dipsacus saponins by inhibiting chondrocyte apoptosis. Exp. Ther. Med. 2017, 14, 4527–4532. [Google Scholar] [CrossRef]
  140. Bai, J.; Xie, N.; Hou, Y.; Chen, X.; Hu, Y.; Zhang, Y.; Meng, X.; Wang, X.; Tang, C. The enhanced mitochondrial dysfunction by cantleyoside confines inflammatory response and promotes apoptosis of human HFLS-RA cell line via AMPK/Sirt 1/NF-κB pathway activation. Biomed Pharmacother. 2022, 149, 112847. [Google Scholar] [CrossRef] [PubMed]
  141. Zhang, R.; Wang, C.M.; Jiang, H.J.; Tian, X.G.; Li, W.; Liang, W.; Yang, J.; Zhong, C.; Chen, Y.; Li, T. Protective Effects of Sweroside on IL-1β-Induced Inflammation in Rat Articular Chondrocytes Through Suppression of NF-κB and mTORC1 Signaling Pathway. Inflammation 2019, 42, 496–505. [Google Scholar] [CrossRef] [PubMed]
  142. Available online: https://www.who.int/news-room/fact-sheets/detail/dementia (accessed on 21 January 2023).
  143. Yu, X.; Wang, L.-N.; Du, Q.-M.; Ma, L.; Chen, L.; You, R.; Liu, L.; Ling, J.-J.; Yang, Z.-L.; Ji, H. Akebia Saponin D attenuates amyloid β-induced cognitive deficits and inflammatory response in rats: Involvement of Akt/NF-κB pathway. Behav. Brain Res. 2012, 235, 200–209. [Google Scholar] [CrossRef]
  144. Qian, Y.-H.; Liu, Y.; Hu, H.-T.; Ren, H.-M.; Chen, X.-L.; Xu, J.-H. The effects of the total saponin of Dipsacus asperoides on the damage of cultured neurons induced by β-amyloid protein 25-35. Anat. Sci. Int. 2002, 77, 196–200. [Google Scholar] [CrossRef] [PubMed]
  145. Xie, Z.-S.; Zhao, J.-P.; Wu, L.-M.; Chu, S.; Cui, Z.-H.; Sun, Y.-R.; Wang, H.; Ma, H.-F.; Ma, D.-R.; Wang, P.; et al. Hederagenin improves Alzheimer’s disease through PPARα/TFEB-mediated autophagy. Phytomedicine 2023, 112, 154711. [Google Scholar] [CrossRef]
  146. Chen, C.; Ai, Q.; Shi, A.; Wang, N.; Wang, L.; Wei, Y. Oleanolic acid and ursolic acid: Therapeutic potential in neurodegenerative diseases, neuropsychiatric diseases and other brain disorders. Nutr. Neurosci. 2023, 26, 414–428. [Google Scholar] [CrossRef]
  147. Brinza, I.; Raey, M.A.E.; El-Kashak, W.; Eldahshan, O.A.; Hritcu, L. Sweroside Ameliorated Memory Deficits in ScopolamineInduced Zebrafish (Danio rerio) Model: Involvement of Cholinergic System and Brain Oxidative Stress. Molecules 2022, 27, 5901. [Google Scholar] [CrossRef]
  148. Ahmedy, O.A.; Abdelghany, T.M.; El-Shamarka, M.E.A.; Khattab, M.A.; El-Tanbouly, D.M. Apigenin attenuates LPS-induced neurotoxicity and cognitive impairment in mice via promoting mitochondrial fusion/mitophagy: Role of SIRT3/PINK1/Parkin pathway. Psychopharmacology 2022, 239, 3903–3917. [Google Scholar] [CrossRef]
  149. Wang, Z.Y.; Zhang, X.D.; Whang, W.K. The Effect of Terpenoids of Dipsacus Asperoides Against Alzheimer’s Disease and Development of Simultaneous Analysis by High Performance Liquid Chromatography. Nat. Prod. Commun. 2021, 16, 1934578X211044603. [Google Scholar] [CrossRef]
  150. Tan, Z.; Sun, H.; Xue, T.; Gan, C.; Liu, H.; Xie, Y.; Yao, Y.; Ye, T. Liver Fibrosis: Therapeutic Targets and Advances in Drug Therapy. Front. Cell Dev. Biol. 2021, 9, 730176. [Google Scholar] [CrossRef]
  151. Gong, J.; Yang, F.; Yang, Q.; Tang, X.; Shu, F.; Xu, L.; Wang, Z.; Yang, L. Sweroside ameliorated carbon tetrachloride (CCl4)-induced liver fibrosis through FXR-miR-29a signaling pathway. J. Nat. Med. 2020, 74, 17–25. [Google Scholar] [CrossRef] [PubMed]
  152. Yang, G.; Jang, J.H.; Kim, S.W.; Han, S.H.; Ma, K.H.; Jang, J.K.; Kang, H.C.; Cho, Y.Y.; Lee, H.S.; Lee, J.Y. Sweroside Prevents Non-Alcoholic Steatohepatitis by Suppressing Activation of the NLRP3 Inflammasome. Int. J. Mol. Sci. 2020, 21, 2790. [Google Scholar] [CrossRef]
  153. Meng, Z.; Zhu, B.; Gao, M.; Wang, G.; Zhou, H.; Lu, J.; Guan, S. Apigenin alleviated PA-induced pyroptosis by activating autophagy in hepatocytes. Food Funct. 2022, 13, 5559–5570. [Google Scholar] [CrossRef]
  154. Gong, L.-L.; Wang, Z.-H.; Li, G.-R.; Liu, L.-H. Protective Effects of Akebia Saponin D Against Rotenone-Induced Hepatic Mitochondria Dysfunction. J. Pharmacol. Sci. 2014, 126, 243–252. [Google Scholar] [CrossRef]
  155. Gong, L.-L.; Yang, S.; Zhang, W.; Han, F.-F.; Lv, Y.-L.; Wan, Z.-R.; Liu, H.; Jia, Y.-J.; Ling-ling, X.; Liu, L.-H. Akebia saponin D alleviates hepatic steatosis through BNip3 induced mitophagy. J. Pharmacol. Sci. 2018, 136, 189–195. [Google Scholar] [CrossRef] [PubMed]
  156. World Health Organization. Available online: https://www.who.int/health-topics/cardiovascular-diseases#tab=tab_1 (accessed on 1 April 2023).
  157. Yang, S.; Zhang, W.; Xuan, L.-L.; Han, F.-F.; Lv, Y.-L.; Wan, Z.-R.; Liu, H.; Ren, L.-L.; Gong, L.-L.; Liu, L.-H. Akebia Saponin D inhibits the formation of atherosclerosis in ApoE−/− mice by attenuating oxidative stress-induced apoptosis in endothelial cells. Atherosclerosis 2019, 285, 23–30. [Google Scholar] [CrossRef]
  158. Li, J.; Zhao, C.; Zhu, Q.; Wang, Y.; Li, G.; Li, X.; Li, Y.; Wu, N.; Ma, C. Sweroside Protects Against Myocardial Ischemia-Reperfusion Injury by Inhibiting Oxidative Stress and Pyroptosis Partially via Modulation of the Keap1/Nrf2 Axis. Front. Cardiovasc. Med. 2021, 8, 650368. [Google Scholar] [CrossRef]
  159. Ma, L.Q.; Yu, Y.; Chen, H.; Li, M.; Ihsan, A.; Tong, H.Y.; Huang, X.J.; Gao, Y. Sweroside Alleviated Aconitine-Induced Cardiac Toxicity in H9c2 Cardiomyoblast Cell Line. Front. Pharmacol. 2018, 9, 1138. [Google Scholar] [CrossRef]
  160. Li, C.; Gao, Y.; Tian, J.; Xing, Y.; Zhu, H.; Shen, J. Long-term oral Asperosaponin VI attenuates cardiac dysfunction, myocardial fibrosis in a rat model of chronic myocardial infarction. Food Chem. Toxicol. 2012, 50, 1432–1438. [Google Scholar] [CrossRef]
  161. Muruganathan, N.; Dhanapal, A.R.; Baskar, V.; Muthuramalingam, P.; Selvaraj, D.; Aara, H.; Shiek Abdullah, M.Z.; Sivanesan, I. Recent Updates on Source, Biosynthesis, and Therapeutic Potential of Natural Flavonoid Luteolin: A Review. Metabolites 2022, 12, 1145. [Google Scholar] [CrossRef] [PubMed]
  162. Huang, L.; Kim, M.-Y.; Cho, J.Y. Immunopharmacological Activities of Luteolin in Chronic Diseases. Int. J. Mol. Sci. 2023, 24, 2136. [Google Scholar] [CrossRef] [PubMed]
  163. Thomas, S.D.; Jha, N.K.; Jha, S.K.; Sadek, B.; Ojha, S. Pharmacological and Molecular Insight on the Cardioprotective Role of Apigenin. Nutrients 2023, 15, 385. [Google Scholar] [CrossRef]
  164. Song, J.-S.; Lim, K.-M.; Kang, S.; Noh, J.-Y.; Kim, K.; Bae, O.-N.; Chung, J.-H. Procoagulant and prothrombotic effects of herbal medicine, Dipsacus asper and its active ingredient, dipsacus saponin C, on human platelets. J. Thromb. Haemost. 2012, 10, 895–906. [Google Scholar] [CrossRef] [PubMed]
  165. Yang, W.; He, L. The protective effect of hederagenin on renal fibrosis by targeting muscarinic acetylcholine receptor. Bioengineered 2022, 13, 8689–8698. [Google Scholar] [CrossRef] [PubMed]
  166. Yang, S.; Hu, T.; Liu, H.; Lv, Y.-L.; Zhang, W.; Li, H.; Xuan, L.; Gong, L.-L.; Liu, L.-H. Akebia saponin D ameliorates metabolic syndrome (MetS) via remodeling gut microbiota and attenuating intestinal barrier injury. Biomed. Pharmacother. 2021, 138, 111441. [Google Scholar] [CrossRef]
  167. Li, F.; Wei, R.; Huang, M.; Chen, J.; Li, P.; Ma, Y.; Chen, X. Luteolin can ameliorate renal interstitial fibrosis-induced renal anaemia through the SIRT1/FOXO3 pathway. Food Funct. 2022, 13, 11896–11914. [Google Scholar] [CrossRef] [PubMed]
  168. Yang, M.; Wang, J.; Wang, Q. Hederagenin Exerts Potential Antilipemic Effect via p38MAPK Pathway in Oleic Acid-induced HepG2 cells and in Hyperlipidemic Rats. Ann. Acad. Bras. Cienc. 2022, 94, e20201909. [Google Scholar] [CrossRef]
  169. Luo, J.-F.; Zhou, H.; Lio, C.-K. Akebia Saponin D Inhibits the Inflammatory Reaction by Inhibiting the IL-6-STAT3-DNMT3b Axis and Activating the Nrf2 Pathway. Molecules 2022, 27, 6236. [Google Scholar] [CrossRef] [PubMed]
  170. Wang, R.; Dong, Z.; Lan, X.; Liao, Z.; Chen, M. Sweroside Alleviated LPS-Induced Inflammation via SIRT1 Mediating NF-κB and FOXO1 Signaling Pathways in RAW264.7 Cells. Molecules 2019, 24, 872. [Google Scholar] [CrossRef]
  171. Piluzza, G.; Bullitta, S. Correlations between phenolic content and antioxidant properties in twenty-four plant species of traditional ethnoveterinary use in the Mediterranean area. Pharm. Biol. 2011, 49, 240–247. [Google Scholar] [CrossRef]
  172. Roman, B.; Muzykiewicz-Szymańska, A.; Ossowicz-Rupniewska, P.; Klimowicz, A.; Janus, E. The application of amino acid ionic liquids as additives in the ultrasound-assisted extraction of plant material. RSC Adv. 2021, 11, 25983. [Google Scholar] [CrossRef] [PubMed]
  173. Xu, M.-Y.; Lee, D.H.; Joo, E.J.; Son, K.H.; Kim, Y.S. Akebia saponin PA induces autophagic and apoptotic cell death in AGS human gastric cancer cells. Food Chem. Toxicol. 2013, 59, 703–708. [Google Scholar] [CrossRef] [PubMed]
  174. Kuhtinskaja, M.; Bragina, O.; Kulp, M.; Vaher, M. Anticancer effect of the iridoid glycoside fraction from Dipsacus fullonum L. leaves. Nat. Prod. Commun. 2020, 15, 1934578X20951417. [Google Scholar] [CrossRef]
  175. Telang, N.; Nair, H.B.; Wong, G.Y.C. Anti-Proliferative and Pro-Apoptotic Effects of Dipsacus Asperoides in a Cellular Model for Triple-Negative Breast Cancer. Arch. Breast Cancer 2022, 9, 66–75. [Google Scholar] [CrossRef]
  176. Carraz, M.; Lavergne, C.; Jullian, V.; Wright, M.; Gairin, J.E.; de la Cruz, M.G.; Bourdy, G. Antiproliferative activity and phenotypic modification induced by selected Peruvian medicinal plants on human hepatocellular carcinoma Hep3B cells. J. Ethnopharmacol. 2015, 166, 185–199. [Google Scholar] [CrossRef] [PubMed]
  177. Chouhan, S.; Sharma, K.; Guleria, S. Antimicrobial Activity of Some Essential Oils-Present Status and Future Perspectives. Medicines 2017, 4, 58. [Google Scholar] [CrossRef]
  178. Liebold, T.; Straubinger, R.K.; Rauwald, H.W. Growth inhibiting activity of lipophilic extracts from Dipsacus sylvestris Huds. roots against Borrelia burgdorferi s. s. in vitro. Pharmazie 2011, 66, 628–630. [Google Scholar] [PubMed]
  179. Feng, J.; Leone, J.; Schweig, S.; Zhang, Y. Evaluation of natural and botanical medicines for activity against Growing and non-growing forms of B. burgdorferi. Front. Med. 2020, 7, 6. [Google Scholar] [CrossRef]
  180. Zhang, J.; Yi, S.; Li, Y.; Xiao, C.; Liu, C.; Jiang, W.; Yang, C.; Zhou, T. The antidepressant effects of asperosaponin VI are mediated by the suppression of microglial activation and reduction of TLR4/NF-κB-induced IDO expression. Psychopharmacology 2020, 237, 2531–2545. [Google Scholar] [CrossRef]
  181. Jiang, X.; Yi, S.; Liu, Q.; Su, D.; Li, L.; Xiao, C.; Zhang, J. Asperosaponin VI ameliorates the CMS-induced depressive-like behaviors by inducing a neuroprotective microglial phenotype in hippocampus via PPAR-γ pathway. J. Neuroinflamm. 2022, 19, 115. [Google Scholar] [CrossRef] [PubMed]
  182. Alghamdi, A.; Almuqbil, M.; Alrofaidi, M.A.; Burzangi, A.S.; Alshamrani, A.A.; Alzahrani, A.R.; Kamal, M.; Imran, M.; Alshehri, S.; Mannasaheb, B.A.; et al. Potential Antioxidant Activity of Apigenin in the Obviating Stress-Mediated Depressive Symptoms of Experimental Mice. Molecules 2022, 27, 9055. [Google Scholar] [CrossRef] [PubMed]
  183. Sun, L.; Yuan, Z.; Jian, L.; Jiang, Q.; Zhang, S.; Tan, J. The Modified Bushen Antai Recipe Upregulates Estrogen and Progesterone Receptors at the Maternal-Fetal Interface in Pregnant Rats with Mifepristone-Induced Pregnancy Loss. Evid. Based Complement. Altern. Med. 2019, 2019, 8312020. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The chemical structures of selected triterpene derivatives, iridoids, phenolic acids, and flavonoids identified in Dipsacus and Scabiosa. The chemical structures of the secondary metabolites were compiled based on the data reported in [103,104,105,106,107,108].
Figure 1. The chemical structures of selected triterpene derivatives, iridoids, phenolic acids, and flavonoids identified in Dipsacus and Scabiosa. The chemical structures of the secondary metabolites were compiled based on the data reported in [103,104,105,106,107,108].
Molecules 28 03754 g001aMolecules 28 03754 g001b
Table 1. The common names, natural occurrence, and traditional usage of Dipsacus and Scabiosa species.
Table 1. The common names, natural occurrence, and traditional usage of Dipsacus and Scabiosa species.
Accepted Species Name
(Synonym Name a)
Common NameGeographical OccurrenceHabitatTraditional Usage (Part of Plant)
D. asperoides C.Y.Cheng & T.M.Ai [8]
D. asper Wall. ex DC. [9]
D. asper Wall. [4]
(D. asper Wall. ex C.B.Clarke (doubtful) [4])
Roots are called Xu duan, chuan xu duan,
(in Chinese); Sichuan teasel, Himalayan Teasel Root (in English) [4,10,11,12,13,14,15,16,17]
China (Hubei, Hunan, Yunnan, Shanxi, Jiangxi, Sichuan, Gansu, Xizang, Guangxi, and Guizhou Provinces), Korea, Japan, Myanmar [9,12,18,19,20,21,22,23]Moist fields, margins of forests, thickets, among herbs, by streams, roadsides, and mountains; 1500–3700 m [10,24,25]Roots are used in traditional Chinese and Korean medicine to treat low back pain, knee pain, osteoporosis or bone diseases, bone fractures, rheumatic arthritis, lumbago, traumatic hematoma, uterine bleeding, and threatened abortion [6,11,14,15,22,26,27,28,29,30,31,32]
D. fullonum L. [4,8,9]
(D. sylvestris Huds. [8,9])
Cardo, Fuller’s teasel, teasel, wild teasel (in English) [4,9,33,34]Europe to the Caucasus, North-Western Africa [9]-Herb is used for treatment of Lyme disease [35] and for eye infection in cattle (in Sardinia) [36]
D. inermis Wall. [4,8,9]
(D. asper Wall. ex. DC. [8])
zang xu duan (in Chinese); Wopal haakh/Wopal Hak (in Kashmiri) [10,37,38]Afghanistan, Bangladesh, India, People’s Republic of China (Yunnan Province), Myanmar, Nepal, Pakistan, Thailand, Vietnam, Kashmir Himalaya [9,10,37]Forests, grassy slopes, by streams; 2100–3900 m [10]Himalayan herb is used in traditional Ayurvedic medicine and Kashmiri traditional medicine against various inflammation-related disorders, body weakness, cough, and sore throat. It has stomachic and carminative properties [37,38,39]
D. japonicus Miq. [4,8,9]ri ben xu duan, (in Chinese); Japanese teasel (in English) [4,10]People’s Republic of China, Korea, central and south Japan, and Vietnam [9,40]Grassy slopes and roadsides, savannas; below 1000–2600 m [10,41]Roots are used in the traditional medicine of China and Vietnam as a remedy for relieving joint pain and inflammation [40]
D. sativus (L.) Honc. [4,8,9]Fuller’s Teasel (in English) [4]Europa (France, Italy), Caucasus [9]-Tea of leaves is used for the treatment of cardiovascular diseases [42]
S. arenaria Forssk. [8,9]-Algeria, Egypt, Libya, Morocco, Palestine, Sinai, and Tunisia [9]--
S. atropurpurea L. [8,9]Mourningbride (in English); Mor uyuzotu or Şeytanotu (in Turkey); Ambarina
(in Northern Peru);
Escabiosa (in north-east Catalonia, Iberian Peninsula) [4,43,44,45]
Algeria, Tunisia, Turkey, Europe (Albania, Azores, Baleares, Bulgaria, Canary Is., Corse, East Aegean Is., France, Greece, Italy, Madeira, Morocco, Portugal, Sicilia, Spain) [9]Around roadsides, dry fields, and dunes
[45]
Flowers/aerial parts are used traditionally in Catalonia for bronchitis, acne, cold and cough, and for measles and furuncleas, in Northern Peru for menstrual regulation, and in the Iberian Peninsula as a veterinary diuretic [43,44,45,46]
S. comosa Fisch. ex Roem & Schult. [8,9]lan pen hua,
(in Chinese) [10]
People’s Republic of China (Gansu, Hebei, Henan, Heilongjiang, Jilin, Liaoning, Shaanxi, Shanxi Provinces, Nei, and Ningxia), Korea, Inner Mongolia, Russia (Far East, Siberia) [9,10]Sandy dunes, dry mountain slopes, steppes; 300–1600(–3000) m [10]In traditional Mongolian and Tibetan medicine, inflorescences are used in the treatment of liver diseases [7,47]
Lomelosia stellata (L.) Raf. [4,8,9]
(S. stellata L. [8,9])
Starflower pincushions
(in English) [4,48]
Endemic to North Africa (Algeria, Libya, Morocco, Tunisia), Europe (France, Portugal, Spain) [9,48]Dry sunny grassland and rocky hillsides [49]Leaves and flowers are used in the traditional medicine of Morocco to treat heel cracks and for the treatment of various respiratory diseases including bronchitis, bronchial pneumonia, influenza, and asthma [48,50]
S. tschiliensis Grüning [8,9]
(synonymous name of S. comosa Fisch. ex Roem & Schult. [8,9])
Meng Gu Shan Luo Bo (in Chinese) [51]Inner Mongolia
autonomy district and China (the west of Hebei Province) [52]
Mountainous regions (300–1500 m) [52,53]Inflorescences are used in inner Mongolia for the treatment of headache, fever, cough, and jaundice [51]
a names of species cited by the authors of publications used in this review, which are synonymous names according to The World Flora Online [8], Plants of the World Online [9], or The Global Biodiversity Information Facility [4].
Table 9. Antioxidant potential of some Dipsacus and Scabiosa species.
Table 9. Antioxidant potential of some Dipsacus and Scabiosa species.
SpeciesPlant MaterialExtract, Fraction, or Pure CompoundAntioxidant AssayAntioxidant ActivityPositive ControlReferences
D. asperRootsMethanolic extract;
3,4-di-O-caffeoylquinic acid; methyl 3,4-di-O-caffeoyl quinate; 3,5-di-O-caffeoylquinic acid; methyl 3,5-di-O-caffeoyl quinate; 4,5-di-O-caffeoylquinic acid; methyl 4,5-di-O-caffeoyl quinate
DPPH; Cu2+ mediated low-density lipoprotein (LDL)DPPH: methanolic extract IC50 = 90.2 µg/mL; 3,4-di-O-caffeoylquinic acid IC50 = 13.4 µM; methyl 3,4-di-O-caffeoyl quinate IC50 = 14.1 µM; 3,5-di-O-caffeoylquinic acid IC50 = 18.2 µM; methyl 3,5-di-O-caffeoyl quinate IC50 = 10.6 µM; 4,5-di-O-caffeoylquinic acid IC50 = 10.4 µM; methyl 4,5-di-O-caffeoyl quinate IC50 = 12.6 µM
Cu2+ mediated LDL: methanolic extract IC50 = 134.4 µg/mL; 3,4-di-O-caffeoylquinic acid IC50 = 2.1 µM; methyl 3,4-di-O-caffeoyl quinate IC50 = 1.9 µM; 3,5-di-O-caffeoylquinic acid IC50 = 2.3 µM; methyl 3,5-di-O-caffeoyl quinate IC50 = 2.0 µM; 4,5-di-O-caffeoylquinic acid IC50 = 2.3 µM; methyl 4,5-di-O-caffeoyl quinate IC50 = 1.8 µM
DPPH: BHT IC50 = 145.8 µM; caffeic acid IC50 = 31.1 µM
Cu2+ mediated LDL: BHT IC50 = 3 µM; caffeic acid IC50 = 5.2 µM; α-tocopherol IC50 = 23.4 µM
[24]
D. asperoidesRootsPolysaccharide fractionDPPH; ABTSDPPH: EC50 = 0.355 mg/mL (at concentration 1 mg/mL 86.2% scavenging rate)
ABTS: EC50 = 5.867 mg/mL (at concentration 10 mg/mL 92.6% scavenging rate)
-[14]
D. fullonumWhole plantsAcetone:water extract (7:3)DPPH; ABTSDPPH: 4.01 mmol TEAC/100 g d.w.
ABTS: 3.58 mmol TEAC/100 g d.w.
-[171]
LeavesAqueous extract; aqua [TEAH]+[Thr]; aqua [TEAH]+[Met]DPPH; FRAP; CUPRACDPPH: aqua radical scavenging activity (RSA) 68.11%–73.81%; aqua [TEAH]+[Thr] 57.55%–62.78%; aqua [TEAH]+[Met] 58.73%–61.17%
FRAP: aqua 13.14–14.85 mg FeSO4/g raw material; aqua [TEAH]+[Thr] 1.45–17.08 mg FeSO4/g raw material; aqua [TEAH]+[Met] 12.44–21.1 mg FeSO4/g raw material;
CUPRAC: aqua 4.93–5.71 mg TEAC/g raw material; aqua [TEAH]+[Thr] 5.08–5.74 mg TEAC/g raw material; aqua [TEAH]+[Met] 4.39–6.09 mg TEAC/g raw material
-[172]
Leaves70% ethanol extract (v/v %); fraction NP7 (contained two chlorogenic acid derivatives, saponarin and isoorientin); fraction NP2 (contained bis-iridoids)ORAC70% ethanol extract 10.8 mmol TEAC/100 mL;
NP7 fraction 12.5 mmol TEAC/100 mL;
NP2 fraction 0.78 mmol TEAC/100 mL
-[34]
Leaves50% methanol extractORAC14.78 mmol TEAC/100 g dry weight-[33]
Roots50% methanol extractORAC10.87 mmol TEAC/100 g dry weight-[33]
D. sativusLeavesMethanol extractIn vivo study with ICR mice pre-treated with D-galactoseSOD level in the peripheral blood plasma ; MDA level in the peripheral blood plasma -[42]
S. arenariaRoots80% (v/v) hydro-methanolic extract; n-butanol, ethyl acetate, and water fractionsDPPH; ABTS; reducing power; β-carotene bleaching inhibitionDPPH: 80% methanolic extract IC50 = 260 µg/mL; n-butanol fraction IC50 = 26 µg/mL; ethyl acetate fraction IC50 = 19 µg/mL; water fraction IC50 = 105 µg/mL
ABTS: 80% methanolic extract IC50 = 210 µg/mL; n-butanol fraction IC50 = 40 µg/mL; ethyl acetate fraction IC50 = 34 µg/mL; water fraction IC50 = 180 µg/mL
reducing power: 80% methanolic extract EC50 = 190 µg/mL; n-butanol fraction EC50 = 28 µg/mL; ethyl acetate fraction EC50 = 66 µg/mL; water fraction IC50 = 510 µg/mL
β-carotene bleaching inhibition: 80% methanolic extract IC50 = 720 µg/mL; n-butanol fraction IC50 = 18 µg/mL; ethyl acetate fraction IC50 = 26 µg/mL; water fraction IC50 = 450 µg/mL
DPPH: BHT IC50 = 18 µg/mL;
ABTS: BHT IC50 = 50 µg/mL;
reducing power: BHT EC50 = 20 µg/mL;
β-carotene bleaching inhibition: BHT IC50 = 40 µg/mL
[73]
Stem and leaves; flowers; fruits80% (v/v) hydro-methanolic extract; n-butanolic, ethyl acetate, and water fractionsDPPH; ABTS; reducing power; β-carotene bleaching inhibitionDPPH: steam and leaves 80% methanolic extract IC50 = 21 µg/mL; n-butanol fraction IC50 = 19 µg/mL; ethyl acetate fraction IC50 = 19 µg/mL; water fraction IC50 = 48 µg/mL;
DPPH: flowers 80% methanolic extract IC50 = 36 µg/mL; n-butanol fraction IC50 = 19 µg/mL; ethyl acetate fraction IC50 = 17 µg/mL; water fraction IC50 = 30 µg/mL;
DPPH: fruits 80% methanolic extract IC50 = 32 µg/mL; n-butanol fraction IC50 = 20 µg/mL; ethyl acetate fraction IC50 = 18 µg/mL; water fraction IC50 = 25 µg/mL;
ABTS: steam and leaves 80% methanolic extract IC50 = 810 µg/mL; ethyl acetate fraction IC50 = 110 µg/mL;
ABTS: flowers n-butanol fraction IC50 = 140 µg/mL; ethyl acetate fraction IC50 = 54 µg/mL;
ABTS: fruits ethyl acetate fraction IC50 = 170 µg/mL;
β-carotene bleaching inhibition: steam and leaves 80% methanolic extract IC50 = 920 µg/mL; n-butanol fraction IC50 = 400 µg/mL; ethyl acetate fraction IC50 = 210 µg/mL;
β-carotene bleaching inhibition: flowers 80% methanolic extract IC50 = 900 µg/mL; n-butanol fraction IC50 = 180 µg/mL; ethyl acetate fraction IC50 = 52 µg/mL;
β-carotene bleaching inhibition: fruits 80% methanolic extract IC50 = 870 µg/mL; n-butanol fraction IC50 = 700 µg/mL; ethyl acetate fraction IC50 = 680 µg/mL; water fraction IC50 = 800 µg/mL;
reducing power: steam and leaves 80% methanolic extract EC50 = 240 µg/mL; n-butanol fraction EC50 = 220 µg/mL; ethyl acetate fraction EC50 = 100 µg/mL; water fraction EC50 = 220 µg/mL;
reducing power: flowers 80% methanolic extract EC50 = 40 µg/mL; n-butanol fraction EC50 = 26 µg/mL; ethyl acetate fraction EC50 = 20 µg/mL; water fraction EC50 = 77 µg/mL;
reducing power: fruits n-butanol fraction EC50 = 800 µg/mL; ethyl acetate fraction EC50 = 52 µg/mL; water fraction EC50 = 64 µg/mL
DPPH: BHT IC50 = 18 µg/mL;
ABTS: BHT EC50 = 50 µg/mL;
β-carotene bleaching inhibition: BHT IC50 = 40 µg/mL;
reducing power: BHT EC50 = 20 µg/mL
[72]
S. artropurpureaStemsDichloromethane, chloroform, ethyl acetate, ethanol extracts; volatile fractions/hydro distillation V1 hexane, V2 chloroformDPPHDichloromethane extract IC50 = 2.7085 mg/mL; chloroform extract IC50 = 2.0951 mg/mL; ethyl acetate extract IC50 = 0.4806 mg/mL; ethanol extract IC50 = 0.1383 mg/mL; V1 fraction IC50 = 0.4798 mg/mL; V2 fraction IC50 = 1.2944 mg/mLAscorbic acid IC50 = 0.084 mg/mL[46]
S. artropurpurea subsp. maritimaFruitsWater extract (silver nanoparticles)DPPH; FRAPDPPH: IC50 = 0.112 mg/mL
FRAP; IC50 = 0.036 mg EAa/g d.w.
DPPH: ascorbic acid IC50 = 0.087 mg/mL;
FRAP: ascorbic acid IC50 = 0.024 mg EAa/g d.w.
[56]
S. comosaInflorescences70% ethanol extract; 3,5-di-O-caffeoylquinic acid; chlorogenic acid; 4,5-di-O-caffeoylquinic acid; 3,4-di-O-caffeoylquinic acid; caffeic acid; luteolin-7-glucoside; luteolin-6-C-glucoside; quercetin-3-glucoside; quercetin-3-rutinosideDPPH; ABTSDPPH: 70% ethanol extract IC50 = 331.1 µg/mL; 3,5-di-O-caffeoylquinic acid IC50 = 3.63 µg/mL; chlorogenic acid IC50 = 4.67 µg/mL; 4,5-di-O-caffeoylquinic acid IC50 = 4.01 µg/mL; 3,4-di-O-caffeoylquinic acid IC50 = 4.78 µg/mL; caffeic acid IC50 = 3.96 µg/mL; protocatechuic acid IC50 = 4.15 µg/mL; luteolin IC50 = 6.03 µg/mL; luteolin-7-glucoside IC50 = 9.16 µg/mL; luteolin-6-C-glucoside IC50 = 5.74 µg/mL; quercetin-3-glucoside IC50 = 6.47 µg/mL; quercetin-3-rutinoside IC50 = 6.15 µg/mL
ABTS: 70% ethanol extract IC50 = 223.5 µg/mL
-[74]
S. stellataWhole plants70% ethanol extract; fraction A 100% water; fraction B 25% methanol; fraction C 50% methanol; fraction D 75% methanol; fraction E 100% methanol; eustomoruside; eustomoside; hyperinDPPH70% ethanol extract IC50 = 86 µg/mL; fraction A IC50 = 133 µg/mL; fraction B IC50 = 48.7 µg/mL; fraction C IC50 = 25 µg/mL; fraction D IC50 = 64.3 µg/mL; fraction E IC50 >200 µg/mL; eustomoruside IC50 = 7.1 µg/mL; eustomoside IC50 = 7.2 µg/mL; hyperin IC50 = 16 µg/mLAscorbic acid IC50 = 6.3 µg/mL[75]
Whole plantsPetroleum ether; ethyl acetate; n-butanol extractDPPH; ABTS; FRAP; CUPRAC; β-carotene; phosphomolybdate; ferrous and metal ions chelatingDPPH: petroleum ether IC50 = 171.61 µg/mL; ethyl acetate IC50 = 25.15 µg/mL; n-butanol extract IC50 = 21.22 µg/mL
ABTS: petroleum ether IC50 = 64.1 µg/mL; ethyl acetate IC50 = 14.00 µg/mL; n-butanol extract IC50 = 24.99 µg/mL
CUPRAC: petroleum ether IC50 = 100.95 µg/mL; ethyl acetate IC50 = 28.5 µg/mL; n-butanol extract IC50 = 42.16 µg/mL
β-carotene: petroleum ether IC50 = 11.18 µg/mL; ethyl acetate IC50 = 10.34 µg/mL n; n-butanol extract IC50 = 50.01 µg/mL
chelation in ferrous iron: ethyl acetate EC50 = 5.026 mg/mL n; n-butanol extract EC50 = 1.652 mg/mL
chelation in metal iron: ethyl acetate EC50 > 200 mg/mL n; n-butanol extract EC50 = 145.35 mg/mL
DPPH: BHA IC50 = 6.82 µg/mL; BHT IC50 = 22.32 µg/mL; tannic acid IC50 = 7.74 µg/mL; ascorbic acid IC50 = 3.1 µg/mL; α-tocopherol IC50 = 13.02 µg/mL
ABTS: BHA IC50 = 1.81 µg/mL; BHT IC50 = 1.29 µg/mL; tannic acid IC50 = 1.01 µg/mL; ascorbic acid IC50 = 1.74 µg/mL; α-tocopherol IC50 = 7.59 µg/mL
CUPRAC: BHA IC50 = 3.64 µg/mL; BHT IC50 = 9.62 µg/mL; tannic acid IC50 = 3.76 µg/mL; ascorbic acid IC50 = 12.43 µg/mL; α-tocopherol IC50 = 19.92 µg/mL
chelation in ferrous iron: EDTA EC50 = 0.0627 µg/mL
chelation in metal iron: EDTA EC50 = 8.57 mg/mL
[50,77]
Whole plantsn-butanol, ethyl acetate, dichloromethane fractionsDPPH; ABTS; FRAPDPPH: n-butanol FRS50 = 64.46µg/mL; ethyl acetate FRS50 = 71.82 µg/mL; dichloromethane FRS50 > 250 µg/mL
ABTS: n-butanol FRS50 = 27.87 µg/mL; ethyl acetate FRS50 = 40.41 µg/mL; dichloromethane FRS50 > 250 µg/mL
FRAP: n-butanol EC50 = 161.11 µg/mL; ethyl acetate EC50 = 202.41 µg/mL; dichloromethane EC50 > 50 µg/mL
DPPH: ascorbic acid FRS50 = 8.21 µg/mL
ABTS: Trolox FRS50 = 12.07 µg/mL
FRAP: BHA EC50 = 18.03 µg/mL
[76]
S. tschilliensisInflorescences70% ethanol extractDPPH; ABTSDPPH: IC50 = 272.8 µg/mL
ABTS: IC50 = 199.7 µg/mL
-[74]
Whole plants90% ethanol extractDPPHDPPH: IC50 = 26.502 µg/mLAscorbic acid IC50 = 5.41 µg/mL[52]
Whole plants in pre-flowering, flowering, and fruiting stages96% ethanol extract; water, n-butanol, ethyl acetate, petroleum ether fractionsDPPH; ABTS; inhibition of lipid peroxidation; OH scavenging activityDPPH: 96% ethanol extract IC50 = 25.65–86.79 µg/mL; acetate fraction of pre-flowering stage of plants IC50 = 8.47 µg/mL
ABTS: acetate fraction of pre-flowering stage of plants IC50 = 58.76 µg/mL
OH scavenging activity: 96% ethanol extract IC50 = 206.47–772.45 µg/mL; acetate fraction of pre-flowering stage of plants IC50 = 67.64 µg/mL
DPPH: ascorbic acid IC50 = 7.6 µg/mL;
ABTS:
lipid peroxidation inhibition:
OH scavenging activity:
[53]
ABTS, 2,2′-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) radical cation assay; BHA, butylated hydroxyanisole; BHT, butylhydroxytoluene; CUPRAC, cupric-reducing antioxidant capacity; DPPH, 2,2-Diphenyl-1-picrylhydrazyl radical scavenging assay; EAa, equivalent ascorbic acid; EC50 and IC50, the half maximal effective and inhibitory concentration of sample, respectively; FRAP, ferric reducing antioxidant assay; FR, free radical scavenge; LDL, low-density lipoprotein; ORAC, oxygen radical absorbance capacity; TEAC, Trolox equivalent antioxidant capacity; , decrease; , increase.
Table 10. Anticancer activities of some species of Dipsacus and Scabiosa.
Table 10. Anticancer activities of some species of Dipsacus and Scabiosa.
SpeciesPlant MaterialExtract or Specialized MetabolitesCell LineAssayResultPositive ControlReferences
D. asperRootsPhenolic acids (2,6-dihydroxycinnamic acid, vanillic acid, 2′-O-caffeoyl-d-glucopyranoside ester, caffeoylquinic acid)
iridoids (dipsanosides C-G, 3′-O-β-d-glucopyranosyl sweroside, loganin, cantleyoside, triplostoside A, lisianthioside, and 6′-O-β-d-apiofuranosyl sweroside)
Lung carcinoma A549, hepatoma Bel7402, gastric carcinoma BGC-823, colon cancer HCT-8, and ovary cancer A2780MTT assay, cell viability (96 h)2,6-dihydroxycinnamic acid, A549 IC50 = 3.883 µg/mL, Bel7402 IC50 = 7.346 µg/mL, BGC-823 IC50 = 4.321 µg/mL;
vanillic acid, Bel7402 IC50 = 6.437 µg/mL, HCT-8 IC50 = 5.218 µg/mL, A2780 IC50 = 7.395 µg/mL;
2′-O-caffeoyl-d-glucopyranoside ester, A549 IC50 = 5.663 µg/mL; Bel7402 IC50 = 5.545 µg/mL, BGC-823 IC50 = 6.432 µg/mL, HCT-8 IC50 = 5.7 µg/mL, A2780 IC50 = 6.380 µg/mL;
caffeoylquinic acid, A549 IC50 = 5.713 µg/mL; Bel7402 IC50 = 5.586 µg/mL, BGC-823 IC50 = 6.204 µg/mL, HCT-8 IC50 = 5.37 µg/mL, A2780 IC50 = 6.679 µg/mL
iridoids were not cytotoxic against A549, Bel7402, BGC-823, HCT-8, and A2780 cell lines
Fluorouracil, A549 IC50 = 0.177 µg/mL, Bel7402 IC50 = 0.542 µg/mL, BGC-823 IC50 = 0.695 µg/mL, HCT-8 IC50 = 0.67 µg/mL, A2780 IC50 = 0.569 µg/mL[89]
RootsAkebia saponin DHuman monocyte-like histiocytic U937 cellsMTT assay, cytotoxicity (48 h); flow cytometric analysis and DNA ladding (level of DNA fragmentation, sub-G1 peak); RT-PCRInduction of cell cytotoxicity in a concentration-dependent manner (0.1–1000 µM); an increase in the percentage of Sub G1 cells and Bax and p53 gene expressionDoxorubicin, the cell viability 25% at a concentration of 0.1 µM[90]
Roots2′,3′-O-diacetyl-3-O-α-l-arabinopyranosyl-23-hydroxyolea-12-en-
28-oic acid;
2′,4′-O-diacetyl-3-O-α-l-arabinopyranosyl-23-hydroxyolea-12-en-
28-oic acid;
3β-O-trans-feruloyl-2α-hydroxy-urs-12-en-28-oic acid;
leontoside A;
4′-O-acetyl-3-O-α-l-arabinopyranosyl-23-hydroxyolea-12-en-28-oic acid;
3′-O-acetyl-3-O-α-l-arabinopyranosyl-23-hydroxyolea-12-en-28-oic acid;
2′-O-acetyl-3-O-α-l-arabinopyranosyl-23-hydroxyolea-12-en-28-oic acid
Lung A549, liver H157 and HepG2, and breast MCF-7 cell linesSRB method, cytotoxicity (48 h)2′,3′-O-diacetyl-3-O-α-L-arabinopyranosyl-23-hydroxyolea-12-en-28-oic acid,
A549 IC50 = 6.67 µM; H157 IC50 = 9.7 µM; HepG2 IC50 = 15.89 µM; MCF-7 IC50 = 15.08 µM;
2′,4′-O-diacetyl-3-O-α-L-arabinopyranosyl-23-hydroxyolea-12-en-28-oic acid, A549 IC50 = 6.67 µM; H157 IC50 = 9.57 µM; HepG2 IC50 = 16.23 µM; MCF-7 IC50 = 15.23 µM;
3β-O-trans-feruloyl-2α-hydroxy-urs-12-en-28-oic acid, A549 IC50 = 12.8 µM; H157 IC50 = 5.66 µM; HepG2 IC50 = 9.5 µM; MCF-7 IC50 = 9.36 µM;
leontoside A, A549 IC50 = 35.24 µM; H157 IC50 = 36.45 µM; HepG2 IC50 = 46.4 µM; MCF-7 IC50 = 52.06 µM;
4′-O-acetyl-3-O-α-l-arabinopyranosyl-23-hydroxyolea-12-en-28-oic acid, A549 IC50 = 22.94 µM; H157 IC50 = 21.21 µM; HepG2 IC50 = 27.13 µM; MCF-7 IC50 = 26.72 µM;
3′-O-acetyl-3-O-α-l-arabinopyranosyl-23-hydroxyolea-12-en-28-oic acid, A549 IC50 = 34.18 µM; H157 IC50 = 30.02 µM; HepG2 IC50 = 34.35 µM; MCF-7 IC50 = 37.32 µM;
2′-O-acetyl-3-O-α-l-arabinopyranosyl-23-hydroxyolea-12-en-28-oic acid, A549 IC50 = 33.14 µM; H157 IC50 = 27.54 µM; HepG2 IC50 = 34.5 µM; MCF-7 IC50 = 35.66 µM
Doxorubicin, A549 IC50 = 1.68 µM; H157 IC50 = 0.85 µM; HepG2 IC50 = 1.57 µM; MCF-7 IC50 = 0.9 µM[12]
Roots3-O-[β-d-xylopyranosyl-(14)-β-d-glucopyranosyl-(14)][α-l-rhamnopyranosyl-(13)]-β-d-glucopyranosyl-(13)-α-l-rhamnopyranosyl-(12)-α-l-arabinopyranosylhederageninLung A549, liver H157 and HepG2, and breast MCF-7 cell linesSRB method, cytotoxicity (48 h)A549 IC50 = 6.94 µM; H157 IC50 = 9.06 µM-[96]
D. asperoidesRootsWater extractHuman mammary carcinoma derived triple negative MDA-MB-231 cellsCell Titre Glo 2.0 assay, cell viability (7 d); flow cytometer (cell cycle); Western Blot; caspase 3/7 activityCell viability IC50 = 15 µg/mL; arrest cycle growth of cells in G2/M phase; induction of apoptosis by increasing pro-apoptotic caspase 3/7 activity and inhibiting the expression of BRAF, p-ERK, MEK, pPI3K, pAKT, and cyclin-dependent kinase 4/6 in a dose-dependent manner-[175]
RootsAkebia saponin PAHuman gastric cancer AGS, MKN-45, SNU-638, and KATO III cell linesMTT assay, cell viability (24 h); Annexing V/propidium (PI) staining; Western BlotCell viability AGS IC50 = 30.3 µM;
cell viability MKN-45 IC50 = 24.1 µM;
cell viability SNU-638 IC50 = 27.6 µM;
cell viability KATO III IC50 = 36.5 µM;
an increase in apoptotic cells (AGS) to 9.46%, 19.33%, and 48.20% at a concentration of 20 µM, 30 µM, and 40 µM, respectively; an increase in AGS cell number in the sub-G1 phase; activation of caspase-3, cleavage of PARP-1, mitogen-activated protein kinases (MAPKs), and p38/c-Jun N-terminal kinase (JNK); induction of autophagy through PI3K/AKT/mTOR and AMPK/mTOR pathways
-[173]
RootsWater-soluble polysaccharide (ADAPW) with the molecular weight of 16 kDaHuman osteosarcoma cell line HOS cellsMTT assay, cell viability (24 h); Annexin V-FITC/PI staining; Western BlotInhibition of cell growth (at the concentration of 400 µg/mL, the cell survival rate ˂35%); induction of apoptosis in a concentration-dependent pattern (at a concentration of 100–400 µg/mL, the number of apoptotic cell increase to 23.7%-55.3%); down-regulation of PI3K and pAkt protein level; reduction of mitochondrial membrane potential; an increase in intracellular ROS level [11]
D. fullonumLeavesIridoid glycosides fraction of 80% methanol extractHuman breast cancer cell lines MCF7 and MDB-MB-231 and human cervical cancer cell line HeLaWST-1 assay, cell viability (72 h)Decrease in cell survival; the viability of cells 64%, 69.5%, and 78.9% for MCF7, MDB-MB-231, and HeLa, respectively-[174]
Leaves and flowers96% ethanol extractHuman hepatocellular carcinoma cell lines Hep3B, HepG2, PLC/PFR/5, and SNU-182ATPlite assay, cell viability (8 h)Hep3B IC50 > 100 µg/mL; HepG2, PLC PFR5, SNU-182 antiproliferative effect not detected-[176]
D. japonicusRootsSaponin XIIacute myeloid leukemia OCI-AML3 cellsCell viability (24 h) and cell cycle progression
were analyzed by flow cytometry to determine the DNA content of cell nuclei stained with propidium iodide (PI)
Apoptosis cell death at a concentration of 1–2 µg/mL (0.648–1.295 µM); an increase in the number of cells in the G0/G1 phase of the cell cycle and a decrease in the number of cells in the S and G2/M phases, and activation of caspase-3-[40]
S. atropurpurea subsp. maritimaLeavesMethanolic extractHuman epithelial colorectal adenocarcinoma Caco-2MTT assay, cell viability (48 h); Annexin-V/PI Double-Staining analysis of apoptotic cells, flow cytometer; Multicaspase assay; quantitative reverse transcription Real-Time PCR (RT-qPCR)Plant extract at a concentration of IC10, IC20, or IC30 enhances the toxicity of doxorubicin; cell viability IC50 = 1.04 µg/mL; the combination of doxorubicin with plant extract at a concentration of IC50 and IC10 results in an increase in the percentage of apoptotic cells, the percentage of caspase-activated cells, mRNA levels of the apoptosis related-genes (Bax, caspase-3, p21), and a decrease in expression level of anti-apoptotic genes (Bcl-2)doxorubicin cell viability IC50 = 2.41 µg/mL[81]
FruitsWater extract, silver nanoparticlesHuman multiple myeloma U266 cell line and human breast cancer cell line MDA-MB-231MTT assay, cell viability (48 h)U266 IC50 = 10 µg/mL;
MDA-MB-231 IC50 = 12 µg/mL
-[56]
S. stellataWhole plantsScabiostellatosides A-HFibrosarcoma HT1080 cell lineWST1 assay, cell viability (72 h)Scabiostellatoside B, IC50 = 49 µM; scabiostellatoside D, IC50 = 40 µM; scabiostellatoside E, IC50 = 38 µM; scabiostellatoside F, IC50 =12 µM; scabiostellatoside H, IC50 = 40 µM;
scabiostellatoside A, scabiostellatoside C, scabiostellatoside G not cytotoxic at a concentration of 50 µM
Doxorubicin, IC50 = 0.59 µM[48]
Whole plantsIridoids (7-O-(E-caffeoyl)-sylvestroside I and 7-O-(E-p-coumaroyl)-sylvestroside IFibrosarcoma HT1080 cell lineWST1 assay, cell viability (72 h)7-O-(E-caffeoyl)-sylvestroside I, IC50 = 35.9 µg/mL;
7-O-(E-p-coumaroyl)-sylvestroside I, IC50 > 100 µg/mL
-[75]
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

Skała, E.; Szopa, A. Dipsacus and Scabiosa Species—The Source of Specialized Metabolites with High Biological Relevance: A Review. Molecules 2023, 28, 3754. https://doi.org/10.3390/molecules28093754

AMA Style

Skała E, Szopa A. Dipsacus and Scabiosa Species—The Source of Specialized Metabolites with High Biological Relevance: A Review. Molecules. 2023; 28(9):3754. https://doi.org/10.3390/molecules28093754

Chicago/Turabian Style

Skała, Ewa, and Agnieszka Szopa. 2023. "Dipsacus and Scabiosa Species—The Source of Specialized Metabolites with High Biological Relevance: A Review" Molecules 28, no. 9: 3754. https://doi.org/10.3390/molecules28093754

Article Metrics

Back to TopTop